Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gene ; 897: 148083, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38101709

RESUMEN

Light is the main source of energy for plant growth. Studies have shown that I. indigotica is a light-demanding plant and its yield and various active components are positively correlated with light intensity, but no studies of light intensity affecting energy metabolism in I. indigotica have been reported. Mitochondria are the main site of energy metabolism, and miRNAs are important factors in regulating gene expression, this experiment attempts to study the effects of different light intensities on energy metabolism from the perspective of mitochondria and miRNAs. The results show that the biomass、mitochondrial structural integrity and energy metabolism in I. indigotica were found to be positively correlated with light intensity. Small RNA and transcriptome sequencing identified 241 miRNAs and 36,372 mRNAs, and degradomic technology identified 72 miRNAs targeting 106 mRNAs, among which 12 pairs of miRNA-mRNAs were annotated on mitochondria. Combined with RT-qPCR validation, it was concluded that miR167a-5p positively regulates LETM1 and affects mitochondrial structure, miR400-5p and mIR169m-p3_1ss15CT negatively regulate GRXS15 and CMC4, respectively, affecting SDH and CCO activities, and miR395a-APS4 may affect the utilization of ATP and sulfate assimilation. In summary, the results of this study complement and enrich knowledge of light effects on mitochondria from the perspective of miRNA, while providing guidance for the cultivation of I. indigotica.


Asunto(s)
Isatis , MicroARNs , Isatis/genética , MicroARNs/genética , MicroARNs/metabolismo , Luz , Desarrollo de la Planta
2.
J Adv Res ; 2023 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-37380102

RESUMEN

INTRODUCTION: Ectopic expression of transcription factor-mediated in vivo neuronal reprogramming provides promising strategy to compensate for neuronal loss, while its further clinical application may be hindered by delivery and safety concerns. As a novel and attractive alternative, small molecules may offer a non-viral and non-integrative chemical approach for reprogramming cell fates. Recent definitive evidences have shown that small molecules can convert non-neuronal cells into neurons in vitro. However, whether small molecules alone can induce neuronal reprogramming in vivo remains largely unknown. OBJECTIVES: To identify chemical compounds that can induce in vivo neuronal reprogramming in the adult spinal cord. METHODS: Immunocytochemistry, immunohistochemistry, qRT-PCR and fate-mapping are performed to analyze the role of small molecules in reprogramming astrocytes into neuronal cells in vitro and in vivo. RESULTS: By screening, we identify a chemical cocktail with only two chemical compounds that can directly and rapidly reprogram cultured astrocytes into neuronal cells. Importantly, this chemical cocktail can also successfully trigger neuronal reprogramming in the injured adult spinal cord without introducing exogenous genetic factors. These chemically induced cells showed typical neuronal morphologies and neuron-specific marker expression and could become mature and survive for more than 12 months. Lineage tracing indicated that the chemical compound-converted neuronal cells mainly originated from post-injury spinal reactive astrocytes. CONCLUSION: Our proof-of-principle study demonstrates that in vivo glia-to-neuron conversion can be manipulated in a chemical compound-based manner. Albeit our current chemical cocktail has a lowreprogramming efficiency, it will bring in vivo cell fate reprogramming closer to clinical application in brain and spinal cord repair. Future studies should focus on further refining our chemical cocktail and reprogramming approach to boost the reprogramming efficiency.

3.
J Neuroinflammation ; 20(1): 16, 2023 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-36710351

RESUMEN

Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system (CNS). Anxiety and depression are the most common psychiatric comorbidities of MS, which seriously affect patients' quality of life, treatment compliance, and prognosis. However, current treatments for anxiety and depression in MS show low therapeutic efficacy and significant side effects. In the present study, we explored the therapeutic effects of a novel low-toxic anti-inflammatory drug, nanoparticulate magnesium hydride (MgH2), on mood disorders of MS. We observed that anxiety/depression-like behaviors in experimental autoimmune encephalomyelitis (EAE) mice were alleviated by MgH2 treatment. In addition, disease severity and inflammatory demyelination were also diminished. Furthermore, we confirmed the suppressive effect of MgH2 on depression in the acute restraint stress model. Mechanistically, MgH2 may play a therapeutic role by promoting microglial M2 polarization, inhibiting microglial M1 polarization, and reducing oxidative stress and mitochondrial damage. Therefore, nanoparticulate MgH2 may be a promising therapeutic drug for psychiatric comorbidities of MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Ratones , Animales , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple/tratamiento farmacológico , Microglía/fisiología , Depresión/tratamiento farmacológico , Depresión/etiología , Calidad de Vida , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Estrés Oxidativo , Ansiedad/tratamiento farmacológico , Ansiedad/etiología , Ratones Endogámicos C57BL
4.
Neuropathology ; 43(1): 72-83, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35789505

RESUMEN

Glioblastoma (GBM) is the most lethal primary tumor in the human brain and lacks favorable treatment options. Sex differences in the outcome of GBM are broadly acknowledged, but the underlying molecular mechanisms remain largely unknown. To identify the sex-dependent critical genes in the progression of GBM, raw data from several microarray datasets with the same array platform were downloaded from the Gene Expression Omnibus (GEO) database. These datasets included tumorous and normal tissue from patients with GBM and crucial sex features. Then, the differentially expressed genes (DEGs) in female and male tumors were identified via bioinformatics analysis, respectively. Functional signatures of the identified DEGs were further annotated by Gene Ontology (GO) and pathway enrichment analyses. Venn diagram and functional protein-protein interaction (PPI) network analyses were performed to screen out the sex-specific DEGs. Survival analysis of patients with differences in the expression level of selected genes was then carried out using the data from The Cancer Genome Atlas (TCGA). Here, we showed that ECT2, AURKA, TYMS, CDK1, NCAPH, CENPU, OIP5, KIF14, ASPM, FBXO5, SGOL2, CASC5, SHCBP1, FN1, LOX, IGFBP3, CSPG4, and CD44 were enriched in female tumor samples, whereas TNFSF13B, CXCL10, CXCL8, CXCR4, TLR2, CCL2, and FCGR2A were enriched in male tumor samples. Among these key genes, interestingly, ECT2 was associated with increased an survival rate for female patients, whileTNFSF13B could be regarded as a potential marker of poor prognosis in male patients. These results suggested that sex differences in patients may be attributed to the heterogeneous gene activity, which might influence the oncogenesis and the outcomes of GBM.


Asunto(s)
Glioblastoma , Transcriptoma , Humanos , Femenino , Masculino , Glioblastoma/patología , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Pronóstico , Proteínas Nucleares/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Adaptadoras de la Señalización Shc/genética , Proteínas Adaptadoras de la Señalización Shc/metabolismo
5.
Nucleic Acids Res ; 50(16): 9319-9338, 2022 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-36029179

RESUMEN

Topoisomerase IIA (TOP2a) has traditionally been known as an important nuclear enzyme that resolves entanglements and relieves torsional stress of DNA double strands. However, its function in genomic transcriptional regulation remains largely unknown, especially during adult neurogenesis. Here, we show that TOP2a is preferentially expressed in neurogenic niches in the brain of adult mice, such as the subventricular zone (SVZ). Conditional knockout of Top2a in adult neural stem cells (NSCs) of the SVZ significantly inhibits their self-renewal and proliferation, and ultimately reduces neurogenesis. To gain insight into the molecular mechanisms by which TOP2a regulates adult NSCs, we perform RNA-sequencing (RNA-Seq) plus chromatin immunoprecipitation sequencing (ChIP-Seq) and identify ubiquitin-specific protease 37 (Usp37) as a direct TOP2a target gene. Importantly, overexpression of Usp37 is sufficient to rescue the impaired self-renewal ability of adult NSCs caused by Top2a knockdown. Taken together, this proof-of-principle study illustrates a TOP2a/Usp37-mediated novel molecular mechanism in adult neurogenesis, which will significantly expand our understanding of the function of topoisomerase in the adult brain.


