Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Int J Mol Sci ; 23(13)2022 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-35806005

RESUMO

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.


Assuntos
Esclerose Lateral Amiotrófica , Interleucina-12 , Traumatismos dos Nervos Periféricos , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/terapia , Animais , Humanos , Interleucina-12/metabolismo , Camundongos , Regeneração Nervosa/fisiologia , Oligodendroglia , Traumatismos dos Nervos Periféricos/metabolismo , Traumatismos dos Nervos Periféricos/patologia , Traumatismos dos Nervos Periféricos/terapia , Nervo Isquiático/lesões
2.
Cells ; 10(10)2021 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-34685754

RESUMO

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.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Neurônios Motores/patologia , Mutação/genética , Fibras Nervosas/patologia , Superóxido Dismutase-1/genética , Animais , Axônios/efeitos dos fármacos , Axônios/patologia , Álcoois Benzílicos/farmacologia , Cálcio/metabolismo , Diferenciação Celular , Glucosídeos/farmacologia , Ácido Glutâmico/farmacologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos Transgênicos , Degeneração Neural , Neuritos/patologia , Análise de Sobrevida
3.
Cells ; 11(1)2021 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-35011683

RESUMO

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.


Assuntos
Fator 1 de Crescimento de Fibroblastos/metabolismo , Integrases/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Camundongos , Camundongos Transgênicos , Ratos
4.
Medicine (Baltimore) ; 99(31): e21423, 2020 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-32756145

RESUMO

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.


Assuntos
Transtorno Bipolar/psicologia , Periodontite/epidemiologia , Adulto , Idoso , Transtorno Bipolar/complicações , Estudos de Coortes , Comorbidade , Bases de Dados Factuais , Feminino , Humanos , Revisão da Utilização de Seguros , Masculino , Pessoa de Meia-Idade , Periodontite/complicações , Taiwan/epidemiologia , Adulto Jovem
5.
Stem Cells Transl Med ; 9(4): 499-517, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31943851

RESUMO

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.


Assuntos
2',3'-Nucleotídeo Cíclico 3'-Fosfodiesterase/metabolismo , Lesões Encefálicas/terapia , Região CA1 Hipocampal/metabolismo , Cognição , Exossomos/enzimologia , Aprendizagem , Células-Tronco Mesenquimais/enzimologia , Neuritos/metabolismo , Neurogênese , Animais , Lesões Encefálicas/patologia , Cognição/efeitos dos fármacos , AMP Cíclico/metabolismo , Proteínas do Domínio Duplacortina , Exossomos/efeitos dos fármacos , Humanos , Isoindóis/farmacologia , Aprendizagem/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neuritos/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Neuropeptídeos/metabolismo , Polimerização , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo , Sulfonamidas/farmacologia , Tubulina (Proteína)/metabolismo
6.
Stem Cell Res ; 42: 101692, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31896484

RESUMO

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.


Assuntos
Conexinas/metabolismo , Perda Auditiva/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Linhagem Celular , Pré-Escolar , Conexina 26 , Humanos , Masculino , Taiwan
7.
Cancers (Basel) ; 11(11)2019 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-31731581

RESUMO

Nasopharyngeal carcinoma (NPC) is a unique malignancy derived from the epithelium of the nasopharynx. Despite great advances in the development of radiotherapy and chemotherapy, relapse and metastasis in NPC patients remain major causes of mortality. Evidence accumulated over recent years indicates that Epstein-Barr virus (EBV) lytic replication plays an important role in the pathogenesis of NPC and inhibition of EBV reactivation is now being considered as a goal for the therapy of EBV-associated cancers. With this in mind, a panel of dietary compounds was screened and emodin was found to have potential anti-EBV activity. Through Western blotting, immunofluorescence, and flow cytometric analysis, we show that emodin inhibits the expression of EBV lytic proteins and blocks virion production in EBV- positive epithelial cell lines. In investigating the underlying mechanism, reporter assays indicated that emodin represses Zta promoter (Zp) and Rta promoter (Rp) activities, triggered by various inducers. Mapping of the Zp construct reveals that the SP1 binding region is important for emodin-triggered repression and emodin is shown to be able to inhibit SP1 expression, suggesting that it likely inhibits EBV reactivation by suppression of SP1 expression. Moreover, we also show that emodin inhibits the tumorigenic properties induced by repeated EBV reactivation, including micronucleus formation, cell proliferation, migration, and matrigel invasiveness. Emodin administration also represses the tumor growth in mice which is induced by EBV activation. Taken together, our results provide a potential chemopreventive agent in restricting EBV reactivation and NPC recurrence.

8.
Sci Rep ; 9(1): 11143, 2019 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-31367001

RESUMO

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.


Assuntos
Fator 1 de Crescimento de Fibroblastos/genética , Genes Reporter/genética , Regiões Promotoras Genéticas/genética , Animais , Células CHO , Proliferação de Células/genética , Cricetulus , Proteínas de Fluorescência Verde/genética , Células Intersticiais do Testículo/fisiologia , Luciferases de Vaga-Lume/genética , Proteínas Luminescentes/genética , Masculino , Camundongos , Camundongos Transgênicos , Espermatogênese/genética , Timidina Quinase/genética
9.
Brain Res ; 1678: 397-406, 2018 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-29155003

RESUMO

Many studies have demonstrated that combining nerve conduits with neural stem cells or growth factors can repair peripheral nerve injury in rodents. However, nerve damage does occur with longer gaps in human than in rodents, thus findings from rodent studies are difficult to translate to clinical practice. Minipigs have a longer gap that is more closely applicable to the challenge of human nerve grafting in extensive traumatic nerve damage. In this study, human amniotic fluid stem cells (AFSCs) and polylactate nerve conduits were used to repair sciatic nerve injury in minipigs. The AFSCs exhibited the properties of mesenchymal stem cells with a propensity toward neural stem cells. Measurements of compound muscle action potential implied that administration of conduits with AFSCs was beneficial in function recovery in the minipig model compared with conduits alone. The results of diffusion tensor magnetic resonance imaging (DTI) based fiber tractography assay in the minipig model suggest that combining AFSCs with conduits could expedite the repair of sciatic nerve injury. Further, MR-based DTI provides an effective and non-invasive method to visualize the sciatic nerve and to monitor the regeneration progress of injured nerve in a longitudinal study.


Assuntos
Líquido Amniótico/citologia , Neuropatia Ciática/cirurgia , Transplante de Células-Tronco/métodos , Animais , Antígenos CD/metabolismo , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Potencial Evocado Motor/efeitos dos fármacos , Potencial Evocado Motor/fisiologia , Fator 1 de Crescimento de Fibroblastos/metabolismo , Citometria de Fluxo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Imageamento por Ressonância Magnética , Células-Tronco Mesenquimais/fisiologia , Músculo Esquelético/fisiopatologia , Regeneração Nervosa , Proteínas do Tecido Nervoso/metabolismo , RNA Mensageiro/metabolismo , Neuropatia Ciática/diagnóstico por imagem , Neuropatia Ciática/patologia , Células-Tronco , Suínos , Porco Miniatura
10.
Mol Ther Nucleic Acids ; 7: 439-452, 2017 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-28624220

RESUMO

Acid fibroblast growth factor (aFGF) has shown neuroprotection in Alzheimer's disease (AD) models in previous studies, yet its mechanism is still uncertain. Here we report that the efficacy of Tat-haFGF14-154 is markedly increased when loaded cationic liposomes for intranasal delivery are intranasally administered to APP/PS1 mice. Our results demonstrated that liposomal Tat-haFGF14-154 treatment significantly ameliorated behavioral deficits, relieved brain Aß burden, and increased the expression and activity of disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) in the brain. Tat-haFGF14-154 antagonized Aß1-42-induced cell death and structural damage in rat primary neurons in an ADAM10-dependent manner, which, in turn, was promoted by the activation of XBP1 splicing and modulated by the PI3K-CREB pathway. Both knockdown of ADAM10 and inhibition of PI3K (LY294002) negated Tat-haFGF14-154 rescue. Thus, Tat-haFGF14-154 activates the IRE1α/XBP1 pathway of the unfolded protein response (UPR) against the endoplasmic reticulum (ER) stress induced by Aß, and, subsequently, the nuclear translocation of spliced XBP1 (XBP1s) promotes transcription of ADAM10. These results highlight the important role of ADAM10 and its activation through the PI3K-CREB-IRE1α/XBP1 pathway as a key factor in the mechanism of neuroprotection for Tat-haFGF14-154.

11.
Int J Nanomedicine ; 12: 421-432, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28138239

RESUMO

Nerve repair in tissue engineering involves the precise construction of a scaffold to guide nerve cell regeneration in the desired direction. However, improvements are needed to facilitate the cell migration/growth rate of nerves in the center of a nerve conduit. In this paper, we propose a nerve guidance conduit with a hybrid structure comprising a microfibrous poly(lactic-co-glycolic acid) (PLGA) bundle wrapped in a micro/nanostructured PLGA membrane. We applied sequential fabrication processes, including photolithography, nano-electroforming, and polydimethylsiloxane casting to manufacture master molds for the repeated production of the PLGA subelements. After demolding it from the master molds, we rolled the microfibrous membrane into a bundle and then wrapped it in the micro/nanostructured membrane to form a nerve-guiding conduit. We used KT98/F1B-GFP cells to estimate the migration rate and guidance ability of the fabricated nerve conduit and found that both elements increased the migration rate 1.6-fold compared with a flat PLGA membrane. We also found that 90% of the cells in the hybrid nano/microstructured membrane grew in the direction of the designed patterns. After 3 days of culturing, the interior of the nerve conduit was filled with cells, and the microfiber bundle was also surrounded by cells. Our conduit cell culture results also demonstrate that the proposed micro/nanohybrid and microfibrous structures can retain their shapes. The proposed hybrid-structured conduit demonstrates a high capability for guiding nerve cells and promoting cell migration, and, as such, is feasible for use in clinical applications.


Assuntos
Regeneração Tecidual Guiada/métodos , Ácido Láctico/química , Membranas Artificiais , Nanoestruturas/química , Ácido Poliglicólico/química , Animais , Movimento Celular , Proliferação de Células , Células Cultivadas , Camundongos , Regeneração Nervosa/fisiologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Alicerces Teciduais
12.
Mol Cell Neurosci ; 79: 1-11, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27865767

RESUMO

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.


Assuntos
Interleucina-12/metabolismo , Regeneração Nervosa , Células-Tronco Neurais/transplante , Neurogênese , Traumatismos dos Nervos Periféricos/terapia , Células de Schwann/citologia , Nervo Isquiático/fisiologia , Animais , Células Cultivadas , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Fator de Transcrição STAT3/metabolismo , Transplante de Células-Tronco
13.
Exp Cell Res ; 344(2): 153-66, 2016 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-27138904

RESUMO

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.


Assuntos
Aurora Quinase A/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Glioblastoma/patologia , Células-Tronco Neoplásicas/enzimologia , Células-Tronco Neoplásicas/patologia , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Animais , Aurora Quinase A/antagonistas & inibidores , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Autorrenovação Celular/efeitos dos fármacos , Separação Celular , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Ativação Enzimática/efeitos dos fármacos , Fator 1 de Crescimento de Fibroblastos/genética , Fator 1 de Crescimento de Fibroblastos/farmacologia , Genes Reporter , Glioblastoma/enzimologia , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Células-Tronco Multipotentes/efeitos dos fármacos , Células-Tronco Multipotentes/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Piperazinas/farmacologia , Regiões Promotoras Genéticas/genética , Transdução de Sinais/efeitos dos fármacos , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo
14.
Methods Mol Biol ; 1516: 289-297, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27044047

RESUMO

Neurosphere assay is a common and robust method for identification of neural stem/progenitor cells, but obtaining large numbers of live single cells from dissociated neurospheres is difficult using nonenzymatic methods. Here, we present an enzyme-free method for high-efficiency neurosphere dissociation into single cells using microfluidic device technology. This method allows single cell dissociation of DC115 and KT98 cells with high cell viabilities (80-85 %), single-cell yield (91-95 %), and recovery (75-93 %).


Assuntos
Técnicas de Cultura de Células/métodos , Técnicas Analíticas Microfluídicas/métodos , Microfluídica/métodos , Células-Tronco Neurais/citologia , Animais , Diferenciação Celular/genética , Camundongos
15.
Mol Biol Cell ; 27(10): 1676-83, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27009199

RESUMO

RBM4 participates in cell differentiation by regulating tissue-specific alternative pre-mRNA splicing. RBM4 also has been implicated in neurogenesis in the mouse embryonic brain. Using mouse embryonal carcinoma P19 cells as a neural differentiation model, we observed a temporal correlation between RBM4 expression and a change in splicing isoforms of Numb, a cell-fate determination gene. Knockdown of RBM4 affected the inclusion/exclusion of exons 3 and 9 of Numb in P19 cells. RBM4-deficient embryonic mouse brain also exhibited aberrant splicing of Numb pre-mRNA. Using a splicing reporter minigene assay, we demonstrated that RBM4 promoted exon 3 inclusion and exon 9 exclusion. Moreover, we found that RBM4 depletion reduced the expression of the proneural gene Mash1, and such reduction was reversed by an RBM4-induced Numb isoform containing exon 3 but lacking exon 9. Accordingly, induction of ectopic RBM4 expression in neuronal progenitor cells increased Mash1 expression and promoted cell differentiation. Finally, we found that RBM4 was also essential for neurite outgrowth from cortical neurons in vitro. Neurite outgrowth defects of RBM4-depleted neurons were rescued by RBM4-induced exon 9-lacking Numb isoforms. Therefore our findings indicate that RBM4 modulates exon selection of Numb to generate isoforms that promote neuronal cell differentiation and neurite outgrowth.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Processamento Alternativo , Animais , Diferenciação Celular/genética , Éxons , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Neuritos/metabolismo , Neurogênese , Crescimento Neuronal , Neurônios/metabolismo , Cultura Primária de Células , Isoformas de Proteínas/metabolismo , Precursores de RNA/metabolismo , RNA Mensageiro/metabolismo , Células-Tronco/metabolismo
17.
Stem Cells Dev ; 24(24): 2853-63, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26414172

RESUMO

Heart disease is the leading cause of human death in the 21st century. Heart transplantation is a promising way to treat this. Because donor resources are limited, cell-based therapy has been developed as an alternative. Therefore, genes that trigger cardiogenesis could have potential in the treatment of heart disease. Fibroblast growth factor 1 (FGF1) is reported to stimulate cardiomyocyte proliferation under conditions of myocardial infarction, but little is known about its function during cardiac differentiation. In this study, we established an in vitro cardiogenesis model through a reliable chemical induction protocol to determine whether FGF1 and its gene expression are involved in cardiogenesis. Oxytocin, not only a well-known hormone but also a cardiac differentiation inducer, was used in a mouse embryonic stem cell line, E14Tg2a, to achieve cardiac differentiation. After differentiation, beating cell clusters appeared and the expression of FGF1B mRNA was upregulated in the late differentiation stage (differentiation days 8-14). Interestingly, FGF1B expression patterns during cardiac differentiation were similar to those of a mature cardiomyocyte marker, troponin T2, cardiac. The blockage of FGF1-FGF receptor (FGFR) signaling reduced not only the appearance of beating cluster formation but also the expression levels of cardiomyocyte-associated genes. Moreover, by investigating FGF1 downstream signaling cascades, we observed that the efficiency of beating cluster formation was mainly regulated through the FGF1-FGFR-PKC signaling axis. Taken together, we provide evidence to support that FGF1 could regulate cardiogenesis primarily through the protein kinase C signaling, but not through the mitogen-activated protein kinase signaling, pathway.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Fator 1 de Crescimento de Fibroblastos/metabolismo , Miócitos Cardíacos/citologia , Regiões Promotoras Genéticas , Proteína Quinase C/metabolismo , Sistemas do Segundo Mensageiro , Animais , Linhagem Celular , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Fator 1 de Crescimento de Fibroblastos/genética , Sistema de Sinalização das MAP Quinases , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ocitocina/farmacologia , Proteína Quinase C/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Troponina T/genética , Troponina T/metabolismo
18.
Lab Chip ; 15(14): 2928-38, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26060987

RESUMO

In vitro culture of single cells facilitates biological studies by deconvoluting complications from cell population heterogeneity. However, there is still a lack of simple yet high-throughput methods to perform single cell culture experiments. In this paper, we report the development and application of a microfluidic device with a dual-well (DW) design concept for high-yield single-cell loading (~77%) in large microwells (285 and 485 µm in diameter) which allowed for cell spreading, proliferation and differentiation. The increased single-cell loading yield is achieved by using sets of small microwells termed "capture-wells" and big microwells termed "culture-wells" according to their utilities for single-cell capture and culture, respectively. This novel device architecture allows the size of the "culture" microwells to be flexibly adjusted without affecting the single-cell loading efficiency making it useful for cell culture applications as demonstrated by our experiments of KT98 mouse neural stem cell differentiation, A549 and MDA-MB-435 cancer cell proliferation, and single-cell colony formation assay with A549 cells in this paper.


Assuntos
Ensaios de Triagem em Larga Escala , Técnicas Analíticas Microfluídicas , Células-Tronco Neurais/citologia , Análise de Célula Única , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Ensaios de Triagem em Larga Escala/instrumentação , Humanos , Camundongos , Técnicas Analíticas Microfluídicas/instrumentação , Análise de Célula Única/instrumentação
19.
Adv Mater ; 27(23): 3518-24, 2015 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-25953204

RESUMO

An injectable, self-healing hydrogel (≈1.5 kPa) is developed for healing nerve-system deficits. Neurosphere-like progenitors proliferate in the hydrogel and differentiate into neuron-like cells. In the zebrafish injury model, the central nervous system function is partially rescued by injection of the hydrogel and significantly rescued by injection of the neurosphere-laden hydrogel. The self-healing hydrogel may thus potentially repair the central nervous system.


Assuntos
Materiais Biocompatíveis/farmacologia , Sistema Nervoso Central/efeitos dos fármacos , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacologia , Fenômenos Mecânicos , Alginatos/química , Animais , Materiais Biocompatíveis/química , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sistema Nervoso Central/citologia , Sistema Nervoso Central/lesões , Quitosana/química , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Injeções , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Peixe-Zebra
20.
Dev Neurobiol ; 75(3): 232-48, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25104610

RESUMO

FGF1 is involved in multiple biological functions and exhibits the importance in neuroprotective effects. Our previous studies indicated that, in human brain and retina, the FGF1B promoter controlled the expression of FGF1. However, the exact function and regulation of FGF1 in brain is still unclear. Here, we generated F1B-GFP transgenic mice that expressed the GFP reporter gene under the control of human FGF1B promoter (-540 to +31). Using the fresh brain sections of F1B-GFP transgenic mice, we found that the F1B-GFP cells expressed strong fluorescent signals in the ventricular system throughout the brain. The results of immunohistochemistry further showed that two distinct populations of F1B-GFP(+) cells existed in the brains of F1B-GFP transgenic mice. We demonstrated that one population of F1B-GFP(+) cells was ependymal cells, which distributed along the entire ventricles, and the second population of F1B-GFP(+) cells was neuronal cells that projected their long processes into multiple directions in specific areas of the brain. The double labeling of F1B-GFP(+) cells and tyrosine hydroxylase indicated that a subpopulation of F1B-GFP(+) -neuronal cells was dopaminergic neurons. Importantly, these F1B-GFP(+) /TH(+) cells were distributed in the main dopaminergic neuronal groups including hypothalamus, ventral tegmental area, and raphe nuclei. These results suggested that human FGF1B promoter was active in ependymal cells, neurons, and a portion of dopaminergic neurons. Thus, the F1B-GFP transgenic mice provide an animal model not only for studying FGF1 gene expression in vivo but also for understanding the role of FGF1 contribution in neurodegenerative disorders such as Parkinson's disease and Alzheimer's disease.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Epêndima/citologia , Fator 1 de Crescimento de Fibroblastos/genética , Proteínas de Fluorescência Verde/metabolismo , Regiões Promotoras Genéticas , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Ventrículos Cerebrais/citologia , Ventrículos Cerebrais/metabolismo , Neurônios Dopaminérgicos/citologia , Epêndima/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Camundongos Transgênicos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...