Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Cell Mol Med ; 28(10): e18396, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38801304

RESUMEN

Previous studies have found that ferroptosis plays an important role in a variety of neurological diseases. However, the precise role of ferroptosis in the multiple sclerosis patients remains uncertain. We defined and validated a computational metric of ferroptosis levels. The ferroptosis scores were computed using the AUCell method, which reflects the enrichment scores of ferroptosis-related genes through gene ranking. The reliability of the ferroptosis score was assessed using various methods, involving cells induced to undergo ferroptosis by six different ferroptosis inducers. Through a comprehensive approach integrating snRNA-seq, spatial transcriptomics, and spatial proteomics data, we explored the role of ferroptosis in multiple sclerosis. Our findings revealed that among seven sampling regions of different white matter lesions, the edges of active lesions exhibited the highest ferroptosis score, which was associated with activation of the phagocyte system. Remyelination lesions exhibit the lowest ferroptosis score. In the cortex, ferroptosis score were elevated in neurons, relevant to a variety of neurodegenerative disease-related pathways. Spatial transcriptomics demonstrated a significant co-localization among ferroptosis score, neurodegeneration and microglia, which was verified by spatial proteomics. Furthermore, we established a diagnostic model of multiple sclerosis based on 24 ferroptosis-related genes in the peripheral blood. Ferroptosis might exhibits a dual role in the context of multiple sclerosis, relevant to both neuroimmunity and neurodegeneration, thereby presenting a promising and novel therapeutic target. Ferroptosis-related genes in the blood that could potentially serve as diagnostic and prognostic markers for multiple sclerosis.


Asunto(s)
Ferroptosis , Esclerosis Múltiple , Proteómica , Ferroptosis/genética , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Esclerosis Múltiple/metabolismo , Humanos , Proteómica/métodos , Transcriptoma , Microglía/metabolismo , Microglía/patología , Perfilación de la Expresión Génica , Biología Computacional/métodos , Neuronas/metabolismo , Neuronas/patología , Multiómica
2.
J Transl Med ; 22(1): 302, 2024 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-38521921

RESUMEN

BACKGROUND: Myasthenia gravis (MG) is a chronic autoimmune disorder characterized by fluctuating muscle weakness. Despite the availability of established therapies, the management of MG symptoms remains suboptimal, partially attributed to lack of efficacy or intolerable side-effects. Therefore, new effective drugs are warranted for treatment of MG. METHODS: By employing an analytical framework that combines Mendelian randomization (MR) and colocalization analysis, we estimate the causal effects of blood druggable expression quantitative trait loci (eQTLs) and protein quantitative trait loci (pQTLs) on the susceptibility of MG. We subsequently investigated whether potential genetic effects exhibit cell-type specificity by utilizing genetic colocalization analysis to assess the interplay between immune-cell-specific eQTLs and MG risk. RESULTS: We identified significant MR results for four genes (CDC42BPB, CD226, PRSS36, and TNFSF12) using cis-eQTL genetic instruments and three proteins (CTSH, PRSS8, and CPN2) using cis-pQTL genetic instruments. Six of these loci demonstrated evidence of colocalization with MG susceptibility (posterior probability > 0.80). We next undertook genetic colocalization to investigate cell-type-specific effects at these loci. Notably, we identified robust evidence of colocalization, with a posterior probability of 0.854, linking CTSH expression in TH2 cells and MG risk. CONCLUSIONS: This study provides crucial insights into the genetic and molecular factors associated with MG susceptibility, singling out CTSH as a potential candidate for in-depth investigation and clinical consideration. It additionally sheds light on the immune-cell regulatory mechanisms related to the disease. However, further research is imperative to validate these targets and evaluate their feasibility for drug development.


Asunto(s)
Predisposición Genética a la Enfermedad , Miastenia Gravis , Humanos , Multiómica , Estudio de Asociación del Genoma Completo , Miastenia Gravis/genética , Sitios de Carácter Cuantitativo/genética , Polimorfismo de Nucleótido Simple/genética
3.
J Neuroinflammation ; 20(1): 63, 2023 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-36890539

RESUMEN

Vitamin D deficiency is associated with worse clinical outcomes after ischemic stroke; nevertheless, the pathophysiological mechanisms remain largely unexplored. In this study, we characterized the molecular mechanisms of how vitamin D signaling modulated stroke progression in male mouse ischemia-reperfusion stroke models. We found that vitamin D receptor (VDR) exhibited a predominant upregulation in peri-infarct microglia/macrophages following cerebral ischemia. Conditional Vdr inactivation in microglia/macrophages markedly augmented infarct volumes and neurological deficits. VDR-deficient microglia/macrophages exhibited a more primed proinflammatory phenotype with substantial secretion of TNF-α and IFN-γ. These inflammatory cytokines further enhanced CXCL10 release from endothelial cells and blood-brain barrier disruption, and ultimately infiltration of peripheral T lymphocytes. Notably, blocking TNF-α and IFN-γ significantly ameliorated stroke phenotypes in Vdr conditional knockout mice. Collectively, VDR signaling in microglia/macrophages plays a crucial role in restraining ischemia-elicited neuroinflammation and stroke progression. Our findings delineate a novel mechanism underlying the association between vitamin D deficiency and poor stroke outcomes, and underline the significance of maintaining a functional vitamin D signaling in the management of acute ischemic stroke.


Asunto(s)
Lesiones Encefálicas , Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Deficiencia de Vitamina D , Ratones , Masculino , Animales , Microglía , Accidente Cerebrovascular Isquémico/complicaciones , Enfermedades Neuroinflamatorias , Factor de Necrosis Tumoral alfa , Vitamina D , Células Endoteliales , Macrófagos , Accidente Cerebrovascular/complicaciones , Isquemia Encefálica/complicaciones , Isquemia Encefálica/genética , Lesiones Encefálicas/complicaciones , Modelos Animales de Enfermedad , Ratones Noqueados , Infarto , Deficiencia de Vitamina D/complicaciones , Infarto de la Arteria Cerebral Media/complicaciones
4.
Front Neuroendocrinol ; 63: 100932, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34273406

RESUMEN

The social and public health burdens of ischemic stroke have been increasing worldwide. In addition to focal brain damage, acute ischemic stroke (AIS) provokes systemic abnormalities across peripheral organs. AIS profoundly alters the autonomic nervous system, hypothalamic-pituitary-adrenal axis, and immune system, which further yield deleterious organ-specific consequences. Poststroke systemic pathological alterations in turn considerably contribute to the progression of ischemic brain injury, which accounts for the substantial impact of systemic complications on stroke outcomes. This review provides a comprehensive and updated pathophysiological model elucidating the systemic effects of AIS. To address their clinical significance and inform stroke management, we also outline the resulting systemic complications at particular stages of AIS and highlight the mechanisms. Future therapeutic strategies should attempt to integrate the treatment of primary brain lesions with interventions for secondary systemic complications, and should be tailored to patient individualized characteristics to optimize stroke outcomes.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Encéfalo , Humanos , Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal
5.
Mult Scler ; 28(3): 453-462, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34595962

RESUMEN

BACKGROUND: Combined central and peripheral demyelination (CCPD) is a disease of inflammatory demyelination that affects central and peripheral nerves simultaneously or temporally separated. OBJECTIVES: This study evaluated the clinical characteristics and the existence of antinodal/paranodal antibodies in patients with CCPD. METHODS: We reviewed the clinical manifestations, laboratory tests, electrophysiological examinations, neuroimaging findings, treatment, and prognosis of 31 patients with CCPD. Using a live cell-based assay, we tested antinodal/paranodal antibodies. RESULTS: The most common symptoms were motor weakness (83.3%), hyporeflexia (63.3%), and sphincter disturbance (58.1%). In total, 16.6% of patients had impaired vision symptoms, whereas 33.3% of patients had abnormal visual-evoked potentials (VEPs). A total of 21.1% (4/19) of patients were positive for anti-AQP4 (aquaporin 4) antibodies, 20.0% (2/10) of patients were positive for anti-NF155 (neurofascin-155) antibodies, and 10.0% (1/10) of patients were positive for anti-MAG (myelin-associated glycoprotein) antibodies. The effective rates of intravenous corticosteroids, intravenous immunoglobulins, and rituximab were 72.2%, 37.5%, and 100%, respectively. At the illness peak, 75% of patients with CCPD had an mRS (modified Rankin Scale) score of 4 or greater. In remission, 37.5% had an mRS score of 4 or greater. CONCLUSION: The clinical manifestations of patients with CCPD are highly heterogeneous. We recommend testing antinodal/paranodal antibodies for patients with CCPD.


Asunto(s)
Autoanticuerpos , Enfermedades Desmielinizantes , Enfermedades Desmielinizantes/tratamiento farmacológico , Humanos , Inmunoglobulinas Intravenosas/uso terapéutico , Pronóstico , Rituximab
6.
Proc Natl Acad Sci U S A ; 116(12): 5558-5563, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30819895

RESUMEN

CD3+CD4-CD8- T cells (double-negative T cells; DNTs) have diverse functions in peripheral immune-related diseases by regulating immunological and inflammatory homeostasis. However, the functions of DNTs in the central nervous system remain unknown. Here, we found that the levels of DNTs were dramatically increased in both the brain and peripheral blood of stroke patients and in a mouse model in a time-dependent manner. The infiltrating DNTs enhanced cerebral immune and inflammatory responses and exacerbated ischemic brain injury by modulating the FasL/PTPN2/TNF-α signaling pathway. Blockade of this pathway limited DNT-mediated neuroinflammation and improved the outcomes of stroke. Our results identified a critical function of DNTs in the ischemic brain, suggesting that this unique population serves as an attractive target for the treatment of ischemic stroke.


Asunto(s)
Isquemia Encefálica/inmunología , Complejo CD3/inmunología , Accidente Cerebrovascular/inmunología , Subgrupos de Linfocitos T/inmunología , Anciano de 80 o más Años , Animales , Encéfalo/inmunología , Antígenos CD4/inmunología , Antígenos CD8/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
7.
FASEB J ; 34(8): 10107-10116, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32725952

RESUMEN

The rs6647 variant G allele in SERPINA1 gene was reported to be associated with the risk of large artery atherosclerotic stroke (LAS), however, the mechanism remains unclear. Here, we performed a functional annotation of the rs6647 variant by using the software HaploReg version 4.1 (HaploReg v4.1). Next, the expression quantitative trait loci (eQTLs) analysis of multiple datasets was conducted for determining the association between the rs6647 and SERPINA1 expression in various tissues. Then, a case-control gene expression analysis was done using two independent ischemic stroke (IS) gene expression datasets. Finally, SERPINA1 expression in whole blood samples from 8 LAS patients and 14 healthy persons were compared. The functional annotation suggested that the rs6647 regulates gene expression in multiple human tissues especially in brain and blood. The eQTLs analysis revealed a significant association of the rs6647 G allele with increased expression of SERPINA1 gene only in whole blood. Compared with the controls, there was an increased expression of SERPINA1 gene in whole blood in both IS patients and LAS patients. SERPINA1 gene expression in whole blood bridges the rs6647 variant G allele with increased LAS risk, providing new insights into the mechanisms underlying role of the rs6647 in determining LAS risk.


Asunto(s)
Aterosclerosis/genética , Expresión Génica/genética , Predisposición Genética a la Enfermedad/genética , Polimorfismo de Nucleótido Simple/genética , Accidente Cerebrovascular/genética , alfa 1-Antitripsina/genética , Adulto , Alelos , Estudios de Casos y Controles , Femenino , Perfilación de la Expresión Génica/métodos , Genotipo , Humanos , Masculino , Sitios de Carácter Cuantitativo/genética
8.
FASEB J ; 33(12): 14811-14824, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31718280

RESUMEN

Reactive oxygen species (ROS) generation and mitochondrial dysfunction are related to neuron loss in multiple sclerosis (MS). Although peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) appears to play a key role in modulating levels of mitochondrial ROS, antioxidants, and uncoupling proteins (UCPs), and PGC-1α expression is reduced in the neocortex of patients with MS, it is unclear what its role is in neurons and in the manifestation of clinical symptoms of MS. Here, we show in wild-type (WT) experimental autoimmune encephalomyelitis (EAE) mice that PGC-1α is decreased 13 d after EAE induction followed by a steady decline up to 20 d. These changes were accompanied by parallel alterations in levels of superoxide dismutase 2, peroxiredoxin 3, thioredoxin 2, UCP4, and UCP5. In transgenic (TG) mice with neuron-specific overexpression of PGC-1α (PGC-1αf/fEno2-Cre), clinical symptoms after EAE induction were delayed and less severe than in WT mice. The degrees of apoptotic neuron loss and demyelination were also less severe in PGC-1α-TG mice. Overexpression of PGC-1α in neuronal neuroblastoma spinal cord 34 cells subjected to EAE inflammatory conditions showed similar results to those obtained in vivo. RNA sequencing analysis showed that apoptotic processes were significantly enriched in the top 10 significant gene ontology (GO) terms of differentially expressed genes, and the apoptotic pathway was significantly enriched in Kyoto Encyclopedia of Genes and Genomes pathway analysis. Our findings indicate that up-regulation of neuronal PGC-1α protected neurons from apoptosis in EAE. Manipulating PGC-1α levels in MS may help stave off this devastating disease.-Dang, C., Han, B., Li, Q., Han, R., Hao, J. Up-regulation of PGC-1α in neurons protects against experimental autoimmune encephalomyelitis.


Asunto(s)
Apoptosis , Encefalomielitis Autoinmune Experimental/metabolismo , Neuronas/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Regulación hacia Arriba , Animales , Línea Celular Tumoral , Células Cultivadas , Encefalomielitis Autoinmune Experimental/genética , Femenino , Ratones , Ratones Endogámicos C57BL , Proteínas Desacopladoras Mitocondriales/genética , Proteínas Desacopladoras Mitocondriales/metabolismo , Peroxiredoxina III/genética , Peroxiredoxina III/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/metabolismo
9.
J Immunol ; 200(5): 1618-1626, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29378913

RESUMEN

Inflammation that complicates many autoimmune diseases, such as multiple sclerosis (MS), has been correlated to abnormal differentiation of Th17 cells. However, the reasons that promote Th17 cell-driven autoimmunity are yet to be discovered. In this study, we sought evidence that DNA-damage-inducible transcript 4 (DDIT4) and its associated long noncoding RNA DDIT4 (lncDDIT4) inhibit Th17 cell differentiation. We recruited 36 patients. Six MS patients and five healthy volunteers (controls) contributed PBMCs as material for microarray analysis. Microarray assays of lncDDIT4 and DDIT4 RNA expression identified outstanding differences between MS and control subjects, which were verified with real-time quantitative PCR. We then interrupted the expression of lncDDIT4 and DDIT4 mRNA in MS patients' naive CD4+ T cells and observed the resulting changes in Th17 cells. The expression of lncDDIT4 and DDIT4 mRNA were higher both in PBMCs and CD4+ T cells of MS patients than in healthy controls. DDIT4 (2.79-fold upregulation) was then recognized as a candidate for the cis-regulated target of lncDDIT4 (4.32-fold upregulation). Isolation of naive CD4+ T cells revealed enhanced levels of lncDDIT4 and DDIT4 after stimulated with Th17-inducing cytokines, but not after Th1, Th2, or T regulatory cell induction. Overexpression of lncDDIT4 in naive CD4+ T cells inhibited IL-17 transcription through increased DDIT4 expression and decreased activation of the DDIT4/mTOR pathway. Consistently, silencing lncDDIT4 in naive CD4+ T cells enhanced Th17 differentiation through increased activation of the DDIT4/mTOR pathway. However, these results vanished when DDIT4 was silenced. This outcome suggests that lncDDIT4 regulates Th17 cell differentiation by directly targeting DDIT4.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Diferenciación Celular/fisiología , ARN Largo no Codificante/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Células Th17/metabolismo , Factores de Transcripción/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Humanos , ARN Mensajero/metabolismo , Linfocitos T Reguladores/metabolismo , Células Th2/metabolismo , Regulación hacia Arriba/fisiología
10.
J Med Genet ; 56(4): 265-270, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30194086

RESUMEN

BACKGROUND: The locus for familial cortical myoclonic tremor with epilepsy (FCMTE) has long been mapped to 8q24 in linkage studies, but the causative mutations remain unclear. Recently, expansions of intronic TTTCA and TTTTA repeat motifs within SAMD12 were found to be involved in the pathogenesis of FCMTE in Japanese pedigrees. We aim to identify the causative mutations of FCMTE in Chinese pedigrees. METHODS: We performed genetic linkage analysis by microsatellite markers in a five-generation Chinese pedigree with 55 members. We also used array-comparative genomic hybridisation (CGH) and next-generation sequencing (NGS) technologies (whole-exome sequencing, capture region deep sequencing and whole-genome sequencing) to identify the causative mutations in the disease locus. Recently, we used low-coverage (~10×) long-read genome sequencing (LRS) on the PacBio Sequel and Oxford Nanopore platforms to identify the causative mutations, and used repeat-primed PCR for validation of the repeat expansions. RESULTS: Linkage analysis mapped the disease locus to 8q23.3-24.23. Array-CGH and NGS failed to identify causative mutations in this locus. LRS identified the intronic TTTCA and TTTTA repeat expansions in SAMD12 as the causative mutations, thus corroborating the recently published results in Japanese pedigrees. CONCLUSIONS: We identified the pentanucleotide repeat expansion in SAMD12 as the causative mutation in Chinese FCMTE pedigrees. Our study also suggested that LRS is an effective tool for molecular diagnosis of genetic disorders, especially for neurological diseases that cannot be positively diagnosed by conventional clinical microarray and NGS technologies.


Asunto(s)
Estudios de Asociación Genética , Intrones , Proteínas del Tejido Nervioso/genética , Linaje , Fenotipo , Secuencias Repetidas en Tándem , Adulto , Hibridación Genómica Comparativa , Epilepsias Mioclónicas/diagnóstico , Epilepsias Mioclónicas/genética , Femenino , Estudios de Asociación Genética/métodos , Humanos , Masculino , Análisis de Secuencia de ADN , Secuenciación del Exoma , Secuenciación Completa del Genoma
11.
Proc Natl Acad Sci U S A ; 114(3): E396-E405, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-27994144