Asunto(s)
Células Madre Adultas , ADN-Topoisomerasas de Tipo II , Enzimas Desubicuitinizantes , Células-Madre Neurales , Neurogénesis , Animales , Ratones , Células Madre Adultas/metabolismo , Enzimas Desubicuitinizantes/genética , Enzimas Desubicuitinizantes/metabolismo , ADN-Topoisomerasas de Tipo II/metabolismo , Ventrículos Laterales/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Activación Transcripcional/genética
6.
Theranostics ; 12(10): 4548-4563, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35832093

RESUMEN

Background: Direct reprogramming of astrocytes into neurons opens up a new avenue for neuroregenerative medicine. However, the poor understanding of the molecular mechanisms underpinning the latent neurogenic program in astrocytes has largely restricted this strategy towards safe and effective clinical therapies. Methods: Immunocytochemistry, immunohistochemistry, western blotting, qRT-PCR, gene knockdown and fate-mapping are performed to analyze the role of NOTCH1 signaling in regulation of the latent neurogenic program in reactive astrocytes after spinal cord injury. Results: Western blotting analysis highlights that NOTCH1 is a key signaling mediating Ascl1- and Neurog2-driven astrocyte-to-neuron conversion. Inhibition of NOTCH1 signaling in cultured astrocytes by shRNA or DAPT (a NOTCH1 inhibitor) is sufficient to reprogram them into neurons by upregulating the expression of pro-neural transcription factors, including NeuroD1, NeuroD2, Pax6, Lmx1a and Lhx6. In the spinal cord of adult mouse, the expression of Notch1 is detected in resident astrocytes, which was significantly increased after spinal cord injury (SCI). Genetical knockdown of NOTCH1 signaling alone successfully triggers endogenous reactive astrocytes reprogramming into neurons in the injured adult spinal cord. Importantly, pharmacologically blocking NOTCH1 signaling with small molecule DAPT alone can also induce in situ astrocyte-to-neuron conversion after SCI. Conclusions: We identify NOTCH1 as a key common signaling pathway in reactive astrocyte that provides a barrier for cell fate conversion. This proof-of-principle study will significantly expand our molecular understanding of astroglial-lineage reprogramming and overcoming the NOTCH1 gatekeeper with small molecules may provide a transgene-free approach for in vivo chemical neuronal reprogramming with potential clinical application in neuroregeneration.


Asunto(s)
Astrocitos , Receptor Notch1 , Traumatismos de la Médula Espinal , Animales , Astrocitos/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/fisiología , Inhibidores de Agregación Plaquetaria , Receptor Notch1/metabolismo , Transducción de Señal/fisiología , Traumatismos de la Médula Espinal/metabolismo
7.
Adv Sci (Weinh) ; 9(24): e2105442, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35758549

RESUMEN

Blood-brain barrier (BBB) impairment is an early prevalent feature of multiple sclerosis (MS), and remains vital for MS progression. Microglial activation precedes BBB disruption and cellular infiltrates in the brain of MS patients. However, little is known about the function of microglia in BBB impairment. Here, microglia acts as an important modulator of BBB integrity in inflammatory demyelination. Microglial depletion profoundly ameliorates BBB impairment in experimental autoimmune encephalomyelitis (EAE). Specifically, miR-126a-5p in microglia is positively correlated with BBB integrity in four types of MS plaques. Mechanistically, microglial deletion of miR-126a-5p exacerbates BBB leakage and EAE severity. The protective effect of miR-126a-5p is mimicked and restored by specific inhibition of MMP9 in microglia. Importantly, Auranofin, an FDA-approved drug, is identified to protect BBB integrity and mitigate EAE progression via a microglial miR-126a-5p dependent mechanism. Taken together, microglia can be manipulated to protect BBB integrity and ameliorate inflammatory demyelination. Targeting microglia to regulate BBB permeability merits consideration in therapeutic interventions in MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental , MicroARNs , Esclerosis Múltiple , Animales , Barrera Hematoencefálica , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Humanos , Metaloproteinasa 9 de la Matriz/farmacología , Metaloproteinasa 9 de la Matriz/uso terapéutico , Ratones , MicroARNs/genética , Microglía
8.
Neurosci Bull ; 37(9): 1314-1324, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34091810

RESUMEN

The exacerbation of progressive multiple sclerosis (MS) is closely associated with obstruction of the differentiation of oligodendrocyte progenitor cells (OPCs). To discover novel therapeutic compounds for enhancing remyelination by endogenous OPCs, we screened for myelin basic protein expression using cultured rat OPCs and a library of small-molecule compounds. One of the most effective drugs was pinocembrin, which remarkably promoted OPC differentiation and maturation without affecting cell proliferation and survival. Based on these in vitro effects, we further assessed the therapeutic effects of pinocembrin in animal models of demyelinating diseases. We demonstrated that pinocembrin significantly ameliorated the progression of experimental autoimmune encephalomyelitis (EAE) and enhanced the repair of demyelination in lysolectin-induced lesions. Further studies indicated that pinocembrin increased the phosphorylation level of mammalian target of rapamycin (mTOR). Taken together, our results demonstrated that pinocembrin promotes OPC differentiation and remyelination through the phosphorylated mTOR pathway, and suggest a novel therapeutic prospect for this natural flavonoid product in treating demyelinating diseases.


Asunto(s)
Remielinización , Animales , Diferenciación Celular , Flavanonas , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Ratas , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
9.
Neurosci Bull ; 35(5): 802-814, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31028571

RESUMEN

The correct differentiation of oligodendrocyte precursor cells (OPCs) is essential for the myelination and remyelination processes in the central nervous system. Determining the regulatory mechanism is fundamental to the treatment of demyelinating diseases. By analyzing the RNA sequencing data of different neural cells, we found that cyclin-dependent kinase 18 (CDK18) is exclusively expressed in oligodendrocytes. In vivo studies showed that the expression level of CDK18 gradually increased along with myelin formation during development and in the remyelination phase in a lysophosphatidylcholine-induced demyelination model, and was distinctively highly expressed in oligodendrocytes. In vitro overexpression and interference experiments revealed that CDK18 directly promotes the differentiation of OPCs, without affecting their proliferation or apoptosis. Mechanistically, CDK18 activated the RAS/mitogen-activated protein kinase kinase 1/extracellular signal-regulated kinase pathway, thus promoting OPC differentiation. The results of the present study suggest that CDK18 is a promising cell-type specific target to treat demyelinating disease.


Asunto(s)
Diferenciación Celular/fisiología , Quinasas Ciclina-Dependientes/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Células Precursoras de Oligodendrocitos/metabolismo , Animales , Animales Recién Nacidos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Ratas , Ratas Sprague-Dawley
10.
Neurosci Bull ; 35(3): 434-446, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30684125

RESUMEN

The obstacle to successful remyelination in demyelinating diseases, such as multiple sclerosis, mainly lies in the inability of oligodendrocyte precursor cells (OPCs) to differentiate, since OPCs and oligodendrocyte-lineage cells that are unable to fully differentiate are found in the areas of demyelination. Thus, promoting the differentiation of OPCs is vital for the treatment of demyelinating diseases. Shikimic acid (SA) is mainly derived from star anise, and is reported to have anti-influenza, anti-oxidation, and anti-tumor effects. In the present study, we found that SA significantly promoted the differentiation of cultured rat OPCs without affecting their proliferation and apoptosis. In mice, SA exerted therapeutic effects on experimental autoimmune encephalomyelitis (EAE), such as alleviating clinical EAE scores, inhibiting inflammation, and reducing demyelination in the CNS. SA also promoted the differentiation of OPCs as well as their remyelination after lysolecithin-induced demyelination. Furthermore, we showed that the promotion effect of SA on OPC differentiation was associated with the up-regulation of phosphorylated mTOR. Taken together, our results demonstrated that SA could act as a potential drug candidate for the treatment of demyelinating diseases.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Remielinización/efectos de los fármacos , Ácido Shikímico/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Enfermedades Desmielinizantes/prevención & control , Encefalitis/prevención & control , Encefalomielitis Autoinmune Experimental/prevención & control , Femenino , Ratones Endogámicos C57BL , Proteína Básica de Mielina/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Ratas , Serina-Treonina Quinasas TOR/metabolismo
11.
Oncogene ; 38(9): 1544-1559, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30337686

RESUMEN

TROY is a component of the Nogo receptor complex and plays the key role in neuronal survival, migration, and differentiation. Here, we show the up-regulation of TROY in human glioma tissues and cells. Inhibition of TROY expression slowed glioma development in vivo and in vitro. Raf kinase inhibitor (RKIP) was found to interact with TROY. The physical interaction of TROY/RKIP was confirmed via co-immunoprecipitation (co-IP) assays. Furthermore, we found that the TROY/RKIP interaction was enhanced by fetal bovine serum (FBS) exposure, and TROY knockdown also led to down-regulation of NF-κB. Finally, disruption of the TROY/RKIP interaction using the TAT-TROY (234-371 aa) protein reduced the glioma development in xenografted mice. This suggests the TROY/RKIP interaction is a potential target for therapy of gliomas.


Asunto(s)
Carcinogénesis/genética , Glioma/genética , Proteínas de Unión a Fosfatidiletanolamina/genética , Receptores del Factor de Necrosis Tumoral/genética , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Glioma/patología , Humanos , Ratones , FN-kappa B/genética , Suero/química , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Neurosci Bull ; 34(2): 247-260, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29397565

RESUMEN

The differentiation and maturation of oligodendrocyte precursor cells (OPCs) is essential for myelination and remyelination in the CNS. The failure of OPCs to achieve terminal differentiation in demyelinating lesions often results in unsuccessful remyelination in a variety of human demyelinating diseases. However, the molecular mechanisms controlling OPC differentiation under pathological conditions remain largely unknown. Myt1L (myelin transcription factor 1-like), mainly expressed in neurons, has been associated with intellectual disability, schizophrenia, and depression. In the present study, we found that Myt1L was expressed in oligodendrocyte lineage cells during myelination and remyelination. The expression level of Myt1L in neuron/glia antigen 2-positive (NG2+) OPCs was significantly higher than that in mature CC1+ oligodendrocytes. In primary cultured OPCs, overexpression of Myt1L promoted, while knockdown inhibited OPC differentiation. Moreover, Myt1L was potently involved in promoting remyelination after lysolecithin-induced demyelination in vivo. ChIP assays showed that Myt1L bound to the promoter of Olig1 and transcriptionally regulated Olig1 expression. Taken together, our findings demonstrate that Myt1L is an essential regulator of OPC differentiation, thereby supporting Myt1L as a potential therapeutic target for demyelinating diseases.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas del Tejido Nervioso/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Remielinización/fisiología , Factores de Transcripción/metabolismo , Animales , Enfermedades Desmielinizantes/inducido químicamente , Lisofosfatidilcolinas/toxicidad , Ratones , Ratones Endogámicos C57BL , Células Precursoras de Oligodendrocitos/citología , Oligodendroglía/citología
13.
Cell Death Dis ; 9(2): 181, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29415995

RESUMEN

Spinal cord injury (SCI) is a devastating type of central nervous system (CNS) trauma with limited therapeutic treatments. The polarization of microglia into the M1 or M2 state has been documented to play important roles in the pathogenesis of SCI, although the complete repertoire of underlying factors has not been identified. Interestingly, the time point at which hematomyelia (intramedullary spinal cord hemorrhage) is alleviated coincides with a decrease in the number of M2 microglia. Here the function of Hemopexin (Hpx), a hematogenous glycoprotein, was examined in the crush model of SCI. Hpx levels were elevated at the lesion site during hematomyelia and were synchronously correlated with the level of the M2 marker Arginase-1 (Arg-1). Ablation of Hpx in vivo affected the polarization state of lipopolysaccharide (LPS)-stimulated microglia, as mirrored by a lower percentage of M2 microglia and a higher percentage of M1 microglia in the lesion site, which delayed the recovery and exacerbated the behavioral dysfunction after SCI. However, Hpx induced a rapid switch from the M1 to M2 phenotype in LPS-stimulated primary cultured microglia in a heme scavenging-independent manner. The supernant of Hpx-treated microglia ameliorated neuronal degeneration, alleviated demyelination, and promoted oligodendrocyte precursor cell (OPC) maturation. This modulatory effect of Hpx on microglia polarization was at least partially mediated by the LRP-1 receptor. Based on these results, Hpx is considered a novel modulator of the polarization of microglia during the pathogenesis of SCI and may play a crucial role in the recovery from SCI.


Asunto(s)
Arginasa/metabolismo , Hemopexina/metabolismo , Microglía/metabolismo , Traumatismos de la Médula Espinal/sangre , Animales , Polaridad Celular/efectos de los fármacos , Polaridad Celular/fisiología , Células Cultivadas , Hemopexina/farmacología , Lipopolisacáridos/farmacología , Ratones , Microglía/efectos de los fármacos , Microglía/patología , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Células Precursoras de Oligodendrocitos/patología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/patología
14.
J Neuroimmunol ; 313: 152-160, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29132838

RESUMEN

Multiple sclerosis (MS) is a chronic autoimmune demyelinating disease of the central nervous system. Currently, there is no drug available to cure this kind of disease. Diosgenin is a plant-derived steroid saponin. A previous study in our lab revealed that diosgenin can promote oligodendrocyte progenitor cell differentiation and accelerate remyelination. In the present study, we found that diosgenin dose-dependently alleviated the progression of experimental autoimmune encephalomyelitis with reduced central nervous system inflammation and demyelination. We also found that diosgenin treatment can significantly inhibit the activation of microglia and macrophages, suppress CD4+ T cell proliferation and hinder Th1/Th17 cell differentiation. Therefore, we suggested that diosgenin may be a potential therapeutic drug for inflammatory demyelinating diseases, such as MS.


Asunto(s)
Antiinflamatorios/uso terapéutico , Diosgenina/uso terapéutico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Animales , Antígenos CD/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Encefalitis/tratamiento farmacológico , Encefalitis/etiología , Encefalomielitis Autoinmune Experimental/complicaciones , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/patología , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Estadísticas no Paramétricas , Resultado del Tratamiento
15.
EMBO Rep ; 18(10): 1801-1816, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28808113

RESUMEN

The regulation of inflammation is pivotal for preventing the development or reoccurrence of multiple sclerosis (MS). A biased ratio of high-M1 versus low-M2 polarized microglia is a major pathological feature of MS Here, using microarray screening, we identify the long noncoding RNA (lncRNA) GAS5 as an epigenetic regulator of microglial polarization. Gain- and loss-of-function studies reveal that GAS5 suppresses microglial M2 polarization. Interference with GAS5 in transplanted microglia attenuates the progression of experimental autoimmune encephalomyelitis (EAE) and promotes remyelination in a lysolecithin-induced demyelination model. In agreement, higher levels of GAS5 are found in amoeboid-shaped microglia in MS patients. Further, functional studies demonstrate that GAS5 suppresses transcription of TRF4, a key factor controlling M2 macrophage polarization, by recruiting the polycomb repressive complex 2 (PRC2), thereby inhibiting M2 polarization. Thus, GAS5 may be a promising target for the treatment of demyelinating diseases.


Asunto(s)
Microglía/fisiología , Esclerosis Múltiple/fisiopatología , ARN Largo no Codificante/genética , Diferenciación Celular , Enfermedades Desmielinizantes/fisiopatología , Encefalomielitis Autoinmune Experimental , Epigénesis Genética , Regulación de la Expresión Génica , Humanos , Inflamación , Macrófagos , Esclerosis Múltiple/genética , ARN Largo no Codificante/metabolismo
16.
Neurosci Bull ; 33(6): 603-615, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28717866

RESUMEN

Multiple sclerosis (MS) is a classical inflammatory demyelinating disease of the central nervous system (CNS). Microglia are the main resident immune cells in the CNS and are closely associated with the pathogenesis of MS. In the present study, we found that miR-30a was highly expressed in jellyfish-like microglia in chronic active lesions of MS patients, as well as in the microglia of mice with experimental autoimmune encephalomyelitis (EAE) at the chronic phase. In vitro, the conditioned supernatant of mouse microglia overexpressing miR-30a promoted the apoptosis of oligodendrocyte precursor cells (OPCs), and inhibited OPC differentiation. In vivo, overexpressing miR-30a in transplanted microglia exacerbated the progression of EAE. Overexpression and knock-down experiments in primary cultured mouse microglia showed that miR-30a increased the expression of IL-1ß and iNOS, which are pro-inflammatory, while inhibiting the expression of Ym-1 and CD206. Mechanistically, miR-30a inhibited the expression of Ppargc1b, which is the co-activator of peroxisome proliferator-activated receptor gamma, resulting in pro-inflammatory effects. Our work shows that miR-30a is an important regulator of the inflammatory response in microglia, and may be a promising therapeutic target for inflammatory diseases like MS in the CNS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , MicroARNs/metabolismo , Microglía/metabolismo , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley
17.
J Neurosci ; 36(35): 9253-66, 2016 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-27581464

RESUMEN

UNLABELLED: T-helper 17 (Th17) cells play an important role in the pathogenesis of multiple sclerosis (MS), an autoimmune demyelinating disease that affects the CNS. In the present study, MicroRNA sequencing (miRNA-seq) was performed in mouse Th0 and Th17 cells to determine the critical miRNAs that are related to Th17 differentiation. We found that miR-30a was significantly downregulated during mouse Th17 differentiation. In addition, the level of miR-30a in CD4(+) T cells from peripheral blood of MS patients and experimental autoimmune encephalomyelitis (EAE) animal models was also decreased and inversely correlated with the expression of interleukin 17a, the canonical cytokine of Th17 cells. Moreover, overexpression of miR-30a inhibited Th17 differentiation and prevented the full development of EAE, whereas interference of miR-30a promoted Th17 differentiation. Mechanism studies showed that miR-30a reduced IRF4 expression by specifically binding with the 3'-untranslated region. Through screening of 640 different Food and Drug Administration (FDA)-approved drugs, we found that disulfiram and diphenhydramine hydrochloride were effective candidates for inhibiting Th17 differentiation and ameliorating EAE development through upregulating miR-30a. To our knowledge, the present work is not only the first miRNA-seq study focusing on Th17 differentiation, but also the first chemical screening for FDA-approved drugs that inhibit Th17 differentiation through regulating miRNA expression. SIGNIFICANCE STATEMENT: The present work is the first miRNA sequencing (miRNA-seq) study focusing on T-helper 17 (Th17) differentiation. By miRNA deep sequencing, we found that miR-30a was downregulated during Th17 differentiation. miR-30a was also decreased in CD4(+) T cells from multiple sclerosis patients and experimental autoimmune encephalomyelitis (EAE) mice. miR-30a reduced IRF4 expression by specific binding with the 3'-untranslated region and thus suppressed Th17 differentiation and prevented the full development of EAE. Interestingly, by performing a chemical screen with Food and Drug Administration-approved small-molecule drugs, we found that disulfiram and diphenhydramine upregulated miR-30a and suppressed Th17-associated autoimmune demyelination.


Asunto(s)
Difenhidramina/farmacología , Disulfiram/farmacología , Encefalomielitis Autoinmune Experimental/metabolismo , Interleucina-17/metabolismo , MicroARNs/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Estudios de Casos y Controles , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/prevención & control , Femenino , Células HEK293 , Humanos , Factores Reguladores del Interferón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Proteolipídica de la Mielina/toxicidad , Glicoproteína Mielina-Oligodendrócito/toxicidad , Fragmentos de Péptidos/toxicidad , Estadísticas no Paramétricas , Transfección
18.
J Neuroimmunol ; 294: 6-13, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27138092

RESUMEN

Multiple sclerosis (MS) is a chronic autoimmune demyelinating disease of the central nervous system (CNS). The high costs, inconvenient administration, and side effects of current Food and Drug Administration (FDA)-approved drugs often lead to poor adherence to the long-term treatment of MS. Molecular hydrogen (H2) has been reported to exhibit anti-oxidant, anti-apoptotic, anti-inflammatory, anti-allergy, and anti-cancer effects. In the present study, we explored the prophylactic and therapeutic effects of hydrogen-rich water (HRW) on the progress of experimental autoimmune encephalomyelitis (EAE), the animal model for MS. We found that prophylactic administration of both 0.36mM and 0.89mM HRW was able to delay EAE onset and reduce maximum clinical scores. Moreover, 0.89mM HRW also reduced disease severity, CNS infiltration, and demyelination when administered after the onset of disease. Furthermore, HRW treatment prevented infiltration of CD4(+) T lymphocytes into the CNS and inhibited Th17 cell development without affecting Th1 cell populations. Because HRW is non-toxic, inexpensive, easily administered, and can readily cross the blood-brain barrier, our experiments suggest that HRW may have great potential in the treatment of MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/complicaciones , Hidrógeno/uso terapéutico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/etiología , Recuperación de la Función/efectos de los fármacos , Análisis de Varianza , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Citometría de Flujo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Enfermedades del Sistema Nervioso/inmunología , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Estadísticas no Paramétricas , Agua
19.
Mol Med Rep ; 12(4): 5701-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26239757

RESUMEN

Excessive apoptosis and high expression levels of interleukin­1ß (IL­1ß) in disc cells have been reported to serve important roles in intervertebral disc degeneration (IVDD). Previous studies investigating mesenchymal stem cells (MSCs) have indicated potential for their use in the treatment of IVDD. However, the therapeutic potential and anti­apoptotic ability of MSCs remains to be fully elucidated. The present study aimed to establish an in vitro model for bone marrow­derived MSC (BMSC) therapy by investigating the anti­apoptotic effects, in addition to the migration of BMSCs to nucleus pulposus (NP) cells stimulated by IL­1ß. A co-culture system of BMSCs and NP cells was founded. Following inflammatory stimulation, the NP cells exhibited increased indexes for inflammation­induced degeneration. The degenerative and apoptotic indexes were significantly reduced when NP cells were co­cultured with BMSCs. Compared with the indirect co-culture group, the direct co-culture group exhibited an improved capacity for anti-apoptosis. In addition, IL­1ß­stimulated NP cells attracted and mediated the migration of BMSCs. Mitochondrial transfer from BMSCs to NP cells by tunneling nanotubes was also observed. In conclusion, the anti­apoptosis and the migration, in addition to mitochondrial transfer associated with BMSC treatments in IVDD, were investigated in vitro in the present study. The interaction between stimulated NP cells and BMSCs is likely involved in to simulating the in vivo process of stem cell­mediated repair.


Asunto(s)
Células de la Médula Ósea/metabolismo , Condrocitos/metabolismo , Degeneración del Disco Intervertebral/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Apoptosis , Transporte Biológico , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Caspasa 3/genética , Caspasa 3/metabolismo , Comunicación Celular , Movimiento Celular , Supervivencia Celular , Condrocitos/citología , Condrocitos/efectos de los fármacos , Técnicas de Cocultivo , Expresión Génica , Interleucina-1beta/farmacología , Degeneración del Disco Intervertebral/genética , Degeneración del Disco Intervertebral/patología , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley
20.
Purinergic Signal ; 11(3): 361-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26099359

RESUMEN

Recruitment of oligodendrocyte precursor cells (OPCs) to the lesions is the most important event for remyelination after central nervous system (CNS) injury or in demyelinating diseases. However, the underlying molecular mechanism is not fully understood. In the present study, we found high concentrations of ATP could increase the number of migrating OPCs in vitro, while after pretreatment with oxidized ATP (a P2X7 receptor antagonist), the promotive effect was attenuated. The promotive effect of 2'(3')-O-(4-benzoylbenzoyl) adenosine 5'-triphosphate (BzATP) (a P2X7 receptor agonist) was more potent than ATP. After incubation with BzATP, the activity of Fyn, one member of the Src family of kinases, was enhanced. Moreover, the interaction between P2X7 and Fyn was identified by co-immunoprecipitation. After blocking the activity of Fyn or down-regulating the expression of Fyn, the migration of OPCs induced by BzATP was inhibited. These data indicate that P2X7 receptors/Fyn may mediate ATP-induced OPC migration under pathological conditions.


Asunto(s)
Adenosina Trifosfato/farmacología , Movimiento Celular/fisiología , Oligodendroglía/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fyn/fisiología , Receptores Purinérgicos P2X7/fisiología , Células Madre/fisiología , Adenosina Trifosfato/análogos & derivados , Animales , Separación Celular , Humanos , Lentivirus/crecimiento & desarrollo , Proteínas Proto-Oncogénicas c-fyn/efectos de los fármacos , Ratas , Receptores Purinérgicos P2X7/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...