Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neural Plast ; 2022: 3995227, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36406589

RESUMO

Voltage-gated sodium channel beta 2 (Nav2.2 or Navß2, coded by SCN2B mRNA), a gene involved in maintaining normal physiological functions of the prefrontal cortex and hippocampus, might be associated with prefrontal cortex aging and memory decline. This study investigated the effects of Navß2 in amyloid-ß 1-42- (Aß1-42-) induced neural injury model and the potential underlying molecular mechanism. The results showed that Navß2 knockdown restored neuronal viability of Aß1-42-induced injury in neurons; increased the contents of brain-derived neurotrophic factor (BDNF), enzyme neprilysin (NEP) protein, and NEP enzyme activity; and effectively altered the proportions of the amyloid precursor protein (APP) metabolites including Aß42, sAPPα, and sAPPß, thus ameliorating cognitive dysfunction. This may be achieved through regulating NEP transcription and APP metabolism, accelerating Aß degradation, alleviating neuronal impairment, and regulating BDNF-related signal pathways to repair neuronal synaptic efficiency. This study provides novel evidence indicating that Navß2 plays crucial roles in the repair of neuronal injury induced by Aß1-42 both in vivo and in vitro.


Assuntos
Disfunção Cognitiva , Canais de Sódio Disparados por Voltagem , Humanos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Neurônios/metabolismo , Canais de Sódio Disparados por Voltagem/metabolismo , Neprilisina/genética , Neprilisina/metabolismo , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/metabolismo
2.
Medicine (Baltimore) ; 100(50): e28201, 2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34918679

RESUMO

RATIONALE: Respiratory distress syndrome (RDS) refers to the symptoms of progressive dyspnea and respiratory failure in newborns shortly after birth. The clinical and genetic characteristics of patients with neonatal RDS have not been extensively reported. PATIENT CONCERNS: A infant was in critical condition with repeated paroxysmal blood oxygen decline. Oxygen inhalation and noninvasive ventilator-assisted breathing relief were not effective. The etiology was unclear, and there was no family history of lung disease. Surface-active substance replacement therapy and positive pressure-assisted ventilation support were ineffective. DIAGNOSIS: The infant was clinically diagnosed with RDS. Genetic tests revealed a heterozygous missense mutation in the c.168 surfactant protein C (SFTPC) gene. INTERVENTIONS: Tracheal intubation was performed with invasive ventilator-assisted breathing, pulmonary surfactant was administered. Supportive treatment for liver protection and administration of a cardiotonic diuretic, vasodilator, human immunoglobulin (intravenous infusion), fresh frozen plasma, and suspended red blood cells were performed. OUTCOMES: The infant showed poor responses to respiratory and circulatory support, antibiotic treatment, and other treatment methods. The patient was discharged from hospital against the advice of us, cut off from us. The long-term prognosis of the patient after discharge remains unknown. LESSONS: SFTPC gene mutations may be an important risk factor for the development of common lung diseases. Because of the important roles of surfactant functions and metabolism, mutations in these genes can affect the production and function of pulmonary surfactant, leading to severe lung disease in term newborns.


Assuntos
Proteína C Associada a Surfactante Pulmonar/genética , Síndrome do Desconforto Respiratório do Recém-Nascido/diagnóstico , Humanos , Recém-Nascido , Pneumopatias/genética , Mutação de Sentido Incorreto , Oxigênio/uso terapêutico , Proteína C , Surfactantes Pulmonares/uso terapêutico , Síndrome do Desconforto Respiratório do Recém-Nascido/genética , Síndrome do Desconforto Respiratório do Recém-Nascido/terapia , Tensoativos
3.
Brain Res Bull ; 162: 218-230, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32579902

RESUMO

Neonatal hypoxic-ischemic encephalopathy (HIE) is major cause of neonatal death or long-term neurodevelopmental disabilities, which becomes a major practical problem currently in clinic. Whereas, its pathophysiology and underlying molecular mechanism is not clear. MicroRNAs are involved in the normal growth and development of neuronal cells. Herein, the objective of this research was to examine the roles of miR-410-3p in neurological deficits, neuronal injury and neuron apoptosis after hypoxic-ischemic and to explore its associated mechanisms. We established the hypoxic-ischemic brain damage (HIBD) model and oxygen glucose deprivation (OGD) model. Zea-longa score and TTC staining were used to detect the acute cerebral dysfunction after HIBD. QPCR verification exhibited notable downregulation of miR-410-3p expression at 24 h in rats after HIBD as well as that in PC12, SY5Y cells and primary cortical neurons post OGD. To further determine the function of miR-410-3p, lentivirus-mediated overexpression virus was applied in vivo and in vitro. Behavioral tests, including Morris water maze, open field test, Y maze test, neurological severity score and rotating rod test, were performed to evaluate long-term behavioral changes of rats at 1 month post HIBD. The results showed that the number of cells together with the axonal length were reduced post OGD. While the increase of cells number and the axonal length was measured after upregulating miR-410-3p. Meanwhile, miR-410-3p overexpression inhibited neuron apoptosis and enhanced neuronal survival. In addition, long-term motor and cognitive functions were remarkably recovered in HIBD rats with miR-410-3p overexpression. Together, miR-410-3p exerts a critical role in protecting neuronal growth as well as promoting motor and cognitive function recovery in neonatal rats subjected to HIBD. The current study therefore provides critical insights to develop the activator of miR-410-3p for the clinical treatment of HIBD in future clinic trial.


Assuntos
Córtex Cerebral/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , MicroRNAs/biossíntese , Neurônios/metabolismo , Animais , Animais Recém-Nascidos , Córtex Cerebral/patologia , Feminino , Expressão Gênica , Humanos , Hipóxia Encefálica , Hipóxia-Isquemia Encefálica/genética , Hipóxia-Isquemia Encefálica/patologia , Aprendizagem em Labirinto/fisiologia , MicroRNAs/genética , Neurônios/patologia , Células PC12 , Gravidez , Ratos , Ratos Sprague-Dawley
4.
Mol Med Rep ; 20(2): 2002-2011, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31257483

RESUMO

Voltage­gated sodium channel ß2 (Navß2), as an unconventional substrate of ß­site amyloid precursor protein cleaving enzyme 1, is involved in regulating the neuronal surface expression of sodium channels. A previous study demonstrated that knockdown of Navß2 protected neurons and induced spatial cognition improvement by partially reducing pathological amyloidogenic processing of amyloid precursor protein (APP) in aged APP/presenilin 1 (PS1) transgenic mice. The present study aimed to investigate whether Navß2 knockdown altered APP metabolism via regulation of the Aß­degrading enzyme neprilysin (NEP). APPswe/PS1ΔE9 mice (APP/PS1 transgenic mice with a C57BL/6J genetic background) carrying a Navß2­knockdown mutation (APP/PS1/Navß2­kd) or without Navß2 knockdown (APP/PS1) were used for cell culture and further analysis. The present results demonstrated that in APP/PS1 mouse­derived neurons, Navß2 knockdown partially reversed the reduction in pathological APP cleavage, and the recovery of neurite extension and neuron area. Additionally, Navß2 knockdown increased NEP activity and levels, and the levels of intracellular domain fragment binding to the NEP promoter. The present findings suggested that knockdown of Navß2 reversed the APP/PS1 mutation­induced deficiency in amyloid ß degradation by regulating NEP.


Assuntos
Neurônios/metabolismo , Neuroproteção/genética , Presenilina-1/genética , Subunidade beta-2 do Canal de Sódio Disparado por Voltagem/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Modelos Animais de Doenças , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Camundongos , Camundongos Transgênicos , Mutação , Neprilisina/genética , Neurônios/patologia , Regiões Promotoras Genéticas/genética
5.
Cell Transplant ; 28(9-10): 1197-1211, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31271053

RESUMO

Transplantation of neural stem cells (NSCs) is a potential strategy for the treatment of spinal cord transection (SCT). Here we investigated whether transplanted NSCs would improve motor function of rats with SCT and explored the underlying mechanism. First, the rats were divided into sham, SCT, and NSC groups. Rats in the SCT and NSC groups were all subjected to SCT in T10, and were administered with media and NSC transplantation into the lesion site, respectively. Immunohistochemistry was used to label Nestin-, TUNEL-, and NeuN-positive cells and reveal the expression and location of type I insulin-like growth factor receptor (IGF-1 R). Locomotor function of hind limbs was assessed by Basso, Beattie, Bresnahan (BBB) score and inclined plane test. The conduction velocity and amplitude of spinal nerve fibers were measured by electrophysiology and the anatomical changes were measured using magnetic resonance imaging. Moreover, expression of IGF-1 R was determined by real-time polymerase chain reaction and Western blotting. The results showed that NSCs could survive and differentiate into neurons in vitro and in vivo. SCT-induced deficits were reduced by NSC transplantation, including increase in NeuN-positive cells and decrease in apoptotic cells. Moreover, neurophysiological profiles indicated that the latent period was decreased and the peak-to-peak amplitude of spinal nerve fibers conduction was increased in transplanted rats, while morphological measures indicated that fractional anisotropy and the number of nerve fibers in the site of spinal cord injury were increased after NSC transplantation. In addition, mRNA and protein level of IGF-1 R were increased in the rostral segment in the NSC group, especially in neurons. Therefore, we concluded that NSC transplantation promotes motor function improvement of SCT, which might be associated with activated IGF-1 R, especially in the rostral site. All of the above suggests that this approach has potential for clinical treatment of spinal cord injury.


Assuntos
Regulação da Expressão Gênica , Locomoção , Regeneração Nervosa , Células-Tronco Neurais/metabolismo , Receptor IGF Tipo 1/biossíntese , Traumatismos da Medula Espinal , Animais , Células-Tronco Neurais/patologia , Células-Tronco Neurais/transplante , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/terapia
6.
Front Neurosci ; 13: 16, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30728764

RESUMO

Neuronal apoptosis is a major pathological hallmark of the neonatal hypoxic-ischemic brain damage (HIBD); however, the role of miR-7a-2-3p in the regulation of HIBD remains unknown. The purpose of this study was to explore the possible roles of miR-7a-2-3p in brain injury using a hypoxia-ischemia model in rats and oxygen-glucose deprivation (OGD) model in vitro. Firstly, we established the hypoxia-ischemia (HI) model and verified the model using Zea Longa scores and MRI in rats. Next, the changes of miR-7a-2-3p were screened in the ischemic cortex of neonatal rats by qRT-PCR at 12, 48, and 96 h after HIBD. We have found that the expression of miR-7a-2-3p in the HI rats decreased significantly, compared with the sham group (P < 0.01). Then, we established the OGD model in PC12 cells, SH-SY5Y cells and primary cortical neurons in vitro and qRT-PCR was used to confirm the changes of miR-7a-2-3p in these cells after the OGD. In order to determine the function of miR-7a-2-3p, PC12 cells, SH-SY5Y cells and rat primary cortical neurons were randomly divided into normal, OGD, mimic negative control (mimic-NC) and miR-7a-2-3p groups. Then, Tuj1+ (neuronal marker) staining, TUNEL assay (to detect apoptotic cells) and MTT assay (to investigate cell viability) were performed. We have found that the number of PC12 cells, SH-SY5Y cells and cortical neurons in the miR-7a-2-3p groups increased significantly (P < 0.01) in comparison to the OGD groups. The survival of cortical neurons in the miR-7a-2-3p group was improved markedly (P < 0.01), while the apoptosis of neurons in the miR-7a-2-3p group was significantly decreased (P < 0.01), compared with the normal group. Lastly, we investigated the target genes of miR-7a-2-3p by using the prediction databases (miRDB, TargetScan, miRWalk, and miRmap) and verified the target genes with qRT-PCR in the HI rats. Bioinformatics prediction showed that Vimentin (VIM), pleiomorphic adenoma gene 1(PLAG1), dual specificity phosphatase 10 (DUSP10), NAD(P)H dehydrogenase, quinone 1 (NQO1) and tumor necrosis factor receptor superfamily member 1B (TNFRSF1B) might be the targets of miR-7a-2-3p and the qRT-PCR confirmed that VIM increased in the HI rats (P < 0.01). In conclusion, miR-7a-2-3p plays a crucial role in the hypoxic-ischemic injury, and is associated with regulation of VIM.

7.
Int J Mol Med ; 43(2): 807-820, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30569108

RESUMO

The aim of the present study was to investigate the putative role and underlying mechanisms of insulin­like growth factor 1 (IGF­1) in mediating neuroplasticity in rats subjected to partial dorsal root ganglionectomies following electro­acupuncture (EA) treatment. The rats underwent bilateral removal of the L1­L4 and L6 dorsal root ganglia (DRG), sparing the L5 DRG, and were subsequently subjected to 28 days of EA treatment at two paired acupoints, zusanli (ST 36)­xuanzhong (GB 39) and futu (ST 32)­sanyinjiao (SP 6), as the EA Model group. Rats that received partial dorsal root ganglionectomies without EA treatment served as a control (Model group). Subsequently, herpes simplex virus (HSV)­IGF­1, HSV­small interfering (si) RNA­IGF­1 and the associated control vectors were injected into the L5 DRG of rats in the EA Model group. HSV­IGF­1 transfection enhanced EA­induced neuroplasticity, which manifested as partial recovery in locomotor function, remission hyperpathia, growth of DRG­derived spared fibers, increased expression of phosphorylated (p­) phosphatidylinositol 3­kinase (PI3K) and Akt, and increased pPI3K/PI3K and pAkt/Akt expression ratios. By contrast, HSV­siRNA­IGF­1 treatment attenuated these effects induced by HSV­IGF­1 transfection. The results additionally demonstrated that HSV­IGF­1 transfection augmented the outgrowth of neurites in cultured DRG neurons, and interference of the expression of IGF­1 retarded neurite outgrowth. Co­treatment with a PI3K inhibitor or Akt siRNA inhibited the aforementioned effects induced by the overexpression of IGF­1. In conclusion, the results of the present study demonstrated the crucial roles of IGF­1 in EA­induced neuroplasticity following adjacent dorsal root ganglionectomies in rats via the PI3K/Akt signaling pathway.


Assuntos
Eletroacupuntura , Gânglios Espinais , Neuroproteção , Animais , Células Cultivadas , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Gânglios Espinais/cirurgia , Ganglionectomia , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos/cirurgia , Ratos Sprague-Dawley , Simplexvirus , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA