Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Foods ; 12(21)2023 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-37959035

RESUMO

As cross-border e-commerce platforms become increasingly integrated into the agricultural supply chain, the establishment of a sustainable supply chain ecosystem is of paramount importance. This study, grounded in the platform theory and the supply chain ecosystem theory, combines the grounded theory and the DEMATEL-ISM-MICMAC model to thoroughly analyze the complex mechanisms driving sustainable development. Utilizing the grounded theory, we construct a system of driving factors comprising five primary indicators and eighteen secondary indicators. The hybrid model reveals the interrelationships, significance, system hierarchy, and dependence-driving relationships among these factors. Notably, the driving factor system is categorized into a six-level hierarchical structure, encompassing profound elements, such as policy optimization and digital empowerment, as well as surface-level factors, such as simplification of customs procedures and consumer demand forecasting. Based on the analysis results, this research proposes a set of pathways to achieve the sustainability of the supply chain. These strategies involve improving cross-border agricultural e-commerce policy frameworks, enhancing digital-driven supply-demand coordination, strengthening logistics infrastructure and transparency, and cultivating brand influence. The study's findings not only enrich the relevant theories but also provide practical guidance for the coordinated advancement of economic, social, environmental, and resilient development. Furthermore, they are conducive to advancing the United Nations Sustainable Development Goals.

2.
Biochem Biophys Res Commun ; 647: 47-54, 2023 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-36716645

RESUMO

The aim of this study was to observe the effect of a simulated liver tissue injury microenvironment on the directed differentiation of umbilical cord mesenchymal stem cells into hepatocytes with CYP450 metabolic activity in vitro, and to explore the mechanisms underlying this directed differentiation. Normal and damaged liver tissue homogenate supernatants (LHS and CCl4-LHS, respectively) were used as induction fluids. After induction for different durations, Western blot and RT-PCR were used to measure the protein and gene expression of the hepatocellular proteins AFP, CK18, ALB, and the CYP450 family. Simultaneously, the metabolic activity of CYP450 in hepatocytes was determined. Compared with the LHS and CCl4-LHS controls, the LHS and CCl4-LHS induction groups showed a significantly elevated protein and gene expression of AFP, CK18, ALB, CYP1A1/2, CYP2A6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4 (P < 0.05). The metabolic activity of CYP450 in hepatocytes was increased (P < 0.05). In addition, compared with the LHS group, the CCl4-LHS group induced cell differentiation more rapidly and with a higher efficiency. The results suggested that a liver injury microenvironment is conducive for the directed differentiation of umbilical cord mesenchymal stem cells into hepatocytes with metabolic enzyme activity.


Assuntos
Células-Tronco Mesenquimais , alfa-Fetoproteínas , Fígado , Hepatócitos/metabolismo , Diferenciação Celular , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Cordão Umbilical , Células Cultivadas
3.
Brain Res Bull ; 173: 162-173, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34044033

RESUMO

AIMS: Cholesteryl ester(CE), generated from the mitochondria associated membrane (MAM), is involved in the pathogenesis of Alzheimer's Disease (AD). In theory, the different neuroprotective effects of progesterone in AD are all linked to MAM, yet the effect on cholesterol esterification has not been reported. Therefore, this study was aimed to investigate the regulation of progesterone on intracerebral CE in AD models and the underlying mechanism. METHODS: APP/PS1 mice and AD cell model induced by Aß 25-35 were selected as the research objects. APP/PS1 mice were daily administrated intragastrically with progesterone and The Morris Water Maze test was performed to detect the learning and memory abilities. Intracellular cholesterol was measured by Cholesterol/Cholesteryl Ester Quantitation Assay. The structure of MAMs were observed with transmission electron microscopy. The expression of acyl-CoA: cholesterol acyltransferase 1 (ACAT1), ERK1/2 and p-ERK1/2 were detected with western blotting, immunohistochemistry or immunofluorescence. RESULTS: Progesterone suppressed the accumulation of intracellular CE, shortened the length of abnormally prolonged MAM in cortex of APP/PS1 mice. Progesterone decreased the expression of ACAT1, which could be blocked by progesterone receptor membrane component 1 (PGRMC1) inhibitor AG205. The ERK1/2 pathway maybe involved in the progesterone mediated regulation of ACAT1 in AD models, rather than the PI3K/Akt and the P38 MEPK pathways. SIGNIFICANCE: The results supported a line of evidence that progesterone regulates CE level and the structure of MAM in neurons of AD models, providing a promising treatment against AD on the dysfunction of cholesterol metabolism.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/efeitos dos fármacos , Ésteres do Colesterol/metabolismo , Progesterona/farmacologia , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Esterificação , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Ratos , Ratos Sprague-Dawley
4.
Life Sci ; 253: 117360, 2020 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-32001269

RESUMO

AIMS: Progesterone receptor membrane component 1 (PGRMC1) has been reported to mediate the neuroprotective effect of progesterone, but the exact mechanism has not been elucidated. Therefore, the purpose of this study was to investigate the signalling pathway downstream of PGRMC1 in progesterone-induced neuroprotection. Recognition of the mechanism of progesterone opens novel perspectives for the treatment of diseases of the nervous system. MAIN METHODS: The PGRMC1 protein level was knocked down in rat primary cortical neurons, and Aß25-35 was used to establish an Alzheimer's disease cell model. The neuroprotective effect of progesterone was assessed by Hoechst 33258 staining and a cell counting kit-8 (CCK-8) assay. Then, proteomic and bioinformatic methods were used to analyse the proteins altered in response to PGRMC1 silencing to identify target proteins and signalling pathways involved in PGRMC1-mediated progesterone-induced neuroprotection. These findings were further verified by using signalling pathway inhibitors and western blotting. KEY FINDINGS: The neuroprotective effect of progesterone was significantly attenuated with PGRMC1 silencing. The expression of many proteins in the Ras signalling pathway was significantly changed in response to PGRMC1 silencing. FTI-277 inhibited progesterone-induced neuroprotection. Progesterone increased the expression of total Ras and Grb2. SIGNIFICANCE: These findings provide new perspectives for understanding the mechanism of and role of PGRMC1 in progesterone-induced neuroprotection. The Ras signalling pathway is the signalling pathway downstream of PGRMC1 in the mediation of progesterone-induced neuroprotection.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/química , Proteínas de Membrana/metabolismo , Neuroproteção/efeitos dos fármacos , Progesterona/metabolismo , Receptores de Progesterona/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Apoptose , Sobrevivência Celular , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Cromatografia Líquida de Alta Pressão , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Proteína Adaptadora GRB2/metabolismo , Técnicas de Inativação de Genes/métodos , Inativação Gênica , Humanos , Metionina/análogos & derivados , Metionina/química , Metionina/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteômica , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Espectrometria de Massas em Tandem
5.
Life Sci ; 238: 116979, 2019 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-31647947

RESUMO

AIMS: Alzheimer's disease (AD) is closely related to abnormal glucose metabolism in the central nervous system. Progesterone has been shown to have obvious neuroprotective effects in the pathogenesis of AD, but the specific mechanism has not been fully elucidated. Therefore, the purpose of this study was to investigate the effect of progesterone on the glucose metabolism of neurons in amyloid precursor protein (APP)/presenilin 1 (PS1) mice and Aß-induced AD cell model. MATERIALS AND METHODS: APP/PS1 mice were treated with 40 mg/kg progesterone for 40 days and primary cultured cortical neurons were treated with 1 µM progesterone for 48 h.Then behavior tests,2-NBDG glucose uptake tests and the protein levels of glucose transporter 3 (GLUT3), GLUT4, cAMP-response element binding protein (CREB) and proliferator-activated receptor γ (PPARγ) were examined. KEY FINDINGS: Progesterone increased the expression levels of GLUT3 and GLUT4 in the cortex of APP/PS1 mice, accompanied by an improvement in learning and memory. Progesterone increased the levels of CREB and PPARγ in the cerebral cortex of APP/PS1 mice. In vitro, progesterone increased glucose uptake in primary cultured cortical neurons, this effect was blocked by the progesterone receptor membrane component 1 (PGRMC1)-specific blocker AG205 but not by the progesterone receptor (PR)-specific blocker RU486. Meanwhile, progesterone increased the expression of GLUT3, GLUT4, CREB and PPARγ, and AG205 blocked this effect. SIGNIFICANCE: These results confirm that progesterone significantly improves the glucose metabolism of neurons.One of the mechanisms of this effect is that progesterone upregulates protein expression of GLUT3 and GLUT4 through pathways PGRMC1/CREB/GLUT3 and PGRMC1/PPARγ/GLUT4.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Precursor de Proteína beta-Amiloide/fisiologia , Modelos Animais de Doenças , Glucose/metabolismo , Neurônios/efeitos dos fármacos , Presenilina-1/fisiologia , Progesterona/farmacologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/farmacologia , Animais , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Neurônios/patologia , Progestinas/farmacologia , Ratos , Ratos Sprague-Dawley
6.
Int Immunopharmacol ; 74: 105669, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31176046

RESUMO

Neuroinflammation and autophagy dysfunction are known to be involved in the pathological procession of Alzheimer's disease (AD). Progesterone (PG), neuroactive steroids, exerts a characteristic neuroprotective function in improving AD syndrome. The NOD-like receptor pyrin 3 (NLRP3)-Caspase-1 inflammasome has specific relevance to AD pathological procession. However, the exact role of PG in regulating NLRP3-Caspase-1 inflammasome remains to be elucidated. We demonstrated Aß up-regulated IL-1ß expression in astrocytes by activating NLRP3-Caspase-1 inflammasome. However, pharmacological activation of autophagy by Rapamycin (RAPA) efficiently suppressed Aß-, lipopolysaccharides (LPS)-induced IL-1ß expression via regulating NLRP3-Caspase-1 inflammasome in astrocytes. Remarkably, PG significantly inhibited Aß-induced NLRP3-Caspase-1 inflammasome activation. Autophagy inhibitor 3-MA blocked the protective effects of PG in mediating NLRP3 inflammasome and IL-1ß processing. Taken together, our observations suggest that autophagy-lysosome pathway is one specific molecular mechanism in regulating Aß-induced NLRP3-Caspase-1 inflammasome activation in astrocytes, particularly uncover the potential neuroprotection of PG in regulating upstream signaling leading to the sequence events of neuroinflammation. That neuroprotective mechanism of PG in regulating NLRP3-Caspase-1 inflammasome can be a potential therapeutic target for ameliorating the pathological procession of AD.


Assuntos
Peptídeos beta-Amiloides , Astrócitos/metabolismo , Caspase 1/metabolismo , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Neuroproteção , Progesterona/metabolismo , Animais , Autofagia , Células Cultivadas , Interleucina-1beta/metabolismo , Camundongos
7.
Int Immunopharmacol ; 54: 336-343, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29197800

RESUMO

Autophagy is an intracellular catabolic mechanism essential for recycling intracellular unfolding protein and eliminating toxic protein aggregates. Several studies have shown that deficient autophagy is implicated in the development of Alzheimer's disease (AD) progression. To date, rapidly emerging evidence suggests that neurosteroid progesterone (PG) may play an important role in ameliorating AD. However, the role of PG and its neuroprotective mechanism in regulating autophagy still require further investigation. Here, we investigated the protective effects of PG against Aß-induced inflammatory responses in astrocytes and its underlying mechanism in mediating autophagy. Remarkably, Aß induced astrocyte dysfunction in autophagic activation and up-regulated inflammatory secretion. However, the autophagy inducer rapamycin (RAPA) significantly suppressed Aß-induced inflammation in astrocytes. In astrocytes, treatment with Aß caused autophagy deficiency, whereas PG significantly increased autophagy activation. Finally, PG suppressed Aß-induced neuroinflammatory production via enhancing autophagy together with regulating mTOR signaling. Taken together, these results show that autophagy is a vital mechanism against Aß-induced neuroinflammatory responses in astrocytes and demonstrate the potential neuroprotective mechanism of PG in suppressing neuroinflammatory responses by enhancing autophagy. Therefore, uncovering the neuroprotective mechanism of PG may provide new insight into novel therapies for the amelioration of AD.


Assuntos
Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/metabolismo , Astrócitos/fisiologia , Fragmentos de Peptídeos/metabolismo , Progesterona/metabolismo , Peptídeos beta-Amiloides/imunologia , Animais , Autofagia , Células Cultivadas , Humanos , Inflamação Neurogênica , Fragmentos de Peptídeos/imunologia , Agregação Patológica de Proteínas , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo
8.
J Neurochem ; 141(1): 137-150, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28129443

RESUMO

Neurosteroids are synthesized in the nervous system from cholesterol or steroidal precursors imported from peripheral sources. These compounds are important allosteric modulators of GABAA receptors, which play a vital role in modulating hippocampal functions. Chronic pain is accompanied by increased neurosteroid production in the spinal cord and thalamus. We hypothesize that hippocampal neurosteroids participate in pain or pain-associated emotions, which we tested with high-performance liquid chromatography/tandem mass spectrometry and pharmacological behavioral tests. We observed increased levels of hippocampal neurosteroids (pregnenolone, progesterone, deoxycorticosterone, and allopregnanolone) in rats with chronic neuropathic pain (28 days after spared nerve injury). Meanwhile, the expression of the translocator protein, the upstream steroidogenesis rate-limiting enzyme, increased in the ventral but not dorsal hippocampus of neuropathic rats. In both naïve and neuropathic rats, in vivo stereotaxic microinjection of PK 11195, the translocator protein inhibitor, into the ventral hippocampus exacerbated anxiety-like behaviors. These results indicate anxiolytic effects of hippocampal neurosteroids in both normal and neuropathic rats. Neurosteroids could be considered as agents for treatment of general and pain-related anxiety disorders.


Assuntos
Ansiolíticos/metabolismo , Hipocampo/metabolismo , Neuralgia/metabolismo , Neuralgia/psicologia , Neurotransmissores/metabolismo , Animais , Ansiolíticos/análise , Hipocampo/química , Masculino , Neuralgia/prevenção & controle , Neurotransmissores/análise , Ratos , Ratos Sprague-Dawley
9.
Neurosci Bull ; 32(4): 311-22, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27325509

RESUMO

Neurosteroids are synthesized in the nervous system from cholesterol or steroidal precursors imported from peripheral sources. These compounds are important allosteric modulators of γ-aminobutyric acid A receptors (GABAARs), which play a vital role in pain modulation in the lateral thalamus, a main gate where somatosensory information enters the cerebral cortex. Using high-performance liquid chromatography/tandem mass spectrometry, we found increased levels of neurosteroids (pregnenolone, progesterone, deoxycorticosterone, allopregnanolone, and tetrahydrodeoxycorticosterone) in the chronic stage of neuropathic pain (28 days after spared nerve injury) in rats. The expression of the translocator protein TSPO, the upstream steroidogenesis rate-limiting enzyme, increased at the same time. In vivo stereotaxic microinjection of neurosteroids or the TSPO activator AC-5216 into the lateral thalamus (AP -3.0 mm, ML ±3.0 mm, DV 6.0 mm) alleviated the mechanical allodynia in neuropathic pain, while the TSPO inhibitor PK 11195 exacerbated it. The analgesic effects of AC-5216 and neurosteroids were significantly attenuated by the GABAAR antagonist bicuculline. These results suggested that elevated neurosteroids in the lateral thalamus play a protective role in the chronic stage of neuropathic pain.


Assuntos
Neurotransmissores/metabolismo , Neurotransmissores/uso terapêutico , Ciática/tratamento farmacológico , Tálamo/metabolismo , Animais , Antineoplásicos/farmacologia , Bicuculina/farmacologia , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Transporte/metabolismo , Modelos Animais de Doenças , Antagonistas GABAérgicos/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Hiperalgesia/tratamento farmacológico , Isoquinolinas/farmacologia , Camundongos , Proteínas dos Microfilamentos/metabolismo , Medição da Dor , Fosfopiruvato Hidratase/metabolismo , Purinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/metabolismo , Tálamo/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
10.
Biomed Res Int ; 2016: 8916534, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27088093

RESUMO

Human umbilical cord-derived mesenchymal stem cells (hUCMSCs) are considered to be an ideal cell source for cell therapy of many diseases. The aim of this study was to investigate the contribution of the microenvironment to the hepatic differentiation potential of hUCMSCs in vitro and in vivo and to explore their therapeutic use in acute liver injury in rats. We established a new model to simulate the liver tissue microenvironment in vivo using liver homogenate supernatant (LHS) in vitro. This induced environment could drive hUCMSCs to differentiate into hepatocyte-like cells within 7 days. The differentiated cells expressed hepatocyte-specific markers and demonstrated hepatocellular functions. We also injected hUCMSCs into rats with CCl4-induced acute hepatic injury. The hUCMSCs were detected in the livers of recipient rats and expressed the human hepatocyte-specific markers, suggesting that hUCMSCs could differentiate into hepatocyte-like cells in vivo in the liver tissue microenvironment. Levels of biochemistry markers improved significantly after transplantation of hUCMSCs compared with the nontransplantation group (P < 0.05). In conclusion, this study demonstrated that the liver tissue microenvironment may contribute to the differentiation of hUCMSCs into hepatocytes both in vitro and in vivo.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/terapia , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Cordão Umbilical/citologia , Animais , Biomarcadores/metabolismo , Tetracloreto de Carbono/toxicidade , Diferenciação Celular/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Hepatócitos/citologia , Humanos , Fígado/patologia , Ratos
11.
Int Immunopharmacol ; 33: 83-9, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26878478

RESUMO

The deposition of amyloid-ß (Aß) and neuroinflammation are critical pathological features of Alzheimer's disease (AD). Astrocytes are considered the principal immunoregulatory cells in the brain. Neurosteroid progesterone (PG) exerts neuromodulatory properties, particularly its potential therapeutic function in ameliorating AD. However, the role of PG and the neuroprotective mechanism involving in the regulation of neuroinflammation in astrocytes warrant further investigation. In this study, we found that Aß significantly increased the processing of neuroinflammatory responses in astrocytes. The processing is induced by an increase activity of PERK/elF2ɑ-dependent endoplasmic reticulum (ER) stress. Additionally, the inhibition of ER stress activation by Salubrinal significantly suppressed the Aß-induced neuroinflammatory responses in astrocytes. While the treatment of astrocytes with Aß caused an increase of neuroinflammatory responses, PG significantly inhibited Aß-induced neuroinflammatory cytokine production by suppressing ER stress activation together with attenuating PERK/elF2ɑ signalling. Taken together, these results indicate that PG exerts a neuroprotective effect against Aß-induced neuroinflammatory responses, and significantly suppresses ER stress activation, which is an important mediator of the neurotoxic events occurring in Aß-induced neuroinflammatory responses in astrocytes. These neuroprotective mechanisms may facilitate the development of therapies to ameliorate AD.


Assuntos
Astrócitos/efeitos dos fármacos , Inflamação Neurogênica/tratamento farmacológico , Progesterona/farmacologia , Peptídeos beta-Amiloides/imunologia , Animais , Astrócitos/fisiologia , Células Cultivadas , Cinamatos/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fator de Iniciação 1 em Eucariotos/metabolismo , Humanos , Neuroproteção , Ratos , Ratos Sprague-Dawley , Tioureia/análogos & derivados , Tioureia/farmacologia , eIF-2 Quinase/metabolismo
12.
Sci Rep ; 6: 18982, 2016 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-26732053

RESUMO

Histone modifications have been implicated in learning and memory. Our previous transcriptome data showed that expression of sirtuins 6 (SIRT6), a member of Histone deacetylases (HDACs) family in the hippocampal cornu ammonis 1 (CA1) was decreased after contextual fear conditioning. However, the role of SIRT6 in the formation of memory is still elusive. In the present study, we found that contextual fear conditioning inhibited translational expression of SIRT6 in the CA1. Microinfusion of lentiviral vector-expressing SIRT6 into theCA1 region selectively enhanced the expression of SIRT6 and impaired the formation of long-term contextual fear memory without affecting short-term fear memory. The overexpression of SIRT6 in the CA1 had no effect on anxiety-like behaviors or locomotor activity. Also, we also found that SIRT6 overexpression significantly inhibited the expression of insulin-like factor 2 (IGF2) and amounts of proteins and/or phosphoproteins (e.g. Akt, pAkt, mTOR and p-mTOR) related to the IGF2 signal pathway in the CA1. These results demonstrate that the overexpression of SIRT6 in the CA1 impaired the formation of long-term fear memory, and SIRT6 in the CA1 may negatively modulate the formation of contextual fear memory via inhibiting the IGF signaling pathway.


Assuntos
Região CA1 Hipocampal/metabolismo , Medo , Expressão Gênica , Memória de Longo Prazo , Sirtuínas/genética , Animais , Ansiedade , Comportamento Animal , Condicionamento Psicológico , Perfilação da Expressão Gênica , Técnicas de Transferência de Genes , Masculino , Atividade Motora , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais , Sirtuínas/metabolismo , Somatomedinas/metabolismo , Transdução Genética
13.
Mol Med Rep ; 13(2): 1586-92, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26709052

RESUMO

Numerous studies have suggested that ketamine administration can induce neuroapoptosis in primary cultured cortical neurons. Neurosteroids modulate neuronal function and serve important roles in the central nervous system, however the role of neurosteroids in neuroapoptosis induced by ketamine remains to be elucidated. The present study aimed to explore whether neurosteroidogenesis was a pivotal mechanism for neuroprotection against ketamine-induced neuroapoptosis, and whether it may be selectively regulated under ketamine-induced neuroapoptosis conditions in primary cultured cortical neurons. To study this hypothesis, the effect of ketamine exposure on neurosteroidogenesis in primary cultured cortical neurons was investigated. Cholesterol, a substrate involved in the synthesis of neurosteroids, was added to the culture medium, and neurosteroids were quantified using high-performance liquid chromatography-tandem mass spectrometry analysis. The data demonstrated that cholesterol blocked ketamine-induced neuroapoptosis by promoting the synthesis of various neurosteroids, and the pathway of neurosteroid testosterone conversion into estradiol was inhibited by ketamine exposure. These data suggest that endogenous neurosteroids biosynthesis is critical for neuroprotection against ketamine-induced neuroapoptosis and inhibiting the biosynthesis of neuroprotective-neurosteroid estradiol is of notable importance for ketamine-induced neuroapoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Ketamina/administração & dosagem , Neurônios/efeitos dos fármacos , Neurotransmissores/biossíntese , Animais , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/patologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Colesterol/administração & dosagem , Neurônios/patologia , Fármacos Neuroprotetores , Cultura Primária de Células , Ratos
14.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 30(6): 754-760, 2016 Jun 08.
Artigo em Chinês | MEDLINE | ID: mdl-29786272

RESUMO

OBJECTIVE: To investigate the effect of blood microenvironment of rats with hepatic fibrosis on differentiation of human umbilical cord mesenchymal stem cells (HUCMSCs) into hepatocytes and its mechanisms. METHODS: Eighteen male adult Sprague Dawley rats [weighing, (200±20) g] were used, liver fibrosis was induced in 12 rats by repeated intraperitoneal injections of thioacetamide. The serum was separated after successful model preparation, and the serum of 6 normal rats was collected. ELISA assay was used to detect the concentrations of epidermal growth factor (EGF), hepatocyte growth factor (HGF), oncostatin M (OSM), and basic fibroblastic growth factor (bFGF). Passage 3 HUCMSCs were divided into 3 groups: cells were cultured for 7 days in DMEM/F12 containing 10% fetal bovine serum and 5?mL/ L serum from rats with hepatic fibrosis (group A), in DMEM/F12 containing 10% fetal bovine serum and 5 mL/ L serum from normal rats (group B), and in DMEM/F12 containing 10% fetal bovine serum (group C). The morphological changes of the cells were observed. The expressions of α-fetoprotein (AFP) and cytokeratin 18 (CK18) were detected by immunofluorescence. The protein levels of albumin (ALB), tryptophan 2, 3-dioxygenase (TPH2), and CYP3A4 and MAPK/ERK signal pathway protein (P-ERK) were detected using Western blot. The content of blood urea nitrogen (BUN) was measured by diacetyl m onoxime method. RESULTS: HE staining showed that the liver tissue of rats was in accordance with the change of fibrosis, indicating successful model preparation. In serum of normal rats and rats with hepatic fibrosis, the concentrations of EGF were (21.42±0.32) pg/mL and (17.57±0.31) pg/mL respectively, showing significant difference (t=14.989, P=0.000); the concentrations of OSM were (129.96±0.65) pg/mL and (98.44±1.32) pg/mL respectively, showing significant difference (t=37.172, P=0.000); the concentrations of HGF were below the detection limit and (1.03±0.12)?ng/ mL respectively; and the concentrations of bFGF were lower than the detection limit in both groups. No morphological changes of cells were observed in both groups at 7 days, and there was no significant difference between groups. At 7 days after culture, the cells in group A could express human hepatocyte biomarkers of AFP, CK18 and hepatocyte-specific-function proteins of ALB, TPH2, and CYP3 A4 while cells in groups B and C did not. Western blot showed that cells in each group could express P-ERK protein. The relative level of P-ERK protein in group A was significantly higher than that in groups B and C (P < 0.05), but no significant difference was found between groups B and C (P > 0.05). The BUN concentration of group A [(0.74±0.07)?mmol/ L] was significantly higher than that of groups B [(0.40±0.04)?mmol/ L] and C [(0.38±0.04) mmol/L] (P < 0.05), but no significant difference was shown between groups B and C (P > 0.05). CONCLUSIONS: Under the condition of hepatic fibrosis, the level of HGF will increase while EGF and OSM will decrease. The formed blood microenvironment?will activate MAPK/ERK signal pathway in HUCMSCs, induce them differentiate into hepatocytes.

15.
Artigo em Chinês | MEDLINE | ID: mdl-26540984

RESUMO

OBJECTIVE: To investigate the differentiation potential of human umbilical cord mesenchymal stem cells (HUCMSCs) into hepatocytes induced by rat fibrotic liver tissue extracts. METHODS: Liver fibrosis was induced in the Sprague Dawley rats (weighting, 180-220 g) by repeated intraperitoneal injections of 3% thioacetamide-saline at a dose of 200 mg/kg twice a week for 4 weeks; fibrotic liver tissues were used to prepare liver homogenate supernatants. The HUCMSCs at passage 3 were cultured in DMEM/F12 with 10% fetal bovine serum (FBS) (control group) and in DMEM/F12 with 10% FBS and 50 g/L liver homogenate supernatants (experimental group) for 7 days. The morphological changes of the cells were recorded; the protein levels of cytokeratin 18 (CK18), alpha fetoprotein (AFP), and CYP3A4 were measured using Western blot. The glycogen storing ability of the cells was detected by periodic acid-schiff (PAS) staining. Furthermore, the synthesis of albumin (ALB) and blood urea nitrogen (BUN) was measured. RESULTS: In experimental group, after 1 day of induction, the stem cells of fusiform shape began to lose sharp edges and progressively shrunk, and then they changed into hepatocyte-like cells with round and irregular shape at 7 days. Positive expressions of AFP, CK18, and CYP3A4 were observed in the experimental group, but negative expression in the control group. The concentrations of BUN and ALB were (0.43 ± 0.07) mmol/L and (8.08 ± 0.41) µg/mL in the control group and were (2.52 ± 0.20) mmol/L and (41.48 ± 4.11) µg/mL in the experimental group, showing significant differences (t=24.160, P = 0.000; t = 19.810, P = 0.000). PAS staining results showed navy blue nucleus and lavender cytoplasm in the control group, but dark purple cell body and visible nucleus in the experimental group. CONCLUSION: HUCMSCs could differentiate into hepatocyte-like cells induced by rat fibrotic liver tissue extracts, which have hepatocyte biomarkers (AFP, CK18, and CYP3A4) and hepatocyte-specific functions of glycogen storage, urea production and ALB secretion, so they could partially replace the function of hepatocytes, that may be one of the therapeutic mechanisms of stem cell transplantation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Sangue Fetal/citologia , Fator de Crescimento de Hepatócito/farmacologia , Hepatócitos/citologia , Extratos Hepáticos/farmacologia , Células-Tronco Mesenquimais/citologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Queratina-18/biossíntese , Fígado/embriologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Ratos , Ratos Sprague-Dawley , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Extratos de Tecidos
16.
Sci Rep ; 5: 13327, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-26289919

RESUMO

Drug addiction is considered an aberrant form of learning, and drug-associated memories evoked by the presence of associated stimuli (drug context or drug-related cues) contribute to recurrent craving and reinstatement. Epigenetic changes mediated by DNA methyltransferase (DNMT) have been implicated in the reconsolidation of fear memory. Here, we investigated the role of DNMT activity in the reconsolidation of cocaine-associated memories. Rats were trained over 10 days to intravenously self-administer cocaine by nosepokes. Each injection was paired with a light/tone conditioned stimulus (CS). After acquisition of stable self-administration behaviour, rats underwent nosepoke extinction (10 d) followed by cue-induced reactivation and subsequent cue-induced and cocaine-priming + cue-induced reinstatement tests or subsequently tested to assess the strength of the cocaine-associated cue as a conditioned reinforcer to drive cocaine seeking behaviour. Bilateral intra-basolateral amygdala (BLA) infusion of the DNMT inhibitor5-azacytidine (5-AZA, 1 µg per side) immediately following reactivation decreased subsequent reinstatement induced by cues or cocaine priming as well as cue-maintained cocaine-seeking behaviour. In contrast, delayed intra-BLA infusion of 5-AZA 6 h after reactivation or 5-AZA infusion without reactivation had no effect on subsequent cue-induced reinstatement. These findings indicate that memory reconsolidation for a cocaine-paired stimulus depends critically on DNMT activity in the BLA.


Assuntos
Complexo Nuclear Basolateral da Amígdala/enzimologia , Cocaína/farmacologia , DNA (Citosina-5-)-Metiltransferases/metabolismo , Memória/efeitos dos fármacos , Animais , Azacitidina/farmacologia , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , Masculino , Ratos Sprague-Dawley , Reforço Psicológico
17.
J Steroid Biochem Mol Biol ; 154: 302-11, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25576906

RESUMO

Progesterone, which acts as a neurosteroid in nervous system, has been shown to have neuroprotective effects in different experiments in vitro and in vivo. Our previous study demonstrates that progesterone exerts neuroprotections in Alzheimer's disease-like rats. Present study attempted to evaluate the protective effects of progesterone on Aß-treated neurons and potential mechanisms involved in neuroprotection. Results showed that treatment with progesterone protected primary cultured rat cortical neurons against Aß(25-35)-induced apoptosis. Furthermore, we observed that progesterone alleviated mitochondrial dysfunction by rescuing mitochondrial membrane potential under Aß challenge. Moreover, progesterone could also attenuate Bax/Bcl-2 proteins ratio upregulation and inhibit the activation of caspase-3 in Aß-treated neurons. These indicate that progesterone attenuates Aß(25-35)-induced neuronal toxicity by inhibiting mitochondria-associated apoptotic pathway. Both classic progesterone receptors (classic PR) and progesterone receptor membrane component 1 (PGRMC1), a special progesterone membrane receptor, are broadly expressed throughout the brain. The protective effect of progesterone was partially abolished by PGRMC1 inhibitor AG205 rather than classic PR antagonist RU486 in this study. Additionally, progesterone protected neurons by inhibiting Aß-induced activation of JNK, which was an upstream signaling component in Aß-induced mitochondria-associated apoptotic pathway. But this process was independent of PGRMC1. Taken together, these results suggest that progesterone exerts a protective effect against Aß(25-35)-induced insults at least in part by two complementary pathways: (1) progesterone receptor membrane component 1-dependent inhibition of mitochondrial apoptotic pathway, and (2) blocking Aß-induced JNK activation. The present study provides new insights into the mechanism by which progesterone brings neuroprotection. This article is part of a Special Issue entitled 'Steroids & Nervous System'.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Apoptose/efeitos dos fármacos , MAP Quinase Quinase 4/antagonistas & inibidores , Proteínas de Membrana/fisiologia , Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Progesterona/farmacologia , Receptores de Progesterona/fisiologia , Animais , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Neurônios/enzimologia , Ratos , Ratos Sprague-Dawley
18.
Brain Res ; 1593: 30-9, 2014 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-25234726

RESUMO

Previous studies have demonstrated that the commonly used anesthetic ketamine can induce widespread neuroapoptosis in the neonatal brain and can cause persistent cognitive impairments as the animal matures. Therefore, searching for adjunctive neuroprotective strategies that inhibit ketamine-induced neuroapoptosis and persistent cognitive impairments is highly warranted. The primary goal of this study was to investigate the protective effect of 17ß-estradiol against ketamine-induced neuroapoptosis and persistent cognitive impairments in adult rats. Starting from postnatal day 7, Sprague-Dawley male rat pups were given a daily administration of ketamine (75mg/kg, i.p.) or 17ß-estradiol (600µg/kg, s.c.) in combination with ketamine (75mg/kg, i.p.). The animals were treated for three consecutive days. 24h after the last injection, the rats were decapitated, and the prefrontal cortex (PFC) was isolated to detect neuroapoptosis by cleaved caspase-3 immunohistochemistry and by using the TUNEL assay. The neuroactive steroid 17ß-estradiol was quantified using high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). The protein levels of BDNF and pAkt were measured by western blot analysis. At two months of age (60 days), the learning and memory abilities were tested using the Morris water maze. The results showed that ketamine triggered significant neuroapoptosis in the neonatal PFC accompanied by the downregulation of 17ß-estradiol, BDNF and pAkt. The co-administration of 17ß-estradiol with ketamine attenuated these changes. Moreover, 17ß-estradiol significantly reversed the learning and memory deficits observed at 60 days of age. In brief, our present data demonstrate that 17ß-estradiol attenuates ketamine-induced neuroapoptosis and reverses long-term cognitive deficits in developing rats and thus may be a potential therapeutic and neuroprotective method for the treatment of neurodevelopmental disorders. This article is part of a Special Issue entitled SI: Brain and Memory.


Assuntos
Transtornos Cognitivos/tratamento farmacológico , Estradiol/farmacologia , Ketamina/toxicidade , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Anestésicos Dissociativos/toxicidade , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Transtornos Cognitivos/induzido quimicamente , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Neurônios/fisiologia , Fosforilação , Córtex Pré-Frontal/crescimento & desenvolvimento , Córtex Pré-Frontal/fisiopatologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Sprague-Dawley , Memória Espacial/efeitos dos fármacos , Memória Espacial/fisiologia
19.
PLoS One ; 9(3): e91019, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24614080

RESUMO

Electromagnetic pulse (EMP) causes central nervous system damage and neurobehavioral disorders, and sevoflurane protects the brain from ischemic injury. We investigated the effects of sevoflurane on EMP-induced brain injury. Rats were exposed to EMP and immediately treated with sevoflurane. The protective effects of sevoflurane were assessed by Nissl staining, Fluoro-Jade C staining and electron microscopy. The neurobehavioral effects were assessed using the open-field test and the Morris water maze. Finally, primary cerebral cortical neurons were exposed to EMP and incubated with different concentration of sevoflurane. The cellular viability, lactate dehydrogenase (LDH) release, superoxide dismutase (SOD) activity and malondialdehyde (MDA) level were assayed. TUNEL staining was performed, and the expression of apoptotic markers was determined. The cerebral cortexes of EMP-exposed rats presented neuronal abnormalities. Sevoflurane alleviated these effects, as well as the learning and memory deficits caused by EMP exposure. In vitro, cell viability was reduced and LDH release was increased after EMP exposure; treatment with sevoflurane ameliorated these effects. Additionally, sevoflurane increased SOD activity, decreased MDA levels and alleviated neuronal apoptosis by regulating the expression of cleaved caspase-3, Bax and Bcl-2. These findings demonstrate that Sevoflurane conferred neuroprotective effects against EMP radiation-induced brain damage by inhibiting neuronal oxidative stress and apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Lesões Encefálicas/patologia , Campos Eletromagnéticos , Éteres Metílicos/farmacologia , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Lesões Encefálicas/complicações , Lesões Encefálicas/tratamento farmacológico , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/patologia , Cognição/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Masculino , Malondialdeído/metabolismo , Éteres Metílicos/uso terapêutico , Degeneração Neural/complicações , Degeneração Neural/tratamento farmacológico , Degeneração Neural/patologia , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/ultraestrutura , Ratos Sprague-Dawley , Sevoflurano , Superóxido Dismutase/metabolismo , Proteína X Associada a bcl-2/metabolismo
20.
Zhonghua Gan Zang Bing Za Zhi ; 22(12): 910-4, 2014 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-25654284

RESUMO

OBJECTIVE: To observe the in vivo migration of human umbilical cord mesenchymal stem cells (hUCMSCs) labeled with the PKH26 red fluorescent dye after transplantation into rats with liver cirrhosis. METHODS: Frozen hUCMSCs were resuscitated and labeled with PKH26. Labeling efficiency and fluorescent maintenance time of the PKH26-1abeled cells were measured. Morphology of the labeled and unlabeled (control) cells was observed by microscopy. The cell growth curve was determined using the MTT method. The PKH26-1abeled hUCMSCs were transplanted via tail vein injection into healthy (control) rats and rats with liver cirrhosis. Migration of the PKH26-1abeled hUCMSCs observed 48 h later in frozen liver sections under a fluorescence microscope. RESULTS: The labeling ratio of PKH26 to hUCMSCs was 100%. Growth of the labeled cells was good. The cell morphology was not significantly different between the labeled and unlabeled cells; all cells were long and spindle-like. Cell proliferation was not impacted significantly by labeling.Fluorescence was maintained for at least 20 days, as detected by in vitro analysis. After transplantation into the rats, the PKH26-1abeled hUCMSCs were mainly distributed in the area surrounding the portal vein, the blood vessels, and the false lobule of the cirrhotic liver; a small amount ofhUCMSCs were present in the spleen and lung. CONCLUSION: PKH26 is an ideal fluorescent dye to label hUCMSCs. The PKH26 labeling technique can be used to study the migration of hUCMSCs in cirrhotic liver.


Assuntos
Cirrose Hepática , Células-Tronco Mesenquimais , Cordão Umbilical , Animais , Proliferação de Células , Corantes Fluorescentes , Humanos , Compostos Orgânicos , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...