RESUMEN

Astrocytes are believed to bridge interactions between infiltrating lymphocytes and neurons during brain ischemia, but the mechanisms for this action are poorly understood. Here we found that interleukin-15 (IL-15) is dramatically up-regulated in astrocytes of postmortem brain tissues from patients with ischemic stroke and in a mouse model of transient focal brain ischemia. We generated a glial fibrillary acidic protein (GFAP) promoter-controlled IL-15-expressing transgenic mouse (GFAP-IL-15tg) line and found enlarged brain infarcts, exacerbated neurodeficits after the induction of brain ischemia. In addition, knockdown of IL-15 in astrocytes attenuated ischemic brain injury. Interestingly, the accumulation of CD8+ T and natural killer (NK) cells was augmented in these GFAP-IL-15tg mice after brain ischemia. Of note, depletion of CD8+ T or NK cells attenuated ischemic brain injury in GFAP-IL-15tg mice. Furthermore, knockdown of the IL-15 receptor α or blockade of cell-to-cell contact diminished the activation and effector function of CD8+ T and NK cells in GFAP-IL-15tg mice, suggesting that astrocytic IL-15 is delivered in trans to target cells. Collectively, these findings indicate that astrocytic IL-15 could aggravate postischemic brain damage via propagation of CD8+ T and NK cell-mediated immunity.


Asunto(s)
Astrocitos/metabolismo , Lesiones Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Inmunidad Celular/inmunología , Interleucina-15/inmunología , Interleucina-15/metabolismo , Anciano de 80 o más Años , Animales , Astrocitos/inmunología , Encéfalo/inmunología , Encéfalo/metabolismo , Lesiones Encefálicas/inmunología , Isquemia Encefálica/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/inmunología , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/inmunología , Ratones Transgénicos/metabolismo , Neuroglía/inmunología , Neuroglía/metabolismo , Neuronas/inmunología , Neuronas/metabolismo , Regiones Promotoras Genéticas/inmunología , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/metabolismo , Regulación hacia Arriba/inmunología
12.
Proc Natl Acad Sci U S A ; 114(30): E6202-E6211, 2017 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-28696300

RESUMEN

The nonneural cholinergic system of immune cells is pivotal for the maintenance of immunological homeostasis. Here we demonstrate the expression of choline acetyltransferase (ChAT) and cholinergic enzymes in murine natural killer (NK) cells. The capacity for acetylcholine synthesis by NK cells increased markedly under inflammatory conditions such as experimental autoimmune encephalomyelitis (EAE), in which ChAT expression escalated along with the maturation of NK cells. ChAT+ and ChAT- NK cells displayed distinctive features in terms of cytotoxicity and chemokine/cytokine production. Transfer of ChAT+ NK cells into the cerebral ventricles of CX3CR1-/- mice reduced brain and spinal cord damage after EAE induction, and decreased the numbers of CNS-infiltrating CCR2+Ly6Chi monocytes. ChAT+ NK cells killed CCR2+Ly6Chi monocytes directly via the disruption of tolerance and inhibited the production of proinflammatory cytokines. Interestingly, ChAT+ NK cells and CCR2+Ly6Chi monocytes formed immune synapses; moreover, the impact of ChAT+ NK cells was mediated by α7-nicotinic acetylcholine receptors. Finally, the NK cell cholinergic system up-regulated in response to autoimmune activation in multiple sclerosis, perhaps reflecting the severity of disease. Therefore, this study extends our understanding of the nonneural cholinergic system and the protective immune effect of acetylcholine-producing NK cells in autoimmune diseases.


Asunto(s)
Acetilcolina/metabolismo , Sistema Nervioso Central/patología , Encefalomielitis/patología , Células Asesinas Naturales/fisiología , Macrófagos/fisiología , Monocitos/fisiología , Animales , Encefalomielitis/inmunología , Humanos , Células Asesinas Naturales/metabolismo , Ratones , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Sistema Colinérgico no Neuronal/inmunología , Sistema Colinérgico no Neuronal/fisiología
13.
BMC Neurol ; 19(1): 206, 2019 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-31443692

RESUMEN

BACKGROUND: Subcortical ischemic vascular dementia (SIVD) is very common among the older people, but has no approved treatment. Preclinical trials show that remote ischemic conditioning (RIC) reduces recurrence of ischemic stroke. We hypothesize that RIC may also be an effective therapy for patients with SIVD. METHODS: Thirty-seven consecutive SIVD cases were enrolled in this randomized control study. Eighteen RIC patients underwent five brief cycles of conditioning (bilateral upper limb compression at 200 mmHg) followed by reperfusion twice daily over 6 consecutive months. Nineteen control patients underwent the same process, but at a pressure of 60 mmHg which caused no restriction on the blood flow of the upper limb. The primary outcome measures were changes in neuropsychological assessments. The secondary outcomes included the changes in high-sensitive C-reactive protein (hs-CRP) concentration, white matter lesion volume (WMLV), diffusion tension imaging (DTI) metrics of white matter. All data were collected at baseline and follow-up. RESULTS: A significant treatment difference favoring RIC at 6 months was observed on performance of Hopkins Verbal Learning Test-Revised (HVLT-R), Controlled Oral Word Association Test (COWAT), Trail Making Test A and B (TMT-A & TMT-B), and Judgment of Line Orientation (JLO) (p < 0.05). The control group did not show much improvement after the treatment, and only with a slight change in HVLT-R and TMT-R (p < 0.05). Covariance analysis of efficacy between the two groups suggested that RIC patients performed better on JLO than control patients at the 6-month follow-up (RIC 23.10 vs. control 18.56; p = 0.013). Although DTI metrics were comparable, Hs-CRP levels and WMLV in RIC patients showed a declining trend. CONCLUSIONS: Over the 6-month treatment period, we found that RIC was safe and effective for improving cognitive function in SIVD patients. TRIAL REGISTRATION: Clinical Trial Registration ( http://www.clinicaltrials.gov ), Unique identifier: NCT03022149; Retrospectively registered; Date of registration: January 16, 2017.


Asunto(s)
Encéfalo/irrigación sanguínea , Cognición , Demencia Vascular , Precondicionamiento Isquémico/métodos , Anciano , Anciano de 80 o más Años , Encéfalo/patología , Encéfalo/fisiopatología , Demencia Vascular/patología , Demencia Vascular/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Extremidad Superior/fisiopatología
14.
J Immunol ; 198(7): 2626-2639, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28228555

RESUMEN

IL-17-producing Th17 cells have gradually become considered as key factors in the pathogenesis of many autoimmune diseases, including multiple sclerosis (MS). Although the involvement of certain microRNAs in the development of MS has been reported, their role in Th17-driven autoimmunity is still poorly understood. In this study, we identified microRNA (miR)-15b as an important factor in Th17-associated effects and determined that the expression of miR-15b is significantly downregulated in MS patients and in mice with experimental autoimmune encephalomyelitis. Overexpression of miR-15b alleviated experimental autoimmune encephalomyelitis, whereas knockdown of miR-15b aggravated it. We demonstrated that miR-15b suppressed Th17 differentiation both in vivo and in vitro. We also found that O-linked N-acetylglucosamine transferase is a potential target of miR-15b, enabling it to affect the transcriptional regulation of retinoic acid-related orphan receptor γT through O-linked N-acetylglucosamine glycosylation of NF-κB. These results contribute to the importance of miR-15b in Th17 differentiation and the pathogenesis of MS.


Asunto(s)
Regulación de la Expresión Génica/inmunología , MicroARNs/inmunología , Esclerosis Múltiple/inmunología , N-Acetilglucosaminiltransferasas/biosíntesis , Células Th17/inmunología , Animales , Western Blotting , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Separación Celular , Regulación hacia Abajo , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Humanos , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Esclerosis Múltiple/genética , N-Acetilglucosaminiltransferasas/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
FASEB J ; 31(2): 519-525, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27756768

RESUMEN

In this study, we strove to substantiate the ability of linc-MAF-4 to act as a regulator of pathogenesis during multiple sclerosis (MS). We recruited 34 patients who were diagnosed with MS according to the revised McDonald criteria. Six patients with MS and 5 healthy volunteers contributed peripheral blood mononuclear cells for microarray analysis. Subsequent knockdown and overexpression of linc-MAF-4 in naive CD4+ T cells from the additional 28 patients with MS was performed to track changes in CD4+ T-cell subsets and their function, as well as to confirm results from the prior microarray analysis. Expression of linc-MAF-4 increased significantly in peripheral blood mononuclear cells of patients with MS compared with those of control participants. In addition, linc-MAF-4 regulated encephalitogenic T helper (Th)1-cell differentiation in patients with MS. Transfection of synthetic linc-MAF-4 into naive CD4+ T cells facilitated Th1-cell differentiation and inhibited Th2-cell differentiation by directly inhibiting MAF, which is a Th2-cell transcription factor. Linc-MAF-4 also promoted activation of CD4+ T cells from patients with MS. Expression level of linc-MAF-4 correlated with the annual relapse rate in patients with MS. Our results suggest that linc-MAF-4 is involved in the pathogenesis of MS, specifically via regulation of encephalitogenic T cells.-Zhang, F., Liu, G., Wei, C., Gao, C., Hao, J. Linc-MAF-4 regulates Th1/Th2 differentiation and is associated with the pathogenesis of multiple sclerosis by targeting MAF.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Esclerosis Múltiple/metabolismo , ARN Largo no Codificante/metabolismo , Células TH1/fisiología , Células Th2/fisiología , Adolescente , Adulto , Regulación hacia Abajo/fisiología , Femenino , Humanos , Elementos de Nucleótido Esparcido Largo , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Largo no Codificante/genética , Regulación hacia Arriba , Adulto Joven
16.
FASEB J ; 31(4): 1756-1766, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28096232

RESUMEN

Experimental autoimmune neuritis (EAN) is a CD4+ T-cell-mediated autoimmune inflammatory demyelinating disease of the peripheral nervous system. It has been replicated in an animal model of human inflammatory demyelinating polyradiculoneuropathy, Guillain-Barré syndrome. In this study, we evaluated the therapeutic efficacy of a selective inhibitor of the immunoproteasome subunit, low-MW polypeptide 7 (PR-957) in rats with EAN. Our results showed that PR-957 significantly delayed onset day, reduced severity and shortened duration of EAN, and alleviated demyelination and inflammatory infiltration in sciatic nerves. In addition to significantly regulating expression of the cytokine profile, PR-957 treatment down-regulated the proportion of proinflammatory T-helper (Th)17 cells in sciatic nerves and spleens of rats with EAN. Data presented show the role of PR-957 in the signal transducer and activator of transcription 3 (STAT3) pathway. PR-957 not only decreased expression of IL-6 and IL-23 but also led to down-regulation of STAT3 phosphorylation in CD4+ T cells. Regulation of the STAT3 pathway led to a reduction in retinoid-related orphan nuclear receptor γ t and IL-17 production. Furthermore, reduction of STAT3 phosphorylation may have directly suppressed Th17-cell differentiation. Therefore, our study demonstrates that PR-957 could potently alleviate inflammation in rats with EAN and that it may be a likely candidate for treating Guillain-Barré syndrome.-Liu, H., Wan, C., Ding, Y., Han, R., He, Y., Xiao, J., Hao, J. PR-957, a selective inhibitor of immunoproteasome subunit low-MW polypeptide 7, attenuates experimental autoimmune neuritis by suppressing Th17-cell differentiation and regulating cytokine production.


Asunto(s)
Diferenciación Celular , Interleucinas/metabolismo , Neuritis Autoinmune Experimental/tratamiento farmacológico , Oligopéptidos/uso terapéutico , Inhibidores de Proteasoma/uso terapéutico , Células Th17/efectos de los fármacos , Animales , Interleucinas/genética , Masculino , Oligopéptidos/farmacología , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Ratas , Ratas Endogámicas Lew , Factor de Transcripción STAT3/metabolismo , Células Th17/citología , Células Th17/metabolismo
17.
J Immunol ; 196(5): 2095-108, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26810225

RESUMEN

Myeloid cells, including proinflammatory monocytes and neutrophils, have important roles in the pathology of multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). These cells infiltrate the CNS in the early stages of disease development and contribute to the inflammatory response that is associated with symptom severity. It is thus crucial to identify and understand new mechanisms that can regulate the CNS infiltration of proinflammatory myeloid cells. Nicotinic acetylcholine receptors (nAChRs) have been increasingly studied for their immune-regulatory properties. In this study, we assessed the ability of nicotine, an nAChR ligand, to modulate proinflammatory myeloid cell numbers within the bone marrow, spleen, blood, and CNS of EAE mice. We found that nicotine significantly inhibits the infiltration of proinflammatory monocytes and neutrophils into the CNS at time points where these cells are known to play critical roles in disease pathology. In contrast, nicotine does not affect the expansion of other monocytes. We also show that nicotine exerts these effects by acting on α7 and α9 nAChR subtypes. Finally, mRNA transcript levels for CCL2 and CXCL2, chemokines involved in the chemotaxis of proinflammatory monocytes and neutrophils, respectively, are reduced in the brain of nicotine-treated EAE mice before the massive infiltration of these cells. Taken together, our data provide evidence that nAChRs can regulate proinflammatory cell infiltration into the CNS, which could be of significant value for the treatment of neuroinflammatory disorders.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Monocitos/inmunología , Neutrófilos/inmunología , Receptores Nicotínicos/inmunología , Animales , Antígenos Ly/inmunología , Encéfalo/inmunología , Separación Celular , Quimiotaxis de Leucocito/efectos de los fármacos , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/inmunología , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores CCR2/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Médula Espinal/inmunología
18.
J Immunol ; 197(10): 3831-3840, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27798164

RESUMEN

Programmed death 1 (PD-1; CD279), a member of the CD28 family, is an inhibitory receptor on T cells and is responsible for T cell dysfunction in infectious diseases and cancers. The ligand for PD-1, programmed death ligand 1 (PD-L1; also known as B7-H1, CD274), is a member of the B7 family. The engagement of PD-1 with programmed death ligand can downregulate autoreactive T cells that participate in multiple autoimmune diseases. Experimental autoimmune neuritis (EAN) is an animal model of Guillain-Barré syndrome, and the pathogenesis of EAN is mediated principally through T cells and macrophages. In this study, we investigated the effects of PD-L1 in EAN rats. For preventative and therapeutic management, we administered PD-L1, which successfully decreased the severity of EAN; it alleviated the neurologic course of EAN, as well as inhibited the infiltration of inflammatory cells and demyelination of sciatic nerves. Our data revealed that PD-L1 treatment inhibited lymphocyte proliferation and altered T cell differentiation by inducing decreases in IFN-γ+CD4+ Th1 cells and IL-17+CD4+ Th17 cells and increases in IL-4+CD4+ Th2 cells and Foxp3+CD4+ regulatory T cells. The expression levels of p-STAT3 and Foxp3 were significantly different in PD-L1-treated groups compared with the control group. Additionally, PD-L1 regulated the expression of Foxp3 and p-STAT3 in EAN, probably by inhibiting PI3K/AKT/mTOR signaling expression. In summary, PD-L1 is a potentially useful agent for the treatment of EAN because of its anti-inflammatory and neuroprotective effects.


Asunto(s)
Antígeno B7-H1/farmacología , Neuritis Autoinmune Experimental/inmunología , Neuritis Autoinmune Experimental/terapia , Sistema Nervioso Periférico/inmunología , Animales , Antígeno B7-H1/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Enfermedades Desmielinizantes/prevención & control , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Síndrome de Guillain-Barré/inmunología , Interferón gamma/efectos de los fármacos , Interleucina-17/inmunología , Interleucina-4/inmunología , Activación de Linfocitos , Neuritis Autoinmune Experimental/fisiopatología , Ratas , Nervio Ciático/efectos de los fármacos , Linfocitos T Reguladores , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th2
19.
J Immunol ; 197(9): 3471-3480, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27671112

RESUMEN

A major hurdle for effective stem cell therapy is ongoing inflammation in the target organ. Reconditioning the lesion microenvironment may be an effective way to promote stem cell therapy. In this study, we showed that engineered neural stem cells (NSCs) with complement factor H-related protein 1, a complement inhibitor protein, can attenuate inflammatory infiltration and immune-mediated damage of astrocytes, an important pathogenic progress in patients with neuromyelitis optica spectrum disorders. Furthermore, we demonstrated that transplantation of the complement factor H-related protein 1-modified NSCs effectively blocked the complement activation cascade and inhibited formation of the membrane attack complex, thus contributing to the protection of endogenous and transplanted NSC-differentiated astrocytes. Therefore, manipulation of the lesion microenvironment contributes to a more effective cell replacement therapeutic strategy for autoimmune diseases of the CNS.


Asunto(s)
Astrocitos/fisiología , Proteínas Inactivadoras del Complemento C3b/metabolismo , Células-Madre Neurales/fisiología , Neuromielitis Óptica/inmunología , Neuroprotección , Trasplante de Células Madre , Adulto , Animales , Acuaporina 4/inmunología , Autoanticuerpos/metabolismo , Autoantígenos/inmunología , Células Cultivadas , Activación de Complemento , Proteínas Inactivadoras del Complemento C3b/genética , Modelos Animales de Enfermedad , Femenino , Terapia Genética , Humanos , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Células-Madre Neurales/trasplante , Neuromielitis Óptica/terapia , Adulto Joven
20.
Cell Mol Life Sci ; 74(20): 3667-3686, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28534084

RESUMEN

O-linked ß-N-acetylglucosaminylation (O-GlcNAcylation) is involved in the regulation of many cellular cascades and neurological diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and stroke. In the brain, the expression of O-GlcNAcylation is notably heightened, as is that of O-linked N-acetylglucosaminyltransferase (OGT) and ß-N-acetylglucosaminidase (OGA), the presence of which is prominent in many regions of neurological importance. Most importantly, O-GlcNAcylation is believed to contribute to the normal functioning of neurons; conversely, its dysregulation participates in the pathogenesis of neurological disorders. In neurodegenerative diseases, O-GlcNAcylation of the brain's key proteins, such as tau and amyloid-ß, interacts with their phosphorylation, thereby triggering the formation of neurofibrillary tangles and amyloid plaques. An increase of O-GlcNAcylation by pharmacological intervention prevents neuronal loss. Additionally, O-GlcNAcylation is stress sensitive, and its elevation is cytoprotective. Increased O-GlcNAcylation ameliorated brain damage in victims of both trauma-hemorrhage and stroke. In this review, we summarize the current understanding of O-GlcNAcylation's physiological and pathological roles in the nervous system and provide a foundation for development of a therapeutic strategy for neurological disorders.


Asunto(s)
Acetilglucosamina/metabolismo , Encéfalo/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas/metabolismo , Acetilglucosaminidasa/metabolismo , Enfermedad de Alzheimer/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Humanos , N-Acetilglucosaminiltransferasas/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA