Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Psychiatry ; 28(8): 3444-3458, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37500828

RESUMO

Adverse experiences in early life can shape neuronal structures and synaptic function in multiple brain regions, leading to deficits of distinct cognitive functions later in life. Focusing on the pyramidal cells of the prelimbic cortex (PrL), a main subregion of the medial prefrontal cortex, the impact of early-life adversity (ELA) was investigated in a well-established animal model generated by changing the rearing environment during postnatal days 2 to 9 (P2-P9), a sensitive developmental period. ELA has enduring detrimental impacts on the dendritic spines of PrL pyramidal cells, which is most apparent in a spatially circumscribed region. Specifically, ELA affects both thin and mushroom-type spines, and ELA-provoked loss of spines is observed on selective dendritic segments of PrL pyramidal cells in layers II-III and V-VI. Reduced postsynaptic puncta represented by postsynaptic density protein-95 (PSD-95), but not synaptophysin-labelled presynaptic puncta, in ELA mice supports the selective loss of spines in the PrL. Correlation analysis indicates that loss of spines and postsynaptic puncta in the PrL contributes to the poor spatial working memory of ELA mice, and thin spines may play a major role in working memory performance. To further understand whether loss of spines affects glutamatergic transmission, AMPA- and NMDA-receptor-mediated synaptic currents (EPSCs) were recorded in a group of Thy1-expressing PrL pyramidal cells. ELA mice exhibited a depressed glutamatergic transmission, which is accompanied with a decreased expression of GluR1 and NR1 subunits in the PrL. Finally, upregulating the activation of Thy1-expressing PrL pyramidal cells via excitatory DREADDs can efficiently improve the working memory performance of ELA mice in a T-maze-based task, indicating the potential of a chemogenetic approach in restoring ELA-provoked memory deficits.


Assuntos
Memória de Curto Prazo , Animais , Camundongos , Espinhas Dendríticas/fisiologia , Transtornos da Memória/metabolismo , Memória de Curto Prazo/fisiologia , Neurônios , Córtex Pré-Frontal/metabolismo , Células Piramidais/metabolismo , Estresse Psicológico
2.
Exp Gerontol ; 179: 112243, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37336370

RESUMO

The differentiation of human induced pluripotent stem cells (hiPSCs) into functional dopaminergic neural precursors is the basis of cell therapy for Parkinson's disease (PD). However, the use of small molecule inhibitors/activators in the differentiation of hiPSCs in vitro leads to cell death and low differentiation efficiency. Moreover, the mechanism of differentiation remains unclear. MiR-210-5p was increased during hiPSCs differentiation. Whether it promotes hiPSCs differentiation and transplantation needs further study. Here, we overexpressed miR-210-5p in hiPSCs to study its roles and mechanisms. We found that miR-210-5p promoted the differentiation of hiPSCs into dopaminergic neural precursors and reduced the expression of SMAD4 and SUFU meanwhile. Luciferase assays showed that miR-210-5p binded to SMAD4 and SUFU, which are key molecules in the key signals (TGF-ß and SHH) of hiPSCs differentiation. Furthermore, in the effect evaluation of cell transplantation into parkinsonian rats, the degree of behavioral recovery and the growth of transplanted cells in the group overexpressed miR-210-5p were similar to those in the positive group with all small molecule inhibitors/activators. Therefore, we conclude that miR-210-5p promotes the differentiation of hiPSCs into dopaminergic neural precursors by targeting SMAD4 and SUFU. In the therapeutic evaluation of cell transplantation, miR-210-5p can replace the use of corresponding small molecule inhibitors/activators to reduce cell death. This study provides an experimental basis and a new target for the miRNA-modified differentiation of hiPSCs and cell transplantation in clinical treatment of PD in the future.


Assuntos
Células-Tronco Pluripotentes Induzidas , MicroRNAs , Humanos , Ratos , Animais , Células-Tronco Pluripotentes Induzidas/metabolismo , Diferenciação Celular , MicroRNAs/genética , MicroRNAs/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Proteínas Repressoras/metabolismo
3.
Aging Dis ; 13(4): 1293-1310, 2022 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-35855335

RESUMO

Memory loss is the key symptom of Alzheimer's disease (AD). As successful drug treatments have not yet been identified, non-pharmaceutical interventions such as physical exercise and training have been employed to improve the memory function of people with dementia. We investigated the effect of prolonged physical running on hippocampal-dependent spatial memory and its underlying mechanisms using a well-established rodent model of AD. 3xTg-AD transgenic mice and non-transgenic mice were subjected to voluntary wheel running for 5 months (1 hour per day, 5 days per week), followed by spatial memory testing. After the behavioral testing, dendritic spines, synapses, and synaptic proteins as well as amyloid-beta (Aß) pathology were analyzed in the dorsal hippocampi. Running improved hippocampal-dependent spatial memory in 3xTg-AD mice. This running strategy prevented both thin and mushroom-type spines on CA1 pyramidal cells in 3xTg-AD mice, whereas the effects of running in non-transgenic mice were limited to thin spines. The enormous effects of running on spines were accompanied by an increased number of synapses and upregulated expression of synaptic proteins. Notably, running downregulated the processing of amyloid precursor protein, decreasing intracellular APP expression and extracellular Aß accumulation, and spatial memory performance correlated with levels of Aß peptides Aß1-40 and Aß1-42. These data suggest that prolonged running may improve memory in preclinical AD via slowing down the amyloid pathology and preventing the loss of synaptic contacts.

4.
Brain Struct Funct ; 226(2): 397-414, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33386419

RESUMO

The effects of early-life adversity (ELA) on dendritic differentiation of striatal neurons were investigated in the dorsal striatum including the dorsomedial striatum and dorsolateral striatum (DMS and DLS, respectively). An animal model of ELA was created by changing the growth environment of newborn mouse pups by giving limited bedding and nesting materials from postnatal day 2 to day 9 (P2-P9). One week after the stress paradigm (P16), the dendritic branches and spines of striatal spiny neurons as well as the synapses represented by postsynaptic density protein-95 (PSD-95) in DMS and DLS were stereologically analyzed. Adverse experience in early life selectively affected the spiny neurons in DLS, leading to abundant proximal dendritic branches and an increased number of filopodia-like protrusions, but a reduced number of dendritic spines. The selective effects of stress on neurons in DLS were further identified by reduced expression of PSD-95, including a reduced optical density of PSD-95 immunoreactivity and fewer individual PSD-95 immunoreactive synapses in this region. Notably, stress in early life affected either D1 or D2 dopamine receptor-expressing DLS neurons. These findings suggest that adverse early-life experience delayed the maturation of dendritic spines on neurons in the dorsolateral striatum. Altered dendritic differentiation provoked by stress in early life may contribute critically to the formation of proper neuronal circuits in the dorsal striatum and, therefore, affect striatum-dependent habitual behavior and emotional function later in life.


Assuntos
Corpo Estriado/metabolismo , Dendritos/metabolismo , Neurônios/metabolismo , Estresse Psicológico/metabolismo , Animais , Espinhas Dendríticas/metabolismo , Masculino , Camundongos , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Sinapses/metabolismo
5.
Mol Med Rep ; 21(3): 1172-1180, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31922229

RESUMO

Alzheimer's disease (AD) is the most prevalent age­related neurodegenerative disorder. It is featured by the progressive accumulation of ß­amyloid (Aß) plaques and neurofibrillary tangles. This can eventually lead to a decrease of cholinergic neurons in the basal forebrain. Stem cell transplantation is an effective treatment for neurodegenerative diseases. Previous studies have revealed that different types of stem or progenitor cells can mitigate cognition impairment in different Alzheimer's disease mouse models. However, understanding the underlying mechanisms of neural stem cell (NSC) therapies for AD requires further investigation. In the present study, the effects and the underlying mechanisms of the treatment of AD by NSCs are reported. The latter were labelled with the enhanced green fluorescent protein (EGFP) prior to implantation into the bilateral hippocampus of an amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic (Tg) mouse model of AD. It was observed that the number of basal forebrain cholinergic neurons was restored and the expression of choline acetyltransferase (ChAT) protein was increased. Moreover, the levels of synaptophysin (SYP), postsynaptic density protein 95 (PSD­95) and microtubule­associated protein (MAP­2) were significantly increased in the hippocampus of NSC­treated AD mice. Notably, spatial learning and memory were both improved after transplantation of NSCs. In conclusion, the present study revealed that NSC transplantation improved learning and memory functions in an AD mouse model. This treatment allowed repairing of basal forebrain cholinergic neurons and increased the expression of the cognition­related proteins SYP, PSD­95 and MAP­2 in the hippocampus.


Assuntos
Doença de Alzheimer , Neurônios Colinérgicos , Aprendizagem , Memória , Células-Tronco Neurais , Presenilina-1 , Transplante de Células-Tronco , Sinapses , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Amiloide/genética , Amiloide/metabolismo , Animais , Neurônios Colinérgicos/metabolismo , Neurônios Colinérgicos/patologia , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Células-Tronco Neurais/transplante , Presenilina-1/biossíntese , Presenilina-1/genética , Sinapses/genética , Sinapses/metabolismo , Sinapses/patologia
6.
Neurobiol Dis ; 132: 104586, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31470103

RESUMO

The effects of prolonged physical training on memory performance and underlying presynaptic mechanisms were investigated in old C57BL/6 mice. Training via voluntary running wheels was initiated at 16 months of age and continued for 5 months (1 h per day, 5 days per week), followed by testing of learning and memory functions and counting of presynaptic puncta and cholinergic inputs in the hippocampus. Trained old mice were compared to their age-matched sedentary controls and adult controls. This training strategy improved hippocampal-dependent spatial memory function tested via a novel location task, and enhanced memory was accompanied by restored presynaptic puncta and cholinergic fibers in area CA1 and DG of the hippocampus in old mice. Particularly, the training selectively affected presynaptic vesicle protein synaptophysin but not growth associated protein GAP-43, and the increased number of synaptophysin puncta positively correlates with improved memory performance. To better understand the neurochemical mechanisms by which prolonged physical training protects against aging-related memory deficits, the cholinergic inputs to the hippocampus were compared among the three groups of mice and correlated with memory performance. While the running prevented age-related loss of cholinergic inputs, it has limited impact on the projection source cells in the medial septum-diagonal band (MS-DB). Importantly, cholinergic fibers in area CA1 and DG positively correlated with spatial memory function. These data suggest that the preservation of presynaptic inputs, particularly those involved in the integrity of memory performance, contributes critically to the beneficial effects of physical running initiated at an older age.


Assuntos
Neurônios Colinérgicos/citologia , Condicionamento Físico Animal/fisiologia , Terminações Pré-Sinápticas/metabolismo , Memória Espacial/fisiologia , Sinaptofisina/metabolismo , Envelhecimento , Animais , Hipocampo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL
7.
J Neuroinflammation ; 16(1): 62, 2019 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-30871577

RESUMO

BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by an abnormal accumulation of amyloid-ß (Aß) plaques, neuroinflammation, and impaired neurogenesis. Urolithin A (UA), a gut-microbial metabolite of ellagic acid, has been reported to exert anti-inflammatory effects in the brain. However, it is unknown whether UA exerts its properties of anti-inflammation and neuronal protection in the APPswe/PS1ΔE9 (APP/PS1) mouse model of AD. METHODS: Morris water maze was used to detect the cognitive function. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay was performed to detect neuronal apoptosis. Immunohistochemistry analyzed the response of glia, Aß deposition, and neurogenesis. The expression of inflammatory mediators were measured by enzyme-linked immunosorbent assay (ELISA) and quantitative real-time polymerase chain reaction (qRT-PCR). The modulating effects of UA on cell signaling pathways were assayed by Western blotting. RESULTS: We demonstrated that UA ameliorated cognitive impairment, prevented neuronal apoptosis, and enhanced neurogenesis in APP/PS1 mice. Furthermore, UA attenuated Aß deposition and peri-plaque microgliosis and astrocytosis in the cortex and hippocampus. We also found that UA affected critical cell signaling pathways, specifically by enhancing cerebral AMPK activation, decreasing the activation of P65NF-κB and P38MAPK, and suppressing Bace1 and APP degradation. CONCLUSIONS: Our results indicated that UA imparted cognitive protection by protecting neurons from death and triggering neurogenesis via anti-inflammatory signaling in APP/PS1 mice, suggesting that UA might be a promising therapeutic drug to treat AD.


Assuntos
Anti-Inflamatórios/uso terapêutico , Cumarínicos/uso terapêutico , Citocinas/metabolismo , Encefalite/tratamento farmacológico , Regulação da Expressão Gênica/efeitos dos fármacos , Transtornos da Memória/tratamento farmacológico , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Citocinas/genética , Modelos Animais de Doenças , Encefalite/etiologia , Feminino , Regulação da Expressão Gênica/genética , Gliose/tratamento farmacológico , Gliose/genética , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Placa Amiloide/tratamento farmacológico , Placa Amiloide/etiologia , Presenilina-1/genética , Presenilina-1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
8.
Brain Behav Immun ; 79: 159-173, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30763768

RESUMO

Neuroinflammation, considered as a pathological hallmark of Alzheimer's disease (AD), has been demonstrated to affect hippocampal neurogenesis and cognitive function. Interleukin-6 (IL-6) is a proinflammatory cytokine known to modulate neurogenesis. However, the mechanisms are still largely unknown. Here, we reported that IL-6 suppressed neurogenesis via a JAK2/STAT3 signaling in neural stem cells (NSCs). Importantly, we found that NeuroD1 (Neurogenic differentiation 1) gene expression, which drives NSCs neurodifferentiation, was regulated by TET3 and DNMT1 in a JAK2/STAT3-dependent manner. We further found that JAK2/STAT3 inhibition enhanced demethylation of NeuroD1 regulatory elements in IL-6-treated cells, which is related to the significant upregulation of TET3 expression as well as the decreased expression of DNMT1. Furthermore, Inhibiting JAK2/STAT3 significantly rescued the memory deficits and hippocampal neurogenesis dysfunction in APP/PS1 mice. Our data suggest that JAK2/STAT3 signaling plays a vital role in suppressing neurogenesis of NSCs exposed to IL-6 at the epigenetic level, by regulating DNA methylation/demethylation.


Assuntos
Janus Quinase 2/metabolismo , Neurogênese/fisiologia , Fator de Transcrição STAT3/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Desmetilação do DNA , Metilação de DNA , Dioxigenases/genética , Dioxigenases/metabolismo , Hipocampo/metabolismo , Humanos , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/metabolismo , Neurogênese/imunologia , Neuroimunomodulação , Transdução de Sinais/imunologia
9.
J Neuroinflammation ; 15(1): 268, 2018 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-30227858

RESUMO

BACKGROUND: Netrin-1 functions largely via combined receptors and downstream effectors. Evidence has shown that astrocytes express netrin-1 receptors, including DCC and UNC5H2. However, whether netrin-1 influences the function of astrocytes was previously unknown. METHODS: Lipopolysaccharide was used to stimulate the primary cultured astrocytes; interleukin release was used to track astrocyte activation. In vivo, shRNA and netrin-1 protein were injected in the mouse brain. Infarct volume, astrocyte activation, and interleukin release were used to observe the function of netrin-1 in neuroinflammation and brain injury after middle cerebral artery occlusion. RESULTS: Our results demonstrated that netrin-1 reduced lipopolysaccharide-induced interleukin-1ß and interleukin-12ß release in cultured astrocytes, and blockade of the UNC5H2 receptor with an antibody reversed this effect. Additionally, netrin-1 increased p-AKT and PPAR-γ expression in primary cultured astrocytes. In vivo studies showed that knockdown of netrin-1 increased astrocyte activation in the mouse brain after middle cerebral artery occlusion (p < 0.05). Moreover, injection of netrin-1 attenuated GFAP expression (netrin-1 0.27 ± 0.06 vs. BSA 0.62 ± 0.04, p < 0.001) and the release of interleukins and reduced infarct volume after brain ischemia (netrin-1 0.27 ± 0.06 vs. BSA 0.62 ± 0.04 mm3, p < 0.05). CONCLUSION: Our results indicate that netrin-1 is an important molecule in regulating astrocyte activation and neuroinflammation in cerebral ischemia and provides a potential target for ischemic stroke therapy.


Assuntos
Astrócitos/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Infarto da Artéria Cerebral Média/fisiopatologia , Netrina-1/farmacologia , Animais , Células Cultivadas , Infarto da Artéria Cerebral Média/induzido quimicamente , Interleucinas/metabolismo , Lipopolissacarídeos , Camundongos , Netrina-1/metabolismo
10.
Neurobiol Aging ; 71: 91-104, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30118927

RESUMO

Aging is a normal physiological process associated with impairments in cognitive function, including learning and memory. Here, the underlying synaptic mechanisms by which aging leads to the decline of spatial learning and memory function were investigated in 25-month-old aged mice versus 2-month-old young mice. Deficits of spatial learning and memory, as well as selective loss of thin spines, but not mushroom-type spines on apical dendrites of CA1 pyramidal cells were found in aged mice. Specifically, loss of thin spines in aged mice with memory deficits was primarily found on dendritic segments located in the Schaffer pathway, and the density of thin spines significantly correlated with spatial memory performance. The loss of thin spines was evidenced by a decrease in small synapses that express diminutive amounts of postsynaptic density protein-95 and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subunit GluR1. Furthermore, mushroom-type spines and GluR1-expressed large synapses were not affected in aged mice with impaired memory. Taken together, these data suggest that the selective loss of those highly plastic thin spines with sparse postsynaptic density protein-95 and GluR1 receptors may significantly contribute to cognitive deficits in aged individuals.


Assuntos
Envelhecimento , Região CA1 Hipocampal/fisiologia , Espinhas Dendríticas/fisiologia , Aprendizagem Espacial/fisiologia , Memória Espacial/fisiologia , Sinapses/fisiologia , Animais , Região CA1 Hipocampal/citologia , Masculino , Camundongos Endogâmicos C57BL , Células Piramidais/citologia , Células Piramidais/fisiologia , Receptores de AMPA/metabolismo
11.
Int J Mol Med ; 41(4): 2037-2049, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29393372

RESUMO

Direct derivation of human induced pluripotent stem cells into neural precursor cells and differentiation of these into neurons holds great promise in the cell therapy of neurodegenerative diseases. However, the availability and survival rate of neurons requires improvement. In the present study, it was found that the addition of 5 ng/ml leukocyte inhibitory factor (LIF) during the process of differentiation significantly improved the expression of neuron­specific class III ß­tubulin (TUJ1) and microtubule­associated protein 2 (MAP2), as detected by immunofluorescence and western blotting. In addition, LIF improved the cell viability, increased the expression of phosphorylated­protein kinase B (AKT), downregulated the expression of proinflammatory cytokines, including interleukin­1α (IL­1α) and tumor necrosis factor­α (TNF-α), and upregulated the expression of anti­inflammatory cytokines, including interleukin­10 (IL­10) and transforming growth factor­ß (TGF-ß). After adding the phosphatidylinositol 3-kinase (PI3K)/AKT signaling inhibitor LY294002 or wortmannin to the LIF differentiation group, LIF-induced changes in the protein expression of TUJ1 and MAP2 were reversed, but this effect could not be prevented by rapamycin, a mechanistic target of rapamycin signaling inhibitor. The expression of cytokines associated with inflammation and cell viability was reversed by LY294002 and wortmannin, but not by rapamycin. In conclusion, LIF could improve neuronal differentiation and survival through the activation of PI3K/AKT signaling and the anti­inflammatory effect. The anti­inflammatory effect may be mediated by the activation of PI3K/AKT.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Linfocinas/imunologia , Células-Tronco Neurais/citologia , Neurogênese , Linhagem Celular , Sobrevivência Celular , Citocinas/imunologia , Humanos , Células-Tronco Pluripotentes Induzidas/imunologia , Células-Tronco Neurais/imunologia , Fosfatidilinositol 3-Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Transdução de Sinais
12.
Brain Behav Immun ; 69: 351-363, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29253574

RESUMO

Alzheimer'sdisease(AD) is characterized by deposition of amyloid-ß (Aß)plaques, neurofibrillary tangles, andneuronal loss, accompaniedbyneuroinflammation. Neuroinflammatoryprocesses are thought to contribute toAD pathophysiology. Metformin has been reported to have anti-inflammatory efficacy. However, whether metformin is responsible for the anti-neuroinflammationand neuroprotection on APPswe/PS1ΔE9 (APP/PS1) mice remains unclear. Here we showed that metformin attenuated spatial memory deficit, neuron loss in the hippocampus and enhanced neurogenesis in APP/PS1 mice. In addition, metformin administration decreased amyloid-ß (Aß)plaque load and chronic inflammation (activated microglia and astrocytes as well as pro-inflammatory mediators) in the hippocampus and cortex. Further study demonstrated that treatment with metformin enhanced cerebral AMPK activation. Meanwhile, metformin notably suppressed the activation of P65 NF-κB, mTOR and S6K, reduced Bace1 protein expression. Our data suggest that metformin can exert functional recovery of memory deficits and neuroprotective effect on APP/PS1 mice via triggering neurogenesis and anti-inflammation mediated by regulating AMPK/mTOR/S6K/Bace1 and AMPK/P65 NF-κB signaling pathways in the hippocampus, which may contribute to improvement in neurological deficits.


Assuntos
Doença de Alzheimer/patologia , Hipocampo/efeitos dos fármacos , Transtornos da Memória/prevenção & controle , Metformina/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Placa Amiloide/prevenção & controle , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Metformina/farmacologia , Camundongos , Camundongos Transgênicos , Fármacos Neuroprotetores/farmacologia , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Presenilina-1/genética , Presenilina-1/metabolismo
13.
Cell Death Discov ; 3: 17059, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28904818

RESUMO

Targeting endoplasmic reticulum (ER) stress is being investigated for its anticancer effect in various cancers, including cervical cancer. However, the molecular pathways whereby ER stress mediates cell death remain to be fully elucidated. In this study, we confirmed that ER stress triggered by compounds such as brefeldin A (BFA), tunicamycin (TM), and thapsigargin (TG) leads to the induction of the unfolded protein response (UPR) in cervical cancer cell lines, which is characterized by elevated levels of inositol-requiring kinase 1α, glucose-regulated protein-78, and C/EBP homologous protein, and swelling of the ER observed by transmission electron microscope (TEM). We found that BFA significantly increased autophagy in tumor cells and induced TC-1 tumor cell death in a dose-dependent manner. BFA increased punctate staining of LC3 and the number of autophagosomes observed by TEM in TC-1 and HeLa cells. The autophagic flux was also assessed. Bafilomycin, which blocked degradation of LC3 in lysosomes, caused both LC3I and LC3II accumulation. BFA initiated apoptosis of TC-1 tumor cells through activation of the caspase-12/caspase-3 pathway. At the same time, BFA enhanced the phosphorylation of IκBα protein and translocation into the nucleus of NF-κB p65. Quinazolinediamine, an NF-κB inhibitor, attenuated both autophagy and apoptosis induced by BFA; meanwhile, it partly enhances survival of cervical cancer cells following BFA treatment. In conclusion, our results indicate that the cross-talk between ER stress, autophagy, apoptosis, and the NF-κB pathways controls the fate of cervical cancer cells. Careful evaluation should be given to the addition of an NF-κB pathway inhibitor to treat cervical cancer in combination with drugs that induce ER stress-mediated cell death.

14.
Int J Mol Med ; 40(3): 814-824, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28713933

RESUMO

Pluripotent stem cells (PSCs) are regarded as potential sources that provide specific neural cells for cell therapy in some nervous system diseases. However, the mechanisms underlying the neural differentiation of PSCs remain largely unknown. MicroRNAs (miRNAs or miRs) are a class of small non­pro-tein-coding RNAs that act as critical regulatory molecules in many cellular processes. In this study, we found that miR­146b­5p expression was markedly increased following the neural induction of mouse embryonic stem cells (ESCs) or induced PSCs (iPSCs). In this study, to further identify the role of miR­146b­5p, we generated stable miR­146b­5p-overexpressing ESC and iPSC cell lines, and induced the differentiation of these cells by the adherent monolayer culture method. In the miR­146b­5p-overexpressing ESC- or iPSC-derived cultures, RT-qPCR analysis revealed that the mRNA expression levels of neuroectoderm markers, such as Sox1, Nestin and Pax6, were markedly increased, and flow cytometric analysis verified that the number of Nestin­positive cells was higher in the miR­146b­5p-overexpressing compared with the control cells. Mechanistically, the miR­146b­5p-overexpressing ESCs or iPSCs exhibited a significant reduction in Oct4 expression, which may be an explanation for these cells having a tendency to differentiate towards the neural lineage. Moreover, we confirmed that miR­146b­5p directly targeted Smad4 and negatively regulated the transforming growth factor (TGF)-ß signaling pathway, which contributed to the neural commitment of PSCs. Collectively, our findings uncover the essential role of miR­146b­5p in the neural conversion of PSCs.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , MicroRNAs/biossíntese , Neurônios/metabolismo , Transdução de Sinais , Proteína Smad4/biossíntese , Animais , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , MicroRNAs/genética , Neurônios/citologia , Proteína Smad4/genética
15.
Stem Cell Reports ; 8(5): 1256-1269, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28494938

RESUMO

Although valproic acid (VPA), has been shown to induce neuronal differentiation of neural stem cells (NSCs), the underlying mechanisms remain poorly understood. Here we investigated if and how mammalian target of rapamycin (mTOR) signaling is involved in the neuronal differentiation of VPA-induced NSCs. Our data demonstrated that mTOR activation not only promoted but also was necessary for the neuronal differentiation of NSCs induced by VPA. We further found that inhibition of mTOR signaling blocked demethylation of neuron-specific gene neurogenin 1 (Ngn1) regulatory element in induced cells. These are correlated with the significant alterations of passive DNA demethylation and the active DNA demethylation pathway in the Ngn1 promoter, but not the suppression of lysine-specific histone methylation and acetylation in the promoter region of Ngn1. These findings highlight a potentially important role for mTOR signaling, by working together with DNA demethylation, to influence the fate of NSCs via regulating the expression of Ngn1 in VPA-induced neuronal differentiation of NSCs.


Assuntos
Epigênese Genética , Células-Tronco Neurais/metabolismo , Neurogênese , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Cultivadas , Metilação de DNA , Inibidores Enzimáticos/farmacologia , Código das Histonas , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Regiões Promotoras Genéticas , Ratos , Ratos Sprague-Dawley , Ácido Valproico/farmacologia
16.
J Bioenerg Biomembr ; 48(3): 189-96, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26782277

RESUMO

Mitochondrial dysfunction is a hallmark of amyloid ß peptide (Aß)-induced neuronal toxicity in Alzheimer's disease (AD). However, the precise mechanism(s) of Aß-induced mitochondrial dysfunction has not been fully understood. There is evidence that Forkhead box O3a (FOXO3a) is normally present in neuronal mitochondria. Using HT22 murine hippocampal neuronal cells and primary hippocampal neurons, the present study investigated whether mitochondrial FOXO3a was involved in mitochondrial dysfunction induced by Aß. It was found that Aß induced dephosphorylation and mitochondrial translocation of FOXO3a. In addition, Aß enhanced association of FOXO3a with mitochondrial DNA (mtDNA), causing a decrease in the expression of cytochrome c oxidase subunit 1 (COX1) and the activity of COX. In addition, Aß-induced mitochondrial dysfunction, indicated by the decrease in 3- (4,5-cimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) conversion, mitochondrial adenosine triphosphate (ATP) production and COX activity, could be suppressed by knockdown of FOXO3a (FOXO3a-KD). These results provide new insights into the mechanism underlying Aß-induced neurotoxicity and open up new therapeutic perspectives for AD.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Proteína Forkhead Box O3/fisiologia , Mitocôndrias/metabolismo , Trifosfato de Adenosina/biossíntese , Doença de Alzheimer , Peptídeos beta-Amiloides/farmacologia , Animais , Linhagem Celular , Células Cultivadas , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Proteína Forkhead Box O3/metabolismo , Hipocampo/patologia , Camundongos , Proteínas Mitocondriais/metabolismo , Proteínas Mitocondriais/fisiologia , Neurônios/metabolismo , Neurônios/patologia , Fosforilação/efeitos dos fármacos , Prostaglandina-Endoperóxido Sintases/metabolismo
17.
Med Sci Monit ; 21: 3608-15, 2015 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-26590375

RESUMO

BACKGROUND: Alzheimer's disease (AD) is the most common type of dementia. It causes progressive brain disorder involving loss of normal memory and thinking skills. The transplantation of neural stem cells (NSCs) has been reported to improve learning and memory function of AD rats, and protects basal forebrain cholinergic neurons. Nerve growth factor - poly (ethylene glycol) - poly (lactic-co-glycolic acid)-nanoparticles (NGF-PEG-PLGA-NPs) can facilitate the differentiation of NSCs in vitro. This study thus investigated the treatment efficacy of NGF-PEG-PLGA-NPs combining NSC transplantation in AD model rats. MATERIAL AND METHODS: AD rats were prepared by injection of 192IgG-saporin into their lateral ventricles. Embryonic rat NSCs were separated, induced by NGF-PEG-PLGA-NPs in vitro, and were transplanted. The Morris water-maze test was used to evaluate learning and memory function, followed by immunohistochemical staining for basal forebrain cholinergic neurons, hippocampal synaptophysin, and acetylcholine esterase (AchE) fibers. RESULTS: Rats in the combined treatment group had significantly improved spatial learning ability compared to AD model animals (p<0.05). The number of basal forebrain cholinergic neurons, hippocampal synaptophysin, and AchE-positive fibers were all significantly larger than in the NSC-transplantation group, with no difference from control animals. CONCLUSIONS: NGF-PEG-PLGA-NPs plus NSC transplantation can significantly improve learning and memory functions of AD rats, replenish basal forebrain cholinergic neurons, and help form hippocampal synapses and AchE-positive fibers. These findings may offer practical support for and insight into treatment of Alzheimer's disease.


Assuntos
Doença de Alzheimer/terapia , Nanopartículas/administração & dosagem , Fator de Crescimento Neural/farmacologia , Animais , Prosencéfalo Basal/fisiopatologia , Encéfalo/fisiopatologia , Neurônios Colinérgicos/patologia , Modelos Animais de Doenças , Feminino , Hipocampo/metabolismo , Aprendizagem , Masculino , Memória , Nanopartículas/uso terapêutico , Células-Tronco Neurais/transplante , Poliésteres , Polietilenoglicóis , Ratos , Ratos Sprague-Dawley
18.
Steroids ; 96: 1-6, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25554578

RESUMO

It has been demonstrated that hypothalamus has a programmatic role in aging development, and this role of hypothalamus is mediated by nuclear factor-κB (NF-κB)-directed gonadotropin-releasing hormone (GnRH) decline. ß-Sitosterol (BS), one of the most common phytosterols in the diet, is able to inhibit pro-inflammatory NF-κB signaling. It has been demonstrated that dietary BS can enter the brain and accumulates in brain cell membranes. However, it is unknown whether and how membrane BS affects GnRH release. Using GT1-7 cells, a cell line of GnRH neurons, this study investigated if membrane BS had an influence on GnRH release. It was found that incorporation of BS into the membrane could prevent tumor necrosis factor-α (TNF-α)-induced GnRH decline. The underlying basis involves inhibition of NF-κß activation via estrogen receptor (ER)-mediated inhibition of inhibitor of nuclear factor κB (Iκß) processing. These results extend existing data regarding the beneficial effects of BS, and suggest the use of BS-enriched foods as anti-aging nutrients.


Assuntos
Membrana Celular/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , NF-kappa B/metabolismo , Sitosteroides/metabolismo , Sitosteroides/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Transporte Biológico , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Receptores de Estrogênio/metabolismo
19.
Mol Neurobiol ; 52(1): 601-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25241646

RESUMO

Increasing evidence demonstrates that local inflammation contributes to neuronal death following cerebral ischemia. Peroxisome proliferator-activated receptor α (PPARα) activation has been reported to exhibit many pharmacological effects including anti-inflammatory functions. The aim of this study was to investigate the neuroprotective effects of PPARα agonist fenofibrate on the behavioral dysfunction induced by global cerebral ischemia/reperfusion (GCI/R) injury in rats. The present study showed that fenofibrate treatment significantly reduced hippocampal neuronal death, and improved memory impairment and hippocampal neurogenesis after GCI/R. Fenofibrate administration also inhibited GCI/R-induced over-activation of microglia but not astrocytes and prevented up-regulations of pro-inflammatory mediators in hippocampus. Further study demonstrated that treatment with fenofibrate suppressed GCI/R-induced activations of P65 NF-κB and P38 MAPK. Our data suggest that the PPARα agonist fenofibrate can exert functional recovery of memory deficits and neuroprotective effect against GCI/R in rats via triggering of neurogenesis and anti-inflammatory effect mediated by inhibiting activation of P65 NF-κB and P38 MAPK in the hippocampus, which can contribute to improvement in neurological deficits.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/fisiopatologia , Fenofibrato/farmacologia , Fenofibrato/uso terapêutico , Aprendizagem/efeitos dos fármacos , Transtornos da Memória/tratamento farmacológico , PPAR alfa/agonistas , Animais , Isquemia Encefálica/complicações , Morte Celular/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/patologia , Inflamação/complicações , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/complicações , Transtornos da Memória/fisiopatologia , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/patologia , PPAR alfa/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/patologia , Fator de Transcrição RelA/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Mol Neurobiol ; 51(1): 300-12, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24854198

RESUMO

In the brains of patients with Alzheimer's disease (AD) and transgenic AD mouse models, astrocytes and microglia activated by amyloid-ß (Aß) contribute to the inflammatory process that develops around injury in the brain. Valproic acid (VPA) has been shown to have anti-inflammatory function. The present study intended to explore the therapeutic effect of VPA on the neuropathology and memory deficits in APPswe/PS1ΔE9 (APP/PS1) transgenic mice. Here, we report that VPA-treated APP/PS1 mice markedly improved memory deficits and decreased Aß deposition compared with the vehicle-treated APP/PS1 mice. Moreover, the extensive astrogliosis and microgliosis as well as the increased expression in interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) in the hippocampus and cortex of APP/PS1 transgenic mice were significantly reduced following administration of VPA, which attenuated neuronal degeneration. Concomitantly, VPA alleviated the levels of p65 NF-κB phosphorylation and enhanced the levels of acetyl-H3, Bcl-2, and phospho-glycogen synthase kinase (GSK)-3ß that occurred in the hippocampus of APP/PS1 transgenic mice. These results demonstrate that VPA could significantly ameliorate spatial memory impairment and Aß deposition at least in part via the inhibition of inflammation, suggesting that administration of VPA could provide a therapeutic approach for AD.


Assuntos
Doença de Alzheimer/complicações , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Transtornos da Memória/complicações , Transtornos da Memória/tratamento farmacológico , Ácido Valproico/uso terapêutico , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Morte Celular/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Histonas/metabolismo , Humanos , Imuno-Histoquímica , Transtornos da Memória/genética , Transtornos da Memória/patologia , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Presenilina-1/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Aprendizagem Espacial/efeitos dos fármacos , Ácido Valproico/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...