Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(13)2022 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-35806005

RESUMEN

Nerve injury of the central nervous system and the peripheral nervous system still poses a major challenge in modern clinics. Understanding the roles of neurotrophic factors and their molecular mechanisms on neuro-regeneration will not only benefit patients with neural damage but could potentially treat neurodegenerative disorders, such as amyotrophic lateral sclerosis. In this study, we showed that human IL12 p40-p40 homodimer (hIL12p80) within PLA and PLGA conduits improved sciatic nerve regeneration in mice. As such, the group of conduits with NSCs and hIL12p80 (CNI) showed the best recovery among the groups in the sciatic functional index (SFI), compound muscle action potential (CMAP), and Rotarod performance analyses. In addition, the CNI group had a faster recovery and outperformed the other groups in SFI and Rotarod performance tests beginning in the fourth week post-surgery. Immunohistochemistry showed that the CNI group increased the diameter of the newly regenerated nerve by two-fold (p < 0.01). In vitro studies showed that hIL12p80 stimulated differentiation of mouse NSCs to oligodendrocyte lineages through phosphorylation of Stat3 at Y705 and S727. Furthermore, implantation using PLGA conduits (C2.0 and C2.1) showed better recovery in the Rotarod test and CMAP than using PLA conduits in FVB mice. In B6 mice, the group with C2.1 + NSCs + hIL12p80 (C2.1NI) not only promoted sciatic functional recovery but also reduced the rate of experimental autotomy. These results suggested that hIL12p80, combined with NSCs, enhanced the functional recovery and accelerated the regeneration of damaged nerves in the sciatic nerve injury mice. Our findings could further shed light on IL12's application not only in damaged nerves but also in rectifying the oligodendrocytes' defects in neurodegenerative diseases, such as amyotrophic lateral sclerosis and multiple sclerosis.


Asunto(s)
Esclerosis Amiotrófica Lateral , Interleucina-12 , Traumatismos de los Nervios Periféricos , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/terapia , Animales , Humanos , Interleucina-12/metabolismo , Ratones , Regeneración Nerviosa/fisiología , Oligodendroglía , Traumatismos de los Nervios Periféricos/metabolismo , Traumatismos de los Nervios Periféricos/patología , Traumatismos de los Nervios Periféricos/terapia , Nervio Ciático/lesiones
2.
Mol Cell Neurosci ; 79: 1-11, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27865767

RESUMEN

Regeneration of injured peripheral nerves is a slow, complicated process that could be improved by implantation of neural stem cells (NSCs) or nerve conduit. Implantation of NSCs along with conduits promotes the regeneration of damaged nerve, likely because (i) conduit supports and guides axonal growth from one nerve stump to the other, while preventing fibrous tissue ingrowth and retaining neurotrophic factors; and (ii) implanted NSCs differentiate into Schwann cells and maintain a growth factor enriched microenvironment, which promotes nerve regeneration. In this study, we identified IL12p80 (homodimer of IL12p40) in the cell extracts of implanted nerve conduit combined with NSCs by using protein antibody array and Western blotting. Levels of IL12p80 in these conduits are 1.6-fold higher than those in conduits without NSCs. In the sciatic nerve injury mouse model, implantation of NSCs combined with nerve conduit and IL12p80 improves motor recovery and increases the diameter up to 4.5-fold, at the medial site of the regenerated nerve. In vitro study further revealed that IL12p80 stimulates the Schwann cell differentiation of mouse NSCs through the phosphorylation of signal transducer and activator of transcription 3 (Stat3). These results suggest that IL12p80 can trigger Schwann cell differentiation of mouse NSCs through Stat3 phosphorylation and enhance the functional recovery and the diameter of regenerated nerves in a mouse sciatic nerve injury model.


Asunto(s)
Interleucina-12/metabolismo , Regeneración Nerviosa , Células-Madre Neurales/trasplante , Neurogénesis , Traumatismos de los Nervios Periféricos/terapia , Células de Schwann/citología , Nervio Ciático/fisiología , Animales , Células Cultivadas , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Factor de Transcripción STAT3/metabolismo , Trasplante de Células Madre
3.
Exp Cell Res ; 344(2): 153-66, 2016 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-27138904

RESUMEN

UNLABELLED: Fibroblast growth factor 1 (FGF1) binds and activates FGF receptors, thereby regulating cell proliferation and neurogenesis. Human FGF1 gene 1B promoter (-540 to +31)-driven SV40 T antigen has been shown to result in tumorigenesis in the brains of transgenic mice. FGF1B promoter (-540 to +31)-driven green fluorescent protein (F1BGFP) has also been used in isolating neural stem cells (NSCs) with self-renewal and multipotency from developing and adult mouse brains. In this study, we provide six lines of evidence to demonstrate that FGF1/FGFR signaling is implicated in the expression of Aurora A (AurA) and the activation of its kinase domain (Thr288 phosphorylation) in the maintenance of glioblastoma (GBM) cells and NSCs. First, treatment of FGF1 increases AurA expression in human GBM cell lines. Second, using fluorescence-activated cell sorting, we observed that F1BGFP reporter facilitates the isolation of F1BGFP(+) GBM cells with higher expression levels of FGFR and AurA. Third, both FGFR inhibitor (SU5402) and AurA inhibitor (VX680) could down-regulate F1BGFP-dependent AurA activity. Fourth, inhibition of AurA activity by two different AurA inhibitors (VX680 and valproic acid) not only reduced neurosphere formation but also induced neuronal differentiation of F1BGFP(+) GBM cells. Fifth, flow cytometric analyses demonstrated that F1BGFP(+) GBM cells possessed different NSC cell surface markers. Finally, inhibition of AurA by VX680 reduced the neurosphere formation of different types of NSCs. Our results show that activation of AurA kinase through FGF1/FGFR signaling axis sustains the stem cell characteristics of GBM cells. IMPLICATIONS: This study identified a novel mechanism for the malignancy of GBM, which could be a potential therapeutic target for GBM.


Asunto(s)
Aurora Quinasa A/metabolismo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Glioblastoma/patología , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Animales , Aurora Quinasa A/antagonistas & inhibidores , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Autorrenovación de las Células/efectos de los fármacos , Separación Celular , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Activación Enzimática/efectos de los fármacos , Factor 1 de Crecimiento de Fibroblastos/genética , Factor 1 de Crecimiento de Fibroblastos/farmacología , Genes Reporteros , Glioblastoma/enzimología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/patología , Células Madre Neoplásicas/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Piperazinas/farmacología , Regiones Promotoras Genéticas/genética , Transducción de Señal/efectos de los fármacos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo
4.
Arch Toxicol ; 88(3): 769-80, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24318646

RESUMEN

Acrylamide is a neurological and reproductive toxicant in humans and laboratory animals; however, the neuron developmental toxicity of acrylamide remains unclear. The aims of this study are to investigate the cytotoxicity and neurite outgrowth inhibition of acrylamide in nerve growth factor (NGF)- or fibroblast growth factor 1 (FGF1)-mediated neural development of PC12 cells. MTS assay showed that acrylamide treatment suppresses NGF- or FGF1-induced PC12 cell proliferation in a time- and dose-dependent manner. Quantification of neurite outgrowth demonstrated that 0.5 mM acrylamide treatment resulted in significant decrease in differentiation of NGF- or FGF1-stimulated PC12 cells. This decrease is accompanied with the reduced expression of growth-associated protein-43, a neuronal marker. Moreover, relative levels of pERK, pAKT, pSTAT3 and pCREB were increased within 5-10 min when PC12 cells were treated with NGF or FGF1. Acrylamide (0.5 mM) decreases the NGF-induced activation of AKT-CREB but not ERK-STAT3 within 20 min. Similarly, acrylamide (0.5 mM) decreases the FGF1-induced activation of AKT-CREB within 20 min. In contrast to the NGF treatment, the ERK-STAT3 activation that was induced by FGF1 was slightly reduced by 0.5 mM acrylamide. We further showed that PI3K inhibitor (LY294002), but not MEK inhibitor (U0126), could synergize with acrylamide (0.5 mM) to reduce the cell viability and neurite outgrowth in NGF- or FGF1-stimulated PC12 cells. Moreover, acrylamide (0.5 mM) increased reactive oxygen species (ROS) activities in NGF- or FGF1-stimulated PC12 cells. This increase was reversed by Trolox (an ROS scavenging agent) co-treatment. Together, our findings reveal that NGF- or FGF1-stimulation of the neuronal differentiation of PC12 cells is attenuated by acrylamide through the inhibition of PI3K-AKT-CREB signaling, along with the production of ROS.


Asunto(s)
Acrilamida/toxicidad , Factor 1 de Crecimiento de Fibroblastos/farmacología , Factor de Crecimiento Nervioso/farmacología , Células PC12/efectos de los fármacos , Animales , Butadienos/farmacología , Diferenciación Celular/efectos de los fármacos , Cromonas/farmacología , Morfolinas/farmacología , Neuritas/efectos de los fármacos , Nitrilos/farmacología , Células PC12/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
5.
J Neurochem ; 126(1): 4-18, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23647222

RESUMEN

Valproic acid (VPA) is the primary mood-stabilizing drug to exert neuroprotective effects and to treat bipolar disorder in clinic. Fibroblast growth factor 1 (FGF1) has been shown to regulate cell proliferation, cell division, and neurogenesis. Human FGF1 gene 1B promoter (-540 to +31)-driven green fluorescence (F1BGFP) has been shown to recapitulate endogenous FGF1 gene expression and facilitates the isolation of neural stem/progenitor cells (NSPCs) from developing and adult mouse brains. In this study, we provide several lines of evidence to demonstrate the underlying mechanisms of VPA in activating FGF-1B promoter activity: (i) VPA significantly increased the FGF-1B mRNA expression and the percentage of F1BGFP(+) cells; (ii) the increase of F1BGFP expression by VPA involves changes of regulatory factor X (RFX) 1-3 transcriptional complexes and the increase of histone H3 acetylation on the 18-bp cis-element of FGF-1B promoter; (iii) treatments of other histone deacetylases (HDAC) inhibitors, sodium butyrate and trichostatin A, significantly increased the expression levels of FGF-1B, RFX2, and RFX3 transcripts; (iv) treatments of glycogen synthase kinase 3 (GSK-3) inhibitor, lithium, or GSK-3 siRNAs also significantly activated FGF-1B promoter; (v) VPA specifically enhanced neuronal differentiation in F1BGFP(+) embryonic stem cells and NSPCs rather than GFP(-) cells. This study suggested, for the first time, that VPA activates human FGF1 gene promoter through inhibiting HDAC and GSK-3 activities.


Asunto(s)
Antimaníacos/farmacología , Factor 1 de Crecimiento de Fibroblastos/efectos de los fármacos , Factor 1 de Crecimiento de Fibroblastos/genética , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/metabolismo , Ácido Valproico/farmacología , Animales , Western Blotting , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Células Madre Embrionarias/efectos de los fármacos , Citometría de Flujo , Humanos , Inmunohistoquímica , Cloruro de Litio/farmacología , Ratones , Células-Madre Neurales/efectos de los fármacos , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética , ARN Interferente Pequeño/farmacología , Activación Transcripcional/efectos de los fármacos
6.
Anal Chem ; 85(24): 11920-8, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24228937

RESUMEN

Obtaining single dissociated cells from neurospheres is difficult using nonenzymatic methods. In this paper we report the development of a microfluidic-chip-based approach that utilizes flow and microstructures to dissociate neurospheres. We show that this microfluidic-chip-based neurosphere-dissociation method can generate high yields of single cells from dissociated neurospheres of mouse KT98 and DC115 cell models (passage number, 3-8; diameter range, 40-250 µm): 90% and 95%, respectively. The microfluidic-chip-dissociated cells had high viabilities (80-85%) and the ability to regrow into neurospheres, demonstrating the applicability of this device to neurosphere assay applications. In addition, the dissociated cells retained their normal differentiation potentials, as shown by their capabilities to differentiate into three neural lineages (neurons, astroglia, and oligodendrocytes) when cultured in differentiation culture conditions. Since this microfluidic-chip-based method does not require the use of enzymatic reagents, the risk of contamination from exogenous substances could be reduced, making it an attractive tool for a wide range of applications where neurosphere dissociation is needed.


Asunto(s)
Técnicas Analíticas Microfluídicas/métodos , Células-Madre Neurales/citología , Análisis de la Célula Individual/métodos , Animales , Diferenciación Celular , Línea Celular , Supervivencia Celular , Diseño de Equipo , Ratones , Técnicas Analíticas Microfluídicas/instrumentación , Análisis de la Célula Individual/instrumentación
7.
J Cell Biochem ; 113(7): 2511-22, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22415835

RESUMEN

Fibroblast growth factor 1 (FGF1) has been shown to regulate cell proliferation, cell division, and neurogenesis. Human FGF1 gene 1B promoter (-540 to +31)-driven green fluorescence (F1BGFP) was shown to recapitulate endogenous FGF1 gene expression. It can also be used to isolate neural stem/progenitor cells (NSPCs) and glioblastoma stem cells (GBM-SCs) from developing mouse brains and human glioblastoma tissues, respectively. However, the regulatory mechanisms of FGF-1B promoter and F1BGFP(+) cells are not clear. In this study, we present several lines of evidence to show the roles of ciliogenic RFX transcription factors in the regulation of FGF-1B gene promoter and F1BGFP(+) cells: (i) RFX1, RFX2, and RFX3 transcription factors could directly bind the 18-bp cis-element (-484 to -467), and contribute to the regulation of FGF1 promoter and neurosphere formation. (ii) We demonstrated RFX2/RFX3 complex could only be detected in the nuclear extract of FGF-1B positive cells, but not in FGF-1B negative cells. (iii) Protein kinase C inhibitors, staurosporine and rottlerin, could decrease the percentage of F1BGFP(+) cells and their neurosphere formation efficiency through reducing the RFX2/3 complex. (iv) RNA interference knockdown of RFX2 could significantly reduce the percentage of F1BGFP(+) cells and their neurosphere formation efficiency whereas overexpression of RFX2 resulted in the opposite effects. Taken together, this study suggests ciliogenic RFX transcription factors regulate FGF-1B promoter activity and the maintenance of F1BGFP(+) NSPCs and GBM-SCs.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Factor 1 de Crecimiento de Fibroblastos/genética , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Acetofenonas/farmacología , Benzopiranos/farmacología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/genética , Genes Reporteros , Glioblastoma/metabolismo , Proteínas Fluorescentes Verdes/genética , Humanos , Proteína Quinasa C/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño , Factores de Transcripción del Factor Regulador X , Factor Regulador X1 , Estaurosporina/farmacología , Factores de Transcripción/genética , Transcripción Genética
8.
Arch Biochem Biophys ; 2012 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-22683470

RESUMEN

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

9.
J Biol Chem ; 285(18): 13885-95, 2010 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-20189986

RESUMEN

Fibroblast growth factor 1 (FGF1) has been suggested to have an important role in cell growth, proliferation, and neurogenesis. Human FGF1 gene 1B promoter (-540 to +31)-driven green fluorescence (F1BGFP) has been shown to monitor endogenous FGF1 expression. F1BGFP could also be used to isolate neural stem/progenitor cells from embryonic, neonatal, and adult mouse brains or to isolate glioblastoma stem cells (GBM-SCs) from human glioblastoma tissues. Here, we present evidence that transcription factor RFX1 could bind the 18-bp cis-elements (-484 to -467) of the F1B promoter, modulate F1BGFP expression and endogenous FGF1 expression, and further regulate the maintenance of GBM-SCs. These observations were substantiated by using yeast one-hybrid assay, electrophoretic mobility shift assay, chromatin immunoprecipitation assay, gain- and loss-of-function assays, and neurosphere assays. Overexpression of RFX1 was shown to down-regulate FGF-1B mRNA expression and neurosphere formation in human glioblastoma cells, whereas RNA interference knockdown of RFX1 demonstrated the opposite effects. Our findings provide insight into FGF1 gene regulation and suggest that the roles of FGF1 and RFX1 in the maintenance of GBM-SCs. RFX1 may negatively regulate the self-renewal of GBM-SCs through modulating FGF-1B and FGF1 expression levels by binding the 18-bp cis-elements of the F1B promoter.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/fisiología , Factor 1 de Crecimiento de Fibroblastos/biosíntesis , Elementos de Respuesta/fisiología , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Factor 1 de Crecimiento de Fibroblastos/genética , Glioblastoma/genética , Glioblastoma/metabolismo , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Humanos , Ratones , Células Madre Neoplásicas/metabolismo , Interferencia de ARN , Factores de Transcripción del Factor Regulador X , Factor Regulador X1 , Factores de Transcripción/genética
10.
Artif Organs ; 35(4): 363-72, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21314831

RESUMEN

Neurotrophic factors such as the glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) promote nerve cell survival and regeneration, but their efficacy in repairing a longer gap defect of rat sciatic nerve (15 mm) has not been established. In this study, two recombinant mammalian vectors containing either rat GDNF gene or BDNF gene were constructed and each was transfected into neural stem cells (NSCs). It was found that the transfection of GDNF or BDNF gene into NSCs led to significantly enhanced expression of GDNF or BDNF mRNA. The amount of GDNF or BDNF protein secreted from the transfected NSCs showed a 3.3-fold or 2.5-fold increase than that from nontransfected NSCs, respectively. The regeneration capacity of rat sciatic nerve in a poly(D,L-lactide) conduit seeded with GDNF or BDNF-transfected NSCs was evaluated by the histology, functional gait, and electrophysiology after 8 weeks of implantation. It was observed that the degree of myelination and the size of regenerated tissue in the conduits seeded with GDNF- and BDNF-transfected NSCs were higher than those seeded with the nontransfected NSCs. Conduits seeded with GDNF-transfected NSCs had the greatest number of blood vessels. The functional recovery assessed by the functional gait and electrophysiology was significantly improved for conduits seeded with GDNF or BDNF-transfected NSCs. It was concluded that the genetically modified NSCs may have potential applications in promoting nerve regeneration and functional recovery.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Regeneración Nerviosa , Células-Madre Neurales/metabolismo , Nervio Ciático/fisiología , Transfección , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Células Cultivadas , Expresión Génica , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Masculino , Ratones , Células-Madre Neurales/citología , Plásmidos/genética , Porosidad , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Nervio Ciático/ultraestructura , Andamios del Tejido/química
11.
Cells ; 11(1)2021 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-35011683

RESUMEN

Fibroblast growth factor 1 (FGF1) regulates many biological and physiological processes. In mice, Fgf1 gene contains at least three upstream promoters and are alternatively spliced to the first protein coding exon, giving rise to different Fgf1 mRNA variants (1A, 1B and 1G). Among them, the Fgf1A transcript is predominantly expressed in the heart. FGF1 can induce cardiomyocyte regeneration and cardiogenesis in vitro and in vivo. Here, we generated a novel mouse line using the Fgf1A promoter (F1A) driving the expression of the inducible Cre recombinase (CreERT2). We firstly demonstrated that the highest mRNA expression of CreERT2 were detected in the heart specifically of F1A-CreERT2 mice, similar to that of Fgf1A mRNA. The F1A-CreERT2 mice were crossed with ROSA26 mice, and the F1 mice were analyzed. The LacZ-positive signals were detected exclusively in the heart after tamoxifen administration. The CreERT2-mediated recombination in the tissues is monitored through LacZ-positive signals, indicating the in situ localization of F1A-positive cells. Consistently, these F1A-positive cells with RFP-positive signals or LacZ-positive blue signals were co-localized with cardiomyocytes expressing cardiac troponin T, suggesting cardiomyocyte-specific activation of Fgf1A promoter. Our data suggested that the F1A-CreERT2 mouse line could be used for time-dependent and lineage tracing of Fgf1A-expressing cells in vivo.


Asunto(s)
Factor 1 de Crecimiento de Fibroblastos/metabolismo , Integrasas/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Ratones , Ratones Transgénicos , Ratas
12.
Cells ; 10(10)2021 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-34685754

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a progressive nervous system disease that causes motor neuron (MN) degeneration and results in patient death within a few years. To recapitulate the cytopathies of ALS patients' MNs, SOD1G85R mutant and corrected SOD1G85G isogenic-induced pluripotent stem cell (iPSC) lines were established. Two SOD1 mutant ALS (SOD1G85R and SOD1D90A), two SOD1 mutant corrected (SOD1G85G and SOD1D90D), and one sporadic ALS iPSC lines were directed toward MNs. After receiving ~90% purity for MNs, we first demonstrated that SOD1G85R mutant ALS MNs recapitulated ALS-specific nerve fiber aggregates, similar to SOD1D90A ALS MNs in a previous study. Moreover, we found that both SOD1 mutant MNs showed ALS-specific neurite degenerations and neurotransmitter-induced calcium hyperresponsiveness. In a small compound test using these MNs, we demonstrated that gastrodin, a major ingredient of Gastrodia elata, showed therapeutic effects that decreased nerve fiber cytopathies and reverse neurotransmitter-induced hyperresponsiveness. The therapeutic effects of gastrodin applied not only to SOD1 ALS MNs but also to sporadic ALS MNs and SOD1G93A ALS mice. Moreover, we found that coactivation of the GSK3ß and IGF-1 pathways was a mechanism involved in the therapeutic effects of gastrodin. Thus, the coordination of compounds that activate these two mechanisms could reduce nerve fiber cytopathies in SOD1 ALS MNs. Interestingly, the therapeutic role of GSK3ß activation on SOD1 ALS MNs in the present study was in contrast to the role previously reported in research using cell line- or transgenic animal-based models. In conclusion, we identified in vitro ALS-specific nerve fiber and neurofunctional markers in MNs, which will be useful for drug screening, and we used an iPSC-based model to reveal novel therapeutic mechanisms (including GSK3ß and IGF-1 activation) that may serve as potential targets for ALS therapy.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Neuronas Motoras/patología , Mutación/genética , Fibras Nerviosas/patología , Superóxido Dismutasa-1/genética , Animales , Axones/efectos de los fármacos , Axones/patología , Alcoholes Bencílicos/farmacología , Calcio/metabolismo , Diferenciación Celular , Glucósidos/farmacología , Ácido Glutámico/farmacología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Transgénicos , Degeneración Nerviosa , Neuritas/patología , Análisis de Supervivencia
13.
Chem Res Toxicol ; 23(9): 1449-58, 2010 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-20734998

RESUMEN

Acrylamide (ACR) has been recognized as a neurological and reproductive toxin in humans and laboratory animals. This study aimed to determine the effects of ACR-induced DNA damage on cell cycle regulation in human astrocytoma cell lines. Treatment of U-1240 MG cells with 2 mM ACR for 48 h resulted in a significant inhibition of cell proliferation as evaluated by Ki-67 protein expression and MTT assay. The analysis of DNA damage with the comet assay showed that treatment of the cells with 0.5, 1, and 2 mM ACR for 48 h caused significant increases in DNA damage by 3.5-, 4-, and 14-fold, respectively. Meanwhile, analysis of cell-cycle arrest with flow cytometry revealed that the ACR treatments resulted in significant increases in the G(0)/G(1)-arrested cells in a time- and dose-dependent manner. Expression of DNA damage-associated/checkpoint-related signaling molecules, including phosphorylated-p53 (pp53), p53, p21, p27, Cdk2, and cyclin D(1), in three human astrocytoma cell lines (U-1240 MG, U-251 MG, and U-87 MG) was also analyzed by immunoblotting. Treatment of the three cell lines with 2 mM ACR for 48 h caused marked increases in pp53 and Cdk2, as well as decreases in cyclin D(1) and p27. Moreover, increases in p53 and p21 were detected in both U-1240 and U-87 MG cells, whereas no marked change in p53 and a decrease in p21 were observed in U-251 MG cells. To address the involvement of ataxia telangiectasia mutated/ATM-Rad3-related (ATM/ATR) kinase in the signaling of ACR-induced G(0)/G(1) arrest, caffeine was used to block the ATM/ATR pathway in U-1240 MG cells. Caffeine significantly attenuated the ACR-induced G(0)/G(1) arrest as well as the expression of DNA damage-associated/checkpoint-related signaling molecules in a dose-dependent manner. This in vitro study clearly demonstrates the critical role of ATM/ATR in the signaling of ACR-induced cell-cycle arrest in astrocytoma cells.


Asunto(s)
Acrilamida/toxicidad , Astrocitos/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Daño del ADN , Acrilamida/química , Astrocitoma , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayo Cometa , Ciclina D1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas de Unión al ADN/metabolismo , Fase G1/efectos de los fármacos , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo
14.
J Biomed Sci ; 17: 33, 2010 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-20429889

RESUMEN

BACKGROUND: Neural induction is a complex process and the detailed mechanism of FGF-induced neurogenesis remains unclear. METHODS: By using a serum-free neural induction method, we showed that FGF1 dose-dependently promoted the induction of Sox1/N-cadherin/nestin triple positive cells, which represent primitive neuroblasts, from mouse embryonic stem (ES) cells. RESULTS: We demonstrated that FGF1, FGF2, and FGF4, but not FGF8b, enhanced this neurogenesis. Especially, FGF-enhanced neurogenesis is not mediated through the rescue of the apoptosis or the enhancement of the proliferation of Sox1+ cells. We further indicated that the inactivation of c-Jun N-terminal kinase-1 (JNK-1) and extracellular signal-related kinase-2 (ERK-2), but not p38 mitogen-activated protein kinase (MAPK), inhibited the neural formation through the inhibition of ES differentiation, but not through the formation of endomesodermal cells. CONCLUSIONS: These lines of evidence delineated the roles of FGF downstream signals in the early neural differentiation of ES cells.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Neurogénesis/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular , Medio de Cultivo Libre de Suero , Cartilla de ADN/genética , Células Madre Embrionarias/metabolismo , Factores de Crecimiento de Fibroblastos/fisiología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Neurogénesis/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Recombinantes/farmacología , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
15.
Mol Cell Neurosci ; 41(3): 348-63, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19409495

RESUMEN

Fibroblast growth factor 1 (FGF1) and FGF2 have been shown to maintain the proliferation, self-renewal and multipotent capacities of neural stem/progenitor cells (NSPCs) in vitro. FGF1 is unique for binding to all known FGF receptors. In this study, we investigated if exogenous EGF and FGF1 could be used in the isolation of NSPCs from embryonic mouse brains. We demonstrated that EGF/FGF1-responsive cells exhibited lower proliferation rate and neurosphere formation efficiency than EGF/FGF2-responsive NSPCs. However, EGF/FGF1-responsive mouse brain cells exhibited better neural differentiation capacities than EGF/FGF2-responsive NSPCs at E11.5. Using F1BGFP reporter, we further demonstrated that F1BGFP+ cells showed similar multipotent capacities to CD133+ NSPCs, and could be induced more efficiently toward neuronal differentiation. Our results suggested that EGF/FGF1-responsive cells from E11.5 mouse brains could self-renew and have better multipotency than EGF/FGF2-responsive NSPCs. Further, CD133+ and F1BGFP+ NSPCs may also represent different subsets of NSPCs during neural development and adult neurogenesis.


Asunto(s)
Células Madre Adultas/metabolismo , Encéfalo/citología , Células Madre Embrionarias/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Regiones Promotoras Genéticas , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Células Madre Adultas/citología , Animales , Encéfalo/embriología , Encéfalo/metabolismo , Diferenciación Celular , Separación Celular/métodos , Células Madre Embrionarias/citología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Proteínas Fluorescentes Verdes , Humanos , Ratones , Neuronas/citología , Neuronas/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Proteínas Recombinantes de Fusión/metabolismo , Regeneración
16.
Stem Cell Res ; 42: 101692, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31896484

RESUMEN

Hearing loss is the most common disorder in the sensory system. Mutations in GJB2 have been reported to be very common in sensorineural hearing loss patients. In this report, we generated an induced pluripotent stem cell (iPSC) line, MMCi001-A, from the peripheral blood mononuclear cells of a 4-year-old male hearing loss patient carrying GJB2 pV37I mutation by using the Sendai virus delivery system. The generated iPSCs were demonstrated to express pluripotent markers and be differentiated into three germ layers in vitro and in vivo. This GJB2-pV37I iPSCs is valuable for studying the pathogenic mechanisms and drug discovery of hearing loss.


Asunto(s)
Conexinas/metabolismo , Pérdida Auditiva/genética , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Línea Celular , Preescolar , Conexina 26 , Humanos , Masculino , Taiwán
17.
Medicine (Baltimore) ; 99(31): e21423, 2020 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-32756145

RESUMEN

Whether periodontitis is a risk factor for developing bipolar disorders (BD) has not been investigated. We aimed to determine whether periodontitis is associated with the subsequent development of BD and examine the risk factors for BD among patients with periodontitis.Using ambulatory and inpatient claims data from the National Health Insurance Research Database (NHIRD), we identified 12,337 patients who were aged at least 20 years and newly diagnosed with periodontitis between 2000 and 2004. The date of the first claim with a periodontitis diagnosis was set as the index date. For each patient with periodontitis, 4 subjects without a history of periodontitis were randomly selected from the NHIRD and frequency-matched with the patients with periodontitis according to sex, age (in 5-year bands), and index year.The periodontitis group had a mean age of 44.0 ±â€Š13.7 years and slight predominance of men (51.3%). Compared with the subjects without periodontitis, the patients with periodontitis had higher prevalence of diabetes mellitus, hyperlipidemia, hypertension, ischemic heart disease, stroke, head injury, major depressive disorder, chronic obstructive pulmonary disease (COPD), and asthma (P < .001). The incidence rate of BD was higher in the periodontitis group than in the non-periodontitis group (2.74 vs 1.46 per 1000 person-year), with an adjusted hazard ratio of 1.82 (95% confidence interval = 1.59-2.08) after adjustment for sex, age, and comorbidities.The patients with periodontitis exhibited a significantly higher risk of developing BD. Keep the better oral hygiene to reduce periodontitis might be a preventive strategy for BD.


Asunto(s)
Trastorno Bipolar/psicología , Periodontitis/epidemiología , Adulto , Anciano , Trastorno Bipolar/complicaciones , Estudios de Cohortes , Comorbilidad , Bases de Datos Factuales , Femenino , Humanos , Revisión de Utilización de Seguros , Masculino , Persona de Mediana Edad , Periodontitis/complicaciones , Taiwán/epidemiología , Adulto Joven
18.
Stem Cells Transl Med ; 9(4): 499-517, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31943851

RESUMEN

Mesenchymal stem cells (MSCs) have been used in clinical studies to treat neurological diseases and damage. However, implanted MSCs do not achieve their regenerative effects by differentiating into and replacing neural cells. Instead, MSC secretome components mediate the regenerative effects of MSCs. MSC-derived extracellular vesicles (EVs)/exosomes carry cargo responsible for rescuing brain damage. We previously showed that EP4 antagonist-induced MSC EVs/exosomes have enhanced regenerative potential to rescue hippocampal damage, compared with EVs/exosomes from untreated MSCs. Here we show that EP4 antagonist-induced MSC EVs/exosomes promote neurosphere formation in vitro and increase neurogenesis and neuritogenesis in damaged hippocampi; basal MSC EVs/exosomes do not contribute to these regenerative effects. 2',3'-Cyclic nucleotide 3'-phosphodiesterase (CNP) levels in EP4 antagonist-induced MSC EVs/exosomes are 20-fold higher than CNP levels in basal MSC EVs/exosomes. Decreasing elevated exosomal CNP levels in EP4 antagonist-induced MSC EVs/exosomes reduced the efficacy of these EVs/exosomes in promoting ß3-tubulin polymerization and in converting toxic 2',3'-cAMP into neuroprotective adenosine. CNP-depleted EP4 antagonist-induced MSC EVs/exosomes lost the ability to promote neurogenesis and neuritogenesis in damaged hippocampi. Systemic administration of EV/exosomes from EP4 -antagonist derived MSC EVs/exosomes repaired cognition, learning, and memory deficiencies in mice caused by hippocampal damage. In contrast, CNP-depleted EP4 antagonist-induced MSC EVs/exosomes failed to repair this damage. Exosomal CNP contributes to the ability of EP4 antagonist-elicited MSC EVs/exosomes to promote neurogenesis and neuritogenesis in damaged hippocampi and recovery of cognition, memory, and learning. This experimental approach should be generally applicable to identifying the role of EV/exosomal components in eliciting a variety of biological responses.


Asunto(s)
2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/metabolismo , Lesiones Encefálicas/terapia , Región CA1 Hipocampal/metabolismo , Cognición , Exosomas/enzimología , Aprendizaje , Células Madre Mesenquimatosas/enzimología , Neuritas/metabolismo , Neurogénesis , Animales , Lesiones Encefálicas/patología , Cognición/efectos de los fármacos , AMP Cíclico/metabolismo , Proteínas de Dominio Doblecortina , Exosomas/efectos de los fármacos , Humanos , Isoindoles/farmacología , Aprendizaje/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neuritas/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neuropéptidos/metabolismo , Polimerizacion , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Sulfonamidas/farmacología , Tubulina (Proteína)/metabolismo
19.
Artif Organs ; 33(1): 26-35, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19178438

RESUMEN

There is a strong need for nerve-tissue engineering using the guide conduit and Schwann cells or neural stem cells (NSCs) with regeneration potential for injured peripheral nerves. In this study, micropatterned poly(D,L-lactide) (PLA) conduits were fabricated by microlithography and solvent-casting. The PLA conduits were seeded with the novel green fluorescent protein (GFP)-positive adult mouse NSCs obtained using the patented method of one of the authors. About 85% of the seeded NSCs were successfully aligned on the micropatterned conduits within 72 h and expressed the genes related to the production of neurotrophic factors. Gene expressions for the neurotrophic factors, such as nerve growth factor and brain-derived neurotrophic factor were upregulated by the micropatterned conduits at 72 h. The micropatterned PLA conduits seeded with the aligned NSCs were used to bridge the 10-mm sciatic nerve gaps in rats and were found to facilitate nerve repair and functional recovery during a period of 6 weeks compared with the nonseeded group. This model can be used to study the role of adult NSCs in peripheral-nerve regeneration in the future.


Asunto(s)
Células Madre Adultas/fisiología , Regeneración Nerviosa , Neuropatía Ciática/terapia , Trasplante de Células Madre/métodos , Ingeniería de Tejidos , Animales , Proliferación Celular , Células Cultivadas , Estudios de Factibilidad , Expresión Génica , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Andamios del Tejido
20.
Sci Rep ; 9(1): 11143, 2019 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-31367001

RESUMEN

Transgenic mice harboring imaging reporters take full advantage of imaging technologies in studies using living mice. Here, we established a tri-fusion multimodal reporter gene containing fragments from firefly luciferase, enhanced green fluorescent protein, and herpes simplex virus type 1 thymidine kinase and generated tri-fusion reporter Tg mice. Fibroblast growth factor type 1 (FGF1), a multifunctional mitogen to a wide range of tissues, regulates proliferation of neural stem cells of the brain, where FGF1 expression is initiated through activation of the FGF1B (F1B) promoter. The reporter mouse under the control of the human F1B promoter enables visualization in vivo where F1B activity is elevated, including tissues not only in the brain but also in the nasopharynx, skull, spine, and testes, particularly in Leydig cells. Treating Tg mice with the alkylating agent busulfan, which is known to eradicate Leydig cells and disrupt spermatogenesis in mice, eliminated the reporter signals. Restoring Leydig cells recovered reporter expression, indicating that the reporter can be used as a surrogate marker for Leydig cells. The F1B tri-fusion reporter mouse model can be utilized in longitudinal monitoring of the health status of the male reproductive system, such as in studies exploring the toxicity of chemicals to spermatogenesis.


Asunto(s)
Factor 1 de Crecimiento de Fibroblastos/genética , Genes Reporteros/genética , Regiones Promotoras Genéticas/genética , Animales , Células CHO , Proliferación Celular/genética , Cricetulus , Proteínas Fluorescentes Verdes/genética , Células Intersticiales del Testículo/fisiología , Luciferasas de Luciérnaga/genética , Proteínas Luminiscentes/genética , Masculino , Ratones , Ratones Transgénicos , Espermatogénesis/genética , Timidina Quinasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA