Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Eur J Neurosci ; 59(2): 192-207, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38145884

RESUMEN

Skeletal muscle is striated muscle that moves autonomously and is innervated by peripheral nerves. Peripheral nerve injury is very common in clinical treatment. However, the commonly used treatment methods often focus on the regeneration of the injured nerve but overlook the pathological changes in the injured skeletal muscle. Acupuncture, as the main treatment for denervated skeletal muscle atrophy, is used extensively in clinical practice. In the present study, a mouse model of lower limb sciatic nerve detachment was constructed and treated with electroacupuncture Stomach 36 to observe the atrophy of lower limb skeletal muscle and changes in skeletal muscle fibre types before and after electroacupuncture Stomach 36 treatment. Mice with skeletal muscle denervation showed a decrease in the proportion of IIa muscle fibres and an increase in the proportion of IIb muscle fibres, after electroacupuncture Stomach 36. The changes were reversed by specific activators of p38 MAPK, which increased IIa myofibre ratio. The results suggest that electroacupuncture Stomach 36 can reverse the change of muscle fibre type from IIb to IIa after denervation of skeletal muscle by inhibiting p38 MAPK. The results provide an important theoretical basis for the treatment of clinical peripheral nerve injury diseases with electroacupuncture, in addition to novel insights that could facilitate the study of pathological changes of denervated skeletal muscle.


Asunto(s)
Electroacupuntura , Traumatismos de los Nervios Periféricos , Ratas , Ratones , Animales , Ratas Sprague-Dawley , Traumatismos de los Nervios Periféricos/terapia , Fibras Musculares Esqueléticas , Músculo Esquelético , Nervio Ciático/lesiones , Atrofia Muscular/terapia , Proteínas Quinasas p38 Activadas por Mitógenos
2.
J Neuroinflammation ; 21(1): 10, 2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38178152

RESUMEN

Myasthenia gravis is an autoimmune disease characterized by pathogenic antibodies that target structures of the neuromuscular junction. However, some patients also experience autonomic dysfunction, anxiety, depression, and other neurological symptoms, suggesting the complex nature of the neurological manifestations. With the aim of explaining the symptoms related to the central nervous system, we utilized a rat model to investigate the impact of dopamine signaling in the central nervous and peripheral circulation. We adopted several screening methods, including western blot, quantitative PCR, mass spectrum technique, immunohistochemistry, immunofluorescence staining, and flow cytometry. In this study, we observed increased and activated dopamine signaling in both the central nervous system and peripheral circulation of myasthenia gravis rats. Furthermore, changes in the expression of two key molecules, Claudin5 and CD31, in endothelial cells of the blood-brain barrier were also examined in these rats. We also confirmed that dopamine incubation reduced the expression of ZO1, Claudin5, and CD31 in endothelial cells by inhibiting the Wnt/ß-catenin signaling pathway. Overall, this study provides novel evidence suggesting that pathologically elevated dopamine in both the central nervous and peripheral circulation of myasthenia gravis rats impair brain-blood barrier integrity by inhibiting junction protein expression in brain microvascular endothelial cells through the Wnt/ß-catenin pathway.


Asunto(s)
Dopamina , Miastenia Gravis , Humanos , Ratas , Animales , Dopamina/metabolismo , Células Endoteliales/metabolismo , Encéfalo , Barrera Hematoencefálica/metabolismo , Vía de Señalización Wnt/fisiología , Miastenia Gravis/metabolismo
3.
Mikrochim Acta ; 191(6): 328, 2024 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-38743383

RESUMEN

The instant screening of patients with a tendency towards developing Alzheimer's disease (AD) is significant for providing preventive measures and treatment. However, the current imaging-based technology cannot meet the requirements in the early stage. Developing biosensor-based liquid biopsy technology could be overcoming this bottleneck problem. Herein, we developed a simple, low-cost, and sensitive electrochemical aptamer biosensor for detecting phosphorylated tau protein threonine 231 (P-tau231), the earliest and one of the most efficacious abnormally elevated biomarkers of AD. Gold nanoparticles (AuNPs) were electrochemically synthesized on a glassy carbon electrode as the transducer, exhibiting excellent conductivity, and were applied to amplify the electrochemical signal. A nucleic acid aptamer was designed as the receptor to capture the P-tau231 protein, specifically through the formation of an aptamer-antigen complex. The proposed biosensor showed excellent sensitivity in detecting P-tau 231, with a broad linear detection range from 10 to 107 pg/mL and a limit of detection (LOD) of 2.31 pg/mL. The recoveries of the biosensor in human serum ranged from 97.59 to 103.26%, demonstrating that the biosensor could be used in complex practical samples. In addition, the results showed that the developed biosensor has good repeatability, reproducibility, and stability, which provides a novel method for the early screening of AD.


Asunto(s)
Enfermedad de Alzheimer , Aptámeros de Nucleótidos , Técnicas Biosensibles , Técnicas Electroquímicas , Oro , Límite de Detección , Nanopartículas del Metal , Proteínas tau , Humanos , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/diagnóstico , Aptámeros de Nucleótidos/química , Proteínas tau/sangre , Técnicas Biosensibles/métodos , Técnicas Electroquímicas/métodos , Técnicas Electroquímicas/instrumentación , Oro/química , Nanopartículas del Metal/química , Fosforilación , Biomarcadores/sangre
4.
Bioinformatics ; 37(16): 2491-2493, 2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-33459772

RESUMEN

SUMMARY: Cancer can be classified into various subtypes by its molecular, histological or clinical characteristics. Discovering cancer-subtype-specific drugs is a crucial step in personalized medicine. SubtypeDrug is a system biology R-based software package that enables the prioritization of subtype-specific drugs based on cancer expression data from samples of many subtypes. This provides a novel approach to identify the subtype-specific drug by considering biological functions regulated by drugs at the subpathway level. The operation modes include extraction of subpathways from biological pathways, identification of dysregulated subpathways induced by each drug, inference of sample-specific subpathway activity profiles, evaluation of drug-disease reverse association at the subpathways level, identification of cancer-subtype-specific drugs through subtype sample set enrichment analysis, and visualization of the results. Its capabilities enable SubtypeDrug to find subtype-specific drugs, which will fill the gaps in the recent tools which only identify the drugs for a particular cancer type. SubtypeDrug may help to facilitate the development of tailored treatment for patients with cancer. AVAILABILITY AND IMPLEMENTATION: The package is implemented in R and available under GPL-2 license from the CRAN website (https://CRAN.R-project.org/package=SubtypeDrug). SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

5.
Bioinformatics ; 36(7): 2303-2305, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31821408

RESUMEN

SUMMARY: Subpathways, which are defined as local gene subregions within a biological pathway, have been reported to be associated with the occurrence and development of cancer. The recent subpathway identification tools generally identify differentially expressed subpathways between normal and cancer samples. psSubpathway is a novel systems biology R-based software package that enables flexible identification of phenotype-specific subpathways in a cancer dataset with multiple categories (such as multiple subtypes and developmental stages of cancer). The operation modes include extraction of subpathways from pathway networks, inference with subpathway activities in the context of gene expression data, identification of subtype-specific subpathways, identification of dynamic-changed subpathways associated with the cancer developmental stage and visualization of subpathway activities of samples in different phenotypes. Its capabilities enable psSubpathway to find specific abnormal subpathways in the datasets with multi-phenotype categories and to fill the gaps in the recent tools. psSubpathway may identify more specific biomarkers to facilitate the development of tailored treatment for patients with cancer. AVAILABILITY AND IMPLEMENTATION: The package is implemented in R and available under GPL-2 license from the CRAN website (https://cran.r-project.org/web/packages/psSubpathway/). SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Neoplasias , Programas Informáticos , Expresión Génica , Humanos , Fenotipo
6.
FASEB J ; 34(10): 13762-13775, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32808351

RESUMEN

Cerebral ischemia causes damage to the structure and function of the blood-brain barrier (BBB) and alleviating BBB destruction will be of great significance for the treatment and prognosis of ischemic stroke. Recently, microRNAs have been shown to play a critical role in BBB integrity. However, the potential mechanism by which microRNA-182 (miR-182) affects the BBB in ischemic stroke remains unclear. We demonstrated for the first time that cerebral ischemia leads to a significant progressive increase in miR-182 after pMCAO, and bEnd.3 cells are the primary target cells of miR-182. In miR-182 KD transgenic mice, infarct volume, and BBB permeability were attenuated, and tight junction (TJ) proteins increased. Inhibition of miR-182 with an antagomir reduced OGD-induced apoptosis of bEnd.3 cells and the loss of ZO-1 and Occludin. To further explore the mechanism by which miR-182 regulates BBB integrity, we detected the apoptotic proteins Bcl-2/Bax and demonstrated that mTOR and FOXO1 were the targets of miR-182. Inhibition of mTOR/FOXO1 by rapamycin/AS1842856 decreased the ratio of Bcl-2/Bax and exacerbated TJ protein loss. Taken together, inhibition of miR-182 protects BBB integrity by reducing endothelial cell apoptosis through the mTOR/FOXO1 pathway. Thus, miR-182 may be a potential target for the treatment of BBB disruption during cerebral ischemia.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , MicroARNs/metabolismo , Transducción de Señal , Animales , Apoptosis , Línea Celular , Células Cultivadas , Regulación hacia Abajo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Infarto de la Arteria Cerebral Media/genética , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Uniones Estrechas/genética , Proteínas de Uniones Estrechas/metabolismo
7.
Eur J Immunol ; 49(12): 2184-2194, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31411745

RESUMEN

MicroRNA 182 is important for the clonal expansion of CD4+ T cells (Th) following IL-2 stimulation and is a potential therapeutic target for autoimmune diseases. In the present study, we investigated the role of microRNA 182 in the differentiation of pro-inflammatory CD4+ T helper cell by overexpressing or silencing microRNA 182 expression both in in vivo and in vitro settings. We report that in the studied Chinese cohort, microRNA 182 is upregulated in patients with relapse and remitting multiple sclerosis (RRMS) and this upregulation is associated with increased IFN-γ producing CD4+ Th1 cells in the circulation. In the murine experimental autoimmune encephalomyelitis (EAE) model, global microRNA 182 overexpression exacerbates clinical symptoms and results in augmented CD4+ IFN-γ+ Th1 and CD4+ IL-17+ Th17 differentiation in vivo. Addition of microRNA 182 mimics in vitro represses both the protein expression and transcriptional activity of hypoxia induced factor 1α (HIF-1α) but increases the level of IFN-γ transcripts in sorted murine CD4+ T cells. Together, our results provide evidence that microRNA 182 may be one of the transitional hubs contribution to regulate Th cells expansion in response to self-antigens and differentiation of antigen specific Th cells during the progression of autoimmune inflammations.


Asunto(s)
Diferenciación Celular/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , MicroARNs/inmunología , Esclerosis Múltiple/inmunología , Células TH1/inmunología , Animales , Encefalomielitis Autoinmune Experimental/patología , Femenino , Interferón gamma/inmunología , Interleucina-17/inmunología , Ratones , Esclerosis Múltiple/patología , Células TH1/patología , Células Th17/inmunología , Células Th17/patología
8.
Mol Cell ; 46(4): 484-94, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22542455

RESUMEN

The NAD-dependent histone deacetylase Sirt1 antagonizes p53 transcriptional activity to regulate cell-cycle progression and apoptosis. We have identified a ubiquitin-specific peptidase, USP22, one of the 11 death-from-cancer signature genes that are critical in controlling cell growth and death, as a positive regulator of Sirt1. USP22 interacts with and stabilizes Sirt1 by removing polyubiquitin chains conjugated onto Sirt1. The USP22-mediated stabilization of Sirt1 leads to decreasing levels of p53 acetylation and suppression of p53-mediated functions. In contrast, depletion of endogenous USP22 by RNA interference destabilizes Sirt1, inhibits Sirt1-mediated deacetylation of p53 and elevates p53-dependent apoptosis. Genetic deletion of the usp22 gene results in Sirt1 instability, elevated p53 transcriptional activity and early embryonic lethality in mice. Our study elucidates a molecular mechanism in suppression of cell apoptosis by stabilizing Sirt1 in response to DNA damage and reveals a critical physiological function of USP22 in mouse embryonic development.


Asunto(s)
Desarrollo Embrionario/fisiología , Endopeptidasas/metabolismo , Sirtuina 1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Daño del ADN , Desarrollo Embrionario/genética , Endopeptidasas/deficiencia , Endopeptidasas/genética , Estabilidad de Enzimas , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sirtuina 1/genética , Activación Transcripcional , Proteasas Ubiquitina-Específicas , Ubiquitinación
9.
J Immunol ; 198(10): 3927-3938, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28424240

RESUMEN

Histone acetyltransferases (HATs) regulate inducible transcription in multiple cellular processes and during inflammatory and immune response. However, the functions of general control nonrepressed-protein 5 (Gcn5), an evolutionarily conserved HAT from yeast to human, in immune regulation remain unappreciated. In this study, we conditionally deleted Gcn5 (encoded by the Kat2a gene) specifically in T lymphocytes by crossing floxed Gcn5 and Lck-Cre mice, and demonstrated that Gcn5 plays important roles in multiple stages of T cell functions including development, clonal expansion, and differentiation. Loss of Gcn5 functions impaired T cell proliferation, IL-2 production, and Th1/Th17, but not Th2 and regulatory T cell differentiation. Gcn5 is recruited onto the il-2 promoter by interacting with the NFAT in T cells upon TCR stimulation. Interestingly, instead of directly acetylating NFAT, Gcn5 catalyzes histone H3 lysine H9 acetylation to promote IL-2 production. T cell-specific suppression of Gcn5 partially protected mice from myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis, an experimental model for human multiple sclerosis. Our study reveals previously unknown physiological functions for Gcn5 and a molecular mechanism underlying these functions in regulating T cell immunity. Hence Gcn5 may be an important new target for autoimmune disease therapy.


Asunto(s)
Histona Acetiltransferasas/metabolismo , Activación de Linfocitos , Linfocitos T/inmunología , Animales , Diferenciación Celular , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Regulación de la Expresión Génica , Histona Acetiltransferasas/deficiencia , Histona Acetiltransferasas/genética , Interleucina-2/deficiencia , Interleucina-2/genética , Interleucina-2/inmunología , Ratones , Factores de Transcripción NFATC/genética , Regiones Promotoras Genéticas , Unión Proteica , Procesamiento Proteico-Postraduccional , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/fisiología , Células TH1/inmunología , Células TH1/fisiología , Células Th2/inmunología , Células Th2/fisiología
10.
Genomics ; 110(6): 337-346, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29366861

RESUMEN

Long non-coding RNAs have the potential to regulate immune responses. Their impact on multiple sclerosis has remained elusive. For illustrating their roles in experimental autoimmune encephalomyelitis (EAE) pathogenesis, we investigated the differential expression of lncRNAs and mRNAs in CD4+Th cells obtained from myelin oligodendrocytic glycoprotein35-55(MOG35-55)-induced EAE and complete Freund's adjuvant (CFA) controls. We observed differential expression of 1112 lncRNAs and 519 mRNAs in CD4+Th cells. The functional network showed lncRNAs had the capacity to modulate EAE pathogenesis via regulating many known EAE regulators such as Ptpn6. Predicting the function of lncRNAs demonstrated that dysregulated lncRNAs were closely associated with the development of EAE. These dysregulated lncRNAs may have function in EAE and they could be novel biomarkers and therapeutic targets of EAE. However, the precise mechanisms and biological functions of these specific lncRNAs in EAE pathogenesis require further study.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Regulación de la Expresión Génica , ARN Largo no Codificante/genética , ARN Mensajero/genética , Linfocitos T Colaboradores-Inductores/metabolismo , Animales , Encefalomielitis Autoinmune Experimental/genética , Femenino , Perfilación de la Expresión Génica , Ratones , Glicoproteína Mielina-Oligodendrócito/farmacología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Linfocitos T Colaboradores-Inductores/efectos de los fármacos
11.
Immunol Invest ; 47(7): 712-724, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29944018

RESUMEN

Interleukin-9 (IL-9) is a multifunctional cytokine involved in protective immunity or immunopathology depending on the microenvironment and specific disease settings. Our early study determined that IL-9 and Th9 cells participate in and promote the progression of experimental autoimmune myasthenia gravis (EAMG). The data from this study showed that exogenous recombinant rat IL-9 (rrIL-9) acted as an IL-9 receptor antagonist, reduced the incidence of EAMG in rats, alleviated the severity of the disease, and reduced the anti-acetylcholine receptor (AChR) IgG antibody levels by altering the Th-subset distribution. These data suggest that administration of rrIL-9 may provide a novel therapeutic strategy against MG or related autoimmune diseases. Abbreviations: 2-Mercaptoethanol (2-ME); antibodies (Abs); ?-bungarotoxin (?-BTX); acetylcholine receptor (AChR); airway hyper-reactivity (AHR); allophycocyanin-conjugated (APC); antigen presenting cells (APCs); complete Freund's adjuvant (CFA); Cyanine dye 3 (Cy3); dendritic cells (DCs); experimental autoimmune encephalomyelitis (EAE); experimental autoimmune myasthenia gravis (EAMG); flow cytometry (FACS); fetal bovine serum (FBS); fetal calf serum (FCS); Fluorescein isothiocyanate (FITC); gamma chain (?c); intraperitoneally (i.p.); Incomplete Freund's adjuvant (IFA); interferon (IFN); immunoglobulin (Ig); Interleukin (IL); Janus kinase (JAK); myasthenia gravis (MG); Mononuclear cells (MNC); neuromuscular junctions (NMJ); optical density (OD); ovalbumin (OVA); phosphate-buffered saline (PBS); phycoerythrin (PE); Peridinin chlorophyll protein complex (Percp); Rat AChR ? subunit (R-AChR97-116); Recombinant Rat (rr); room temperature (RT); signal transducer and activator of transcription (STAT); T helper cells (Th).


Asunto(s)
Inmunoterapia/métodos , Interleucina-9/inmunología , Miastenia Gravis Autoinmune Experimental/terapia , Miastenia Gravis/terapia , Proteínas Recombinantes/inmunología , Animales , Autoanticuerpos/sangre , Autoantígenos/inmunología , Femenino , Humanos , Interleucina-9/uso terapéutico , Miastenia Gravis/inmunología , Miastenia Gravis Autoinmune Experimental/inmunología , Péptidos/inmunología , Ratas , Ratas Endogámicas Lew , Receptores Colinérgicos/inmunología , Receptores de Interleucina-9/antagonistas & inhibidores , Proteínas Recombinantes/uso terapéutico
12.
Mol Cell Neurosci ; 83: 65-73, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28684360

RESUMEN

Regulator of G protein signaling 5 (RGS5) acts as a GTPase-activating protein (GAP) for the Gαi subunit and negatively regulates G protein-coupled receptor signaling. However, its presence and function in postmitotic differentiated primary neurons remains largely uncharacterized. During neural development, sonic hedgehog (Shh) signaling is involved in cell signaling pathways via Gαi activity. In particular, Shh signaling is essential for embryonic neural tube patterning, which has been implicated in neuronal polarization involving neurite outgrowth. Here, we examined whether RGS5 regulates Shh signaling in neurons. RGS5 transcripts were found to be expressed in cortical neurons and their expression gradually declined in a time-dependent manner in culture system. When an adenovirus expressing RGS5 was introduced into an in vitro cell culture model of cortical neurons, RGS5 overexpression significantly reduced neurite outgrowth and FM4-64 uptake, while cAMP-PKA signaling was also affected. These findings suggest that RGS5 inhibits Shh function during neurite outgrowth and the presynaptic terminals of primary cortical neurons mature via modulation of cAMP.


Asunto(s)
Proteínas Hedgehog/metabolismo , Neuronas/metabolismo , Proteínas RGS/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Corteza Cerebral/citología , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ratones , Ratones Endogámicos C57BL , Proyección Neuronal , Neuronas/citología , Proteínas RGS/genética
13.
EMBO J ; 32(18): 2477-90, 2013 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-23942232

RESUMEN

In rheumatoid arthritis (RA), macrophage is one of the major sources of inflammatory mediators. Macrophages produce inflammatory cytokines through toll-like receptor (TLR)-mediated signalling during RA. Herein, we studied macrophages from the synovial fluid of RA patients and observed a significant increase in activation of inositol-requiring enzyme 1α (IRE1α), a primary unfolded protein response (UPR) transducer. Myeloid-specific deletion of the IRE1α gene protected mice from inflammatory arthritis, and treatment with the IRE1α-specific inhibitor 4U8C attenuated joint inflammation in mice. IRE1α was required for optimal production of pro-inflammatory cytokines as evidenced by impaired TLR-induced cytokine production in IRE1α-null macrophages and neutrophils. Further analyses demonstrated that tumour necrosis factor (TNF) receptor-associated factor 6 (TRAF6) plays a key role in TLR-mediated IRE1α activation by catalysing IRE1α ubiquitination and blocking the recruitment of protein phosphatase 2A (PP2A), a phosphatase that inhibits IRE1α phosphorylation. In summary, we discovered a novel regulatory axis through TRAF6-mediated IRE1α ubiquitination in regulating TLR-induced IRE1α activation in pro-inflammatory cytokine production, and demonstrated that IRE1α is a potential therapeutic target for inflammatory arthritis.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Citocinas/metabolismo , Endorribonucleasas/metabolismo , Activación Enzimática/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Toll-Like/metabolismo , Respuesta de Proteína Desplegada/fisiología , Animales , Western Blotting , Línea Celular , Sistemas de Liberación de Medicamentos , Endorribonucleasas/antagonistas & inhibidores , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Inmunoprecipitación , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Reacción en Cadena en Tiempo Real de la Polimerasa , Líquido Sinovial/citología , Factor 6 Asociado a Receptor de TNF/farmacología
14.
Clin Immunol ; 173: 109-116, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27664932

RESUMEN

MicroRNA 182 has been found to have a distinct contribution in the clonal expansion of activated- and functioning of specialized-helper T cells. In this study we knocked down microRNA 182 in vivo and induced experimental autoimmune encephalomyelitis (EAE) to determine the influences of microRNA 182 in the Treg cells functional specialization through Foxo1 dependent pathway in the peripheral lymphoid organs. Down-regulation of microRNA 182 significantly increased the proportions of Foxp3+ T cells in the peripheral lymph nodes and spleen. In vivo study verified a positive correlation between microRNA 182 levels and symptom severity of EAE, and a negative correlation between microRNA 182 and the transcriptional factor Foxp3. In vitro polarization study also confirmed the contribution of Foxo1 in microRNA 182 mediated down-regulation of Foxp3+ T cells. Together, our results provide evidence that during the development of EAE, microRNA 182 repressed Treg cells differentiation through the Foxo1 dependent pathway.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Proteína Forkhead Box O1/inmunología , MicroARNs/inmunología , Linfocitos T Reguladores/inmunología , Animales , Diferenciación Celular , Femenino , Ganglios Linfáticos/citología , Ratones Endogámicos C57BL , Bazo/citología , Linfocitos T Reguladores/fisiología
15.
Dig Dis Sci ; 60(5): 1325-34, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25445161

RESUMEN

BACKGROUND: Cirrhosis, or liver fibrosis, which is mainly triggered by cirrhosis fat-storing cells (CFSCs) activation, has traditionally been considered an irreversible disease. However, recent observations indicate that even advanced fibrosis is still reversible by removing the causative agents. Anti-fibrotic effects of bone marrow-derived stromal cells (BMSCs) have been demonstrated by inhibiting CFSCs via cytokines secretion; however, the mechanisms are still unclear. AIMS: The purpose of this study was to explore the underlying mechanisms by which BMSCs modulate the function of activated CFSCs. METHODS: After the co-culture of CFSCs with BMSCs supernatants with or without the addition of recombinant rat adrenomedullin (AM)/AM-specific siRNA, western blot analysis was mainly used to detect the differences of relative protein expression on CFSCs. RESULTS: BMSC-secreted adrenomedullin (AM) effectively inhibited the proliferation and activation of CFSCs by suppressing the expression of Ang II and its binding receptor, AT1, which resulted in a reduction of p47-phox formation. CONCLUSIONS: Our data suggested that BMSCs inhibited CFSC activation in vitro via the AM-Ang II-p47-phox signaling pathway, and since CFSC activation is an essential part of hepatic fibrosis process, this inhibition by BMSCs implies us new insights into the potential treatment of hepatic fibrosis via BMSCs.


Asunto(s)
Adrenomedulina/metabolismo , Células Estrelladas Hepáticas/metabolismo , Metabolismo de los Lípidos , Cirrosis Hepática/metabolismo , Hígado/metabolismo , Células Madre Mesenquimatosas/metabolismo , Comunicación Paracrina , Actinas/metabolismo , Adrenomedulina/genética , Angiotensina II/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Colágeno Tipo I/metabolismo , Hígado/patología , Hígado/fisiopatología , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Cirrosis Hepática/fisiopatología , NADPH Oxidasas/metabolismo , Interferencia de ARN , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Transfección
16.
Proc Natl Acad Sci U S A ; 109(3): 899-904, 2012 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-22219356

RESUMEN

The type III histone deacetylase sirtuin 1 (Sirt1) is a suppressor of both innate and adoptive immune responses. We have recently found that Sirt1 expression is highly induced in anergic T cells. However, the transcriptional program to regulate Sirt1 expression in T cells remains uncharacterized. Here we report that the early responsive genes 2 and 3, which can be up-regulated by T-cell receptor-mediated activation of nuclear factor of activated T-cell transcription factors and are involved in peripheral T-cell tolerance, bind to the sirt1 promoter to transcript sirt1 mRNA. In addition, the forkhead transcription factor, FoxO3a, interacts with early responsive genes 2/3 on the sirt1 promoter to synergistically regulate Sirt1 expression. Interestingly, IL-2, a cytokine that can reverse T-cell anergy, suppresses sirt1 transcription by sequestering FoxO3a to the cytoplasm through activating the PI3K-AKT pathway. Expression of the constitutively active form of FoxO3a blocks IL-2-mediated reversal of T-cell tolerance by retaining sirt1 expression. Our findings here provide a molecular explanation of IL-2-mediated reversion of T-cell anergy.


Asunto(s)
Tolerancia Inmunológica/inmunología , Interleucina-2/inmunología , Sirtuina 1/genética , Linfocitos T/inmunología , Animales , Secuencia de Bases , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Anergia Clonal/efectos de los fármacos , Anergia Clonal/genética , Anergia Clonal/inmunología , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Proteína 3 de la Respuesta de Crecimiento Precoz/metabolismo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/genética , Interleucina-2/farmacología , Ratones , Modelos Inmunológicos , Datos de Secuencia Molecular , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Sirtuina 1/metabolismo , Linfocitos T/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
17.
Immunology ; 143(3): 396-405, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24850614

RESUMEN

Interleukin-9 (IL-9) was initially thought to be a type 2 T helper (Th2)-associated cytokine involved in the regulation of autoimmune responses by affecting multiple cell types. However, it was recently shown that IL-9-producing CD4+ T cells represent a discrete subset of Th cells, designated Th9 cells. Although Th9 cells have been shown to be important in many diseases, their roles in myasthenia gravis (MG) are unclear. The aim of this study was to determine whether IL-9 and Th9 cells promote the progression of experimental autoimmune myasthenia gravis (EAMG). The results showed that the percentage of Th9 cells changed during the progression of EAMG, accompanied by an up-regulation of IL-9. Blocking IL-9 activity with antibodies against IL-9 inhibited EAMG-associated pathology in rats and reduced serum anti-acetylcholine receptor IgG levels. Neutralization of IL-9 altered the Th subset distribution in EAMG, reducing the number of Th1 cells and increasing the number of regulatory T cells. Administration of an anti-IL-9 antibody may represent an effective therapeutic strategy for MG-associated pathologies or other T-cell- or B-cell-mediated autoimmune diseases.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Inmunidad Humoral , Interleucina-9/antagonistas & inhibidores , Miastenia Gravis Autoinmune Experimental/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Interleucina-9/metabolismo , Ratas , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
18.
Clin Immunol ; 152(1-2): 90-100, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24650778

RESUMEN

In this study, the capacity for t-PA to affect T cell-brain microvascular endothelial cell adhesion by acting as a cytokine was investigated. Following the treatment of a brain-derived endothelial cell line, bEnd.3, with various concentrations of t-PA, adhesion and transwell migration assays were performed. In the presence of t-PA, enhanced adhesion of T cells to bEnd.3 cells was observed. Using western blot analysis, an increase in ICAM-1 expression was detected for both t-PA-treated bEnd.3 cells and bEnd.3 cells treated with a non-enzymatic form of t-PA. In contrast, when LRP1 was blocked using a specific antibody, upregulation of ICAM-1 was inhibited and cAMP-PKA signaling was affected. Furthermore, using an EAE mouse model, administration of t-PA was associated with an increase in ICAM-1 expression by brain endothelial cells. Taken together, these findings suggest that t-PA can induce ICAM-1 expression in brain microvascular endothelial cells, and this may promote the development of EAE.


Asunto(s)
Encéfalo/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Molécula 1 de Adhesión Intercelular/biosíntesis , Esclerosis Múltiple/inmunología , Activador de Tejido Plasminógeno/fisiología , Animales , Anticuerpos Monoclonales/inmunología , Encéfalo/irrigación sanguínea , Adhesión Celular/efectos de los fármacos , Línea Celular Transformada , Movimiento Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Células Endoteliales/inmunología , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Femenino , Molécula 1 de Adhesión Intercelular/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/patología , Receptores de LDL/inmunología , Transducción de Señal/inmunología , Linfocitos T/citología , Linfocitos T/metabolismo , Activador de Tejido Plasminógeno/administración & dosificación , Proteínas Supresoras de Tumor/inmunología
19.
J Neuroinflammation ; 11: 79, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24742325

RESUMEN

BACKGROUND: Stroke is accompanied by a distinguished inflammatory reaction that is initiated by the infiltration of immunocytes, expression of cytokines, and other inflammatory mediators. As natural killer cells (NK cells) are a type of cytotoxic lymphocyte critical to the innate immune system, we investigated the mechanism of NK cells-induced brain injuries after cerebral ischemia and the chemotactic effect of IP-10 simultaneously. METHODS: NK cells infiltration, interferon-gamma (IFN-γ) and IP-10 expression were detected by immunohistochemistry, immunofluorescence, PCR and flow cytometry in human and C57/BL6 wild type mouse ischemic brain tissues. The ischemia area was detected via 2,3,5-triphenyltetrazolium chloride staining. CXCR3 mean fluorescence intensity of isolated NK cells was measured by flow cytometry. The neuronal injury made by NK cells was examined via apoptosis experiment. The chemotactic of IP-10 was detected by migration and permeability assays. RESULTS: In human ischemic brain tissue, infiltrations of NK cells were observed and reached a peak at 2 to 5 days. In a permanent middle cerebral artery occlusion (pMCAO) model, infiltration of NK cells into the ischemic infarct region reached their highest levels 12 hours after ischemia. IFN-γ-positive NK cells and levels of the chemokine IP-10 were also detected within the ischemic region, from 6 hours up to 4 days after pMCAO was performed, and IFN-γ levels decreased after NK cells depletion in vivo. Co-culture experiments of neural cells with NK cells also showed that neural necrosis was induced via IFN-γ. In parallel experiments with IP-10, the presence of CXCR3 indicates that NK cells were affected by IP-10 via CXCR3, and the effect was dose-dependent. After IP-10 depletion in vivo, NK cells decreased. In migration assays and permeability experiments, disintegration of the blood-brain barrier (BBB) was observed following the addition of NK cells. Moreover, in the presence of IP-10 this injury was aggravated. CONCLUSIONS: All findings support the hypothesis that NK cells participate in cerebral ischemia and promote neural cells necrosis via IFN-γ. Moreover, IP-10 intensifies injury to the BBB by NK cells via CXCR3.


Asunto(s)
Isquemia Encefálica/patología , Encéfalo/patología , Quimiocina CXCL10/metabolismo , Células Asesinas Naturales/fisiología , Animales , Animales Recién Nacidos , Antígenos Ly/metabolismo , Apoptosis/fisiología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/citología , Movimiento Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Infarto de la Arteria Cerebral Media/patología , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Receptores CXCR3/metabolismo
20.
Cell Immunol ; 290(2): 217-25, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25086239

RESUMEN

Extracellular adenosine is an essential negative regulator of immune reactions that acts by signaling via 4 distinct adenosine receptors. We evaluated adenosine receptor expression in Lewis rats presenting with experimental autoimmune myasthenia gravis (EAMG) to determine whether the expression of adenosine receptors are changed in the development and progression of EAMG. Lymphocyte A1AR and A2AAR mRNA and protein levels from lymphocytes harvested from the lymph nodes, spleen, and peripheral blood mononuclear cells (PBMCs) of EAMG rats were decreased. A modest but not significant increase in A2BAR levels was observed in EAMG lymphocytes harvested from lymph nodes and PBMCs. No changes in A3AR expression were observed in lymphocytes harvested from lymph nodes, spleen, or PBMCs following EAMG induction. Results presented in this report showed that the expression levels and the distribution pattern of adenosine receptors were altered in EAMG lymphocytes.


Asunto(s)
Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/metabolismo , Receptores Purinérgicos P1/biosíntesis , Animales , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Ratas , Ratas Endogámicas Lew , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Purinérgicos P1/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA