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

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biochem Genet ; 60(6): 2052-2068, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35235083

RESUMEN

Severe Acute Respiratory Syndrome Coronavirus Type 2 (SARS-CoV-2) is an enveloped single-stranded RNA virus that can lead to respiratory symptoms and damage many organs such as heart, kidney, intestine, brain and liver. It has not been clearly documented whether myocardial injury is caused by direct infection of cardiomyocytes, lung injury, or other unknown mechanisms. The gene expression profile of GSE150392 was obtained from the Gene Expression Omnibus (GEO) database. The processing of high-throughput sequencing data and the screening of differentially expressed genes (DEGs) were implemented by R software. The R software was employed to analyze the Gene Ontology (GO) analysis and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. The protein-protein interaction (PPI) network of the DEGs was constructed by the STRING website. The Cytoscape software was applied for the visualization of PPI network and the identification of hub genes. The statistical analysis was performed by the GraphPad Prism software to verify the hub genes. A total of 516 up-regulated genes and 191 down-regulated genes were screened out. The top 1 enrichment items of GO in biological process (BP), Cellular Component (CC), and Molecular Function (MF) were type I interferon signaling pathway, sarcomere, and receptor ligand activity, respectively. The top 10 enrichment pathways, including TNF signaling pathway, were identified by KEGG enrichment analysis. A PPI network was established, consisting of 613 nodes and 3,993 edges. The 12 hub genes were confirmed as statistically significant, which was verified by GSE151879 dataset. In conclusion, the hub genes of human iPSC-cardiomyocytes infected with SARS-CoV-2 were identified through bioinformatics analysis, which may be used as biomarkers for further research.


Asunto(s)
COVID-19 , Células Madre Pluripotentes Inducidas , Humanos , SARS-CoV-2 , Perfilación de la Expresión Génica , Miocitos Cardíacos , COVID-19/genética , Biología Computacional , Transducción de Señal/genética
2.
Exp Eye Res ; 181: 15-24, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30633922

RESUMEN

Methyl 3,4 dihydroxybenzoate (MDHB) is a small molecule that shows neuroprotective effects in vitro and in a photoreceptor-degenerative mouse model. Here we investigated whether MDHB protects retina in a mouse model of acute ocular hypertension (AOH) and explores the underlying mechanisms. AOH was induced in mice by increasing intraocular pressure to approximately 90 mmHg for 60 min, then MDHB or vehicle was intraperitoneally injected daily up to 7 days. Immunostaining and multi-electrode array recordings were performed to examine the structure and function of retinas receiving the treatments. Western-blotting was applied to test the expression of several proteins related to oxidative stress and brain-derived neurotrophic factor (BDNF)-initiated signaling. Results showed that AOH injury reduced the number of Brn3a-stained retinal ganglion cells (RGCs) and ChAT-amacrine cells; thinned the inner retinal layers and induced apoptosis. Physiologically, AOH decreased the response of OFF and ON-OFF RGCs. All of these changes were reversed by MDHB-treatment. Mechanistically, MDHB appeared to work on three parallel pathways: (1) MDHB decreased the production of reactive oxygen species, the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and cytosol heme oxygenase 1 (HO-1); (2) It upregulated the expression of BDNF and its receptor tropomyosin-related kinase B (TrkB), and activated the downstream AKT pathways; (3) It inhibited reactive gliosis by reducing GFAP and Iba-1 expression. Thus our results suggest that MDHB protects retina against AOH injury by inhibiting oxidative stress, activating the BDNF/AKT signaling and inhibiting inflammatory pathways. Therefore, MDHB may serve as a promising candidate to treat retinal ischemia.


Asunto(s)
Hidroxibenzoatos/uso terapéutico , Hipertensión Ocular/tratamiento farmacológico , Animales , Apoptosis , Biomarcadores/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Hidroxibenzoatos/farmacología , Masculino , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Hipertensión Ocular/metabolismo , Hipertensión Ocular/fisiopatología , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/fisiología , Transducción de Señal/efectos de los fármacos
3.
Cell Physiol Biochem ; 48(1): 237-250, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30007960

RESUMEN

BACKGROUND/AIMS: Thermal injury causes pulmonary edema and can lead to death. Intercellular junctions are composed of adhesive (p120ctn, E-cadherin, α-catenin and ß-catenin) and compact (occludin and ZO-1) junctions. Heat deteriorates intercellular junctions and increases cell gaps to ultimately induce pulmonary edema, but the underlying mechanism remains elusive. METHODS: Mouse lung epithelial (MLE-12) cells pre-treated with the c-Src inhibitor PP2, p120ctn catenin (p120ctn) small interfering RNA and p120ctn catenin (p120ctn) complementary DNA were subjected to heat treatment. Western blotting and real-time polymerase chain reaction assays were used to evaluate junction protein expression changes after heat treatment, and co-immunoprecipitation was used to test the binding state of junction proteins. In addition, hematoxylin and eosin staining and immunohistochemistry were used to evaluate changes in junction protein expression and lung injury in a Wistar rat model of thermal inhalation injury. RESULTS: Heat increased cell permeability; induced ZO-1, occludin, α-catenin and ß-catenin degradation; and decreased E-cadherin distribution in cell membranes. Heat also activated c-Src and decreased both p120ctn expression levels and occludin and ZO-1 association. c-Src inhibitor (PP2) treatment and p120ctn overexpression reversed these effects and attenuated lung injury in vivo. CONCLUSION: Heat induces junction protein degradation and dissociation to increase membrane permeability and cause lung edema via c-Src kinase activation and p120ctn expression downregulation.


Asunto(s)
Cateninas/metabolismo , Familia-src Quinasas/metabolismo , Animales , Proteína Tirosina Quinasa CSK , Cadherinas/metabolismo , Cateninas/antagonistas & inhibidores , Cateninas/genética , Línea Celular , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Regulación hacia Abajo , Expresión Génica/efectos de los fármacos , Calor , Pulmón/citología , Pulmón/metabolismo , Masculino , Ocludina/metabolismo , Proteolisis , Pirimidinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína de la Zonula Occludens-1/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/genética , Catenina delta
4.
Int J Mol Sci ; 19(6)2018 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-29874838

RESUMEN

Genetic studies have elucidated mechanisms that regulate aging; however, there has been little progress in identifying drugs that retard ageing. Caenorhabditis elegans is among the classical model organisms in ageing research. Methyl 3,4-dihydroxybenzoate (MDHB) can prolong the life-span of C. elegans, but the underlying molecular mechanisms are not yet fully understood. Here, we report that MDHB prolongs the life-span of C. elegans and delays age-associated declines of physiological processes. Besides, MDHB can lengthen the life-span of eat-2 (ad1113) mutations, revealing that MDHB does not work via caloric restriction (CR). Surprisingly, the life-span⁻extending activity of MDHB is completely abolished in daf-2 (e1370) mutations, which suggests that daf-2 is crucial for a MDHB-induced pro-longevity effect in C. elegans. Moreover, MDHB enhances the nuclear localization of daf-16/FoxO, and then modulates the expressions of genes that positively correlate with defenses against stress and longevity in C. elegans. Therefore, our results indicate that MDHB at least partially acts as a modulator of the daf-2/daf-16 pathway to extend the lifespan of C. elegans, and MDHB might be a promising therapeutic agent for age-related diseases.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Factores de Transcripción Forkhead/genética , Hidroxibenzoatos/farmacología , Longevidad/genética , Receptor de Insulina/genética , Envejecimiento/efectos de los fármacos , Envejecimiento/genética , Envejecimiento/fisiología , Animales , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiología , Restricción Calórica , Humanos , Longevidad/efectos de los fármacos , Mutación , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética
5.
Exp Eye Res ; 162: 86-96, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28709891

RESUMEN

Retinitis pigmentosa is a photoreceptor-degenerative disease that is currently untreatable and eventually causes blindness. Methyl 3,4 dihydroxybenzoate (MDHB) is a small molecule that exerts neuroprotective effects in vitro. The present study tests whether MDHB protects the retina of rd10 mice, a model of retinitis pigmentosa. MDHB or an equal volume of vehicle was intraperitoneally injected in rd10 mice daily from postnatal day 12 (P12) to P26. Retinal morphology was evaluated by immunostaining, and retinal function by electroretinogram (ERG) and by visual behavior. TUNEL, Iba1, GFAP staining and western blotting were applied to explore the neuroprotective mechanism of MDHB in retina. MDHB treatment significantly promoted photoreceptor survival and preserved cone morphology compared to the untreated animals. The visual behavior and ERG responses were also greatly enhanced in MDHB-treated rd10 mice. Mechanistically, following MDHB treatment, the number of TUNEL-positive cells was decreased in rd10 retina, and the expression of brain-derived neurotrophic factor (BDNF) protein and phosphorylated tropomyosin-related kinase B (TrkB) receptor were increased. Furthermore, blocking TrkB using the antagonist ANA-12 prevented the protective effect of MDHB on photoreceptor survival and structure. MDHB treatment also inhibited microglial activation and Muller cell gliosis in rd10 retina. In conclusion, MDHB treatment delays retinal degeneration in rd10 mice and preserves retinal structure and functions. These effects are likely mediated by the BDNF-TrkB pathway. Due to its neurotrophic effects and ability to reduce reactive gliosis, MDHB may be useful to treat degenerative diseases in retina and brain.


Asunto(s)
Hidroxibenzoatos/farmacología , Fármacos Neuroprotectores/farmacología , Células Fotorreceptoras Retinianas Conos/patología , Retinitis Pigmentosa/tratamiento farmacológico , Animales , Apoptosis , Western Blotting , Supervivencia Celular , Modelos Animales de Enfermedad , Electrorretinografía , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Células Fotorreceptoras Retinianas Conos/efectos de los fármacos , Células Fotorreceptoras Retinianas Conos/metabolismo , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología
6.
Mol Cell Neurosci ; 71: 80-91, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26711806

RESUMEN

The Nogo-66 receptor (NgR1), a receptor for Nogo-A, contributes to the inhibition of axonal regeneration in the adult central nervous system after traumatic injuries. Thus, NgR1 has been considered a critical target in axon regeneration therapy. Here, we identified a specific NgR1 antagonist peptide (HIYTALV, named NAP2) which promotes neurite regeneration in vitro from a phage display heptapeptide library. NAP2 was co-localized with NgR1 on the surface of PC12 cells and cerebellar granule cells (CGCs) by immunofluorescence assay. Horseradish peroxidase (HRP)-streptavidin-biotin assay further showed that NAP2 binds to NgR1 and the dissociation constant (Kd) was 0.45 µM Functional analyses indicated that NAP2 could reduce the inhibitory effects of Nogo-66 on neurite outgrowth in differentiated PC12 cells and CGCs by blocking the Nogo-66-induced activation of Rho-associated coiled coil-containing protein kinase (ROCK), collapsin response mediator protein 2 (CRMP2) and myosin light chain (MLC). Taken together, the small molecule NgR1 antagonist peptide NAP2 (MW: 815.98Da) has a potential ability in crossing blood brain barrier and will be a promising therapeutic agent for the treatment of spinal cord injury and neurodegenerative diseases.


Asunto(s)
Proteínas de la Mielina/antagonistas & inhibidores , Regeneración Nerviosa , Neuritas/efectos de los fármacos , Oligopéptidos/farmacología , Animales , Células Cultivadas , Cerebelo/citología , Cerebelo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ligandos , Proteínas de la Mielina/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuritas/metabolismo , Neuritas/fisiología , Proteínas Nogo , Células PC12 , Unión Proteica , Ratas , Ratas Sprague-Dawley , Quinasas Asociadas a rho/metabolismo
7.
Molecules ; 21(8)2016 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-27556437

RESUMEN

This study investigated the neuroprotective effects of methyl 3,4-dihydroxybenzoate (MDHB) against t-butyl hydroperoxide (TBHP) induced oxidative damage in SH-SY5Y (human neuroblastoma cells) and the underlying mechanisms. SH-SY5Y were cultured in DMEM + 10% FBS for 24 h and pretreated with different concentrations of MDHB or N-acetyl-l-cysteine (NAC) for 4 h prior to the addition of 40 µM TBHP for 24 h. Cell viability was analyzed using the methylthiazolyltetrazolium (MTT) and lactate dehydrogenase (LDH) assays. An annexin V-FITC assay was used to detect cell apoptosis rates. The 2',7'-dichlorofluorescin diacetate (DCFH-DA) assay was used to determine intracellular ROS levels. The activities of antioxidative enzymes (GSH-Px and SOD) were measured using commercially available kits. The oxidative DNA damage marker 8-OHdG was detected using ELISA. Western blotting was used to determine the expression of Bcl-2, Bax, caspase 3, p-Akt and Akt proteins in treated SH-SY5Y cells. Our results showed that MDHB is an effective neuroprotective compound that can mitigate oxidative stress and inhibit apoptosis in SH-SY5Y cells.


Asunto(s)
Daño del ADN/efectos de los fármacos , Hidroxibenzoatos/farmacología , Neuronas/citología , Fármacos Neuroprotectores/farmacología , terc-Butilhidroperóxido/efectos adversos , Acetilcisteína/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular , Regulación de la Expresión Génica/efectos de los fármacos , Glutatión Peroxidasa/metabolismo , Humanos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Superóxido Dismutasa/metabolismo
8.
Appl Microbiol Biotechnol ; 99(14): 5997-6007, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25758955

RESUMEN

Nogo-66, a hydrophilic loop of 66 amino acids flank two hydrophobic domains of the Nogo-A C terminus, interacts with the Nogo-66 receptor (NgR) to exert numerous functions in the central nervous system (CNS). Nogo-66 has important roles in aspects of neuronal development, including cell migration, axon guidance, fasciculation, and dendritic branching, and in aspects of CNS plasticity, including oligodendrocyte differentiation and myelination. Here, the small ubiquitin-related modifier (SUMO) was fused to the target gene, Nogo-66, and the construct was expressed in Escherichia coli (E. coli). Under the optimal fermentation conditions, the soluble expression level of the fusion protein was 33 % of the total supernatant protein. After cleaving the fusion proteins with SUMO protease and purifying them by Ni-NTA affinity chromatography, the yield and purity of recombinant Nogo-66 obtained by 10-L scale fermentation were 23 ± 1.5 mg/L and greater than 93 %, respectively. The authenticity of the recombinant Nogo-66 was confirmed by an electrospray ionization-mass spectrometry analysis. The functional analyses indicated that the recombinant Nogo-66 was capable of binding the NgR specifically. The immunofluorescence results showed that the recombinant Nogo-66 could significantly inhibit neurite outgrowth of rat pheochromocytoma (PC12) cells stimulated by nerve growth factor and cerebellar granule cells (CGCs). Furthermore, Nogo-66 inhibited neurite outgrowth by increasing the level of phosphorylated Rho-associated coiled-coil-containing protein kinase 2 (ROCK2), collapsin response mediator protein 2 (CRMP2), and myosin light chain (MLC). This study provided a feasible and convenient production method for generating sufficient recombinant Nogo-66 for experimental and clinical applications.


Asunto(s)
Proteínas de la Mielina/metabolismo , Neuritas/efectos de los fármacos , Proteína SUMO-1/metabolismo , Animales , Línea Celular Tumoral , Cromatografía de Afinidad , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Péptidos y Proteínas de Señalización Intercelular , Proteínas de la Mielina/genética , Proteínas de la Mielina/aislamiento & purificación , Cadenas Ligeras de Miosina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nogo , Unión Proteica , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína SUMO-1/genética , Espectrometría de Masa por Ionización de Electrospray , Quinasas Asociadas a rho/metabolismo
9.
J Pharmacol Sci ; 125(1): 51-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24849190

RESUMEN

In the present study, we investigated the protective effect of methyl 3,4-dihydroxybenzoate (MDHB) against H2O2-induced apoptosis in RGC-5 cells. The RGC-5 cells were cultured in plates for 24 h, which were then pretreated with dimethyl sulfoxide, different concentrations of MDHB, or probucol for 12 h prior to addition of 300 µM H2O2 for 24 h. The cell viability was detected by MTT assay. The rate of apoptosis, level of lipid peroxidation, and mitochondrial membrane potential (MMP) were detected by flow cytometry. Western blot analysis was also used to measure the expression level of Bcl-2, Bax, caspase 9, and caspase 3 proteins in H2O2-treated RGC-5 cells. Our study showed that the cell viability of RGC-5 cells significantly decreased after treatment with 300 µM H2O2 for 24 h, but MDHB (8, 16, 32 µM) increased RGC-5 cell survival, suppressed the rate of apoptosis, scavenged reactive oxygen species, and restored MMP. MDHB also obstructed H2O2-induced apoptosis by regulating the expression of Bcl-2 and Bax, as well as suppressing the activation of caspase 9 and caspase 3. Our results showed that MDHB is an effective neuroprotective compound that mitigates oxidative stress and inhibits apoptosis in RGC-5 cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Peróxido de Hidrógeno/efectos adversos , Hidroxibenzoatos/farmacología , Fármacos Neuroprotectores/farmacología , Células Ganglionares de la Retina/patología , Apoptosis/genética , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Expresión Génica/efectos de los fármacos , Humanos , Hidroxibenzoatos/uso terapéutico , Peroxidación de Lípido/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Degeneración Retiniana/tratamiento farmacológico , Células Ganglionares de la Retina/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
10.
Yao Xue Xue Bao ; 49(10): 1372-6, 2014 Oct.
Artículo en Zh | MEDLINE | ID: mdl-25577865

RESUMEN

Alzheimer's disease (AD) is a kind of neurodegenerative diseases, the most common cause of dementia. Although AD has been studied more than, 100 years and the Aß and tau theory are most widely accepted among the theories achieved, yet it is not really clear what the mechanism related to AD works up to now. However, it is certain that AD is a kind of diseases resulting from multi-causes. Except for causes correlated with heredity, aging and life habits, environmental role is worth taking into consideration as well. Some metals, such as copper, aluminum, zinc and iron et al, can also have close relationship with AD. Now, we make an overview on the correlative researches in the field.


Asunto(s)
Enfermedad de Alzheimer/patología , Metales/efectos adversos , Aluminio , Cobre , Humanos , Hierro , Zinc
11.
J Neurosci Res ; 91(9): 1215-25, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23861072

RESUMEN

Amyloid-ß peptides (Aß), which can aggregate into oligomers or fibrils in neurons, play a critical role in the pathogenesis of Alzheimer's disease (AD). Methyl 3,4-dihydroxybenzoate (MDHB), a phenolic acid compound, has been reported to have antioxidative and neurotrophic effects. The present study investigated the neuroprotective effects of MDHB against Aß-induced apoptosis in rat primary cortical neutons. The primary cortical neurons were pretreated with different concentrations of MDHB for 24 hr, then incubated with 10 µM Aß25-35 for 24 hr. The results showed that Aß25-35 could induce neurotoxicity as evidenced by the decreased cell viability and the increased apoptotic rate. In parallel, Aß25-35 significantly increased the reactive oxygen species accumulation and decreased mitochondrial membrane potential. However, pretreatment of the primary cortical neurons with MDHB could effectively suppress these cellular events caused by Aß25-35 exposure. In addition, MDHB could increase the level of Bcl-2, decrease the level of Bax, and inhibit the activation of caspase-9 and caspase-3 in Aß25-35 -treated primary cortical neurons. All these results were beneficial in their protective effect against Aß-induced neurotoxicity. Our results suggest that MDHB has a neuroprotective effect that provides a pharmacological basis for its clinical use in the treatment of AD.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Hidroxibenzoatos/farmacología , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/toxicidad , Análisis de Varianza , Animales , Animales Recién Nacidos , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Líquido Intracelular/efectos de los fármacos , Líquido Intracelular/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Proteína X Asociada a bcl-2/metabolismo
12.
Zhong Yao Cai ; 36(9): 1477-80, 2013 Sep.
Artículo en Zh | MEDLINE | ID: mdl-24620696

RESUMEN

OBJECTIVE: To study the neurotrophic effects of senegenin on the expression of MAP2 mRNA and BDNF mRNA in cultured cerebral cortical neurons. METHODS: The newborn rat cerebral cortex neurons were cultured in vitro. LDH assay was used to investigate the effect of senegenin on the neuronal viability and reverse transcription polymerase chain reaction (RT-PCR) was carried out to determine the expression level of MAP2 mRNA and BDNF mRNA. RESULTS: LDH assay showed that senegenin at the concentration of 0. 5 micromol/L,1 micromol/L and 2 micromol/L could obviously enhance the survival of cells and the survival rates were in dose-dependent manner to some extent. Moreover, the low, medium and high concentrations of senegenin significantly increased the expression of MAP2 mRNA and BDNF mRNA. CONCLUSION: The study suggests that suitable dose of senegenin can increase the expression of MAP2 mRNA and BDNF mRNA in cultured cerebral cortical neurons, and its mechanism needs further study.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Corteza Cerebral/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Animales Recién Nacidos , Factor Neurotrófico Derivado del Encéfalo/genética , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/administración & dosificación , Femenino , L-Lactato Deshidrogenasa/metabolismo , Masculino , Neuritas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/administración & dosificación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Int J Neurosci ; 122(5): 248-54, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22176219

RESUMEN

In this study, we evaluated the neurotrophic effects of magnesium fructose 1, 6-diphosphate (FDP-Mg) on cortical neurons. The results demonstrated that FDP-Mg promoted dendrite outgrowth and neuronal survival in a dose-dependent manner. In order to investigate the associated mechanisms, we determined adenosine triphosphate (ATP) levels and brain-derived neurotrophic factor (BDNF) mRNA expression in cortical neurons. Treatment with FDP-Mg significantly increased ATP levels and BDNF mRNA expression in cortical neurons. These data suggest that FDP-Mg can exert neurotrophic effects on cortical neurons. The increases in BDNF mRNA expression and cellular ATP levels are involved in the neurotrophic effects produced by FDP-Mg.


Asunto(s)
Corteza Cerebral/citología , Dendritas/efectos de los fármacos , Fructosadifosfatos/farmacología , Magnesio/farmacología , Neuronas/efectos de los fármacos , Animales , Animales Recién Nacidos , Supervivencia Celular/efectos de los fármacos , Dendritas/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Neuronas/citología , Neuronas/fisiología , Ratas
14.
Zhong Yao Cai ; 35(11): 1813-6, 2012 Nov.
Artículo en Zh | MEDLINE | ID: mdl-23627096

RESUMEN

OBJECTIVE: To investigate the effect of protocatechuic acid on the mRNA expression of APP in double transfected (human APP gene and presenlin-1 gene) Chinese hamster ovary (CHO) cells (M146L). METHODS: Abeta42 overexpressing cell model was established in vitro by culturing Chinese hamster ovary cells stably expressing amyloid beta-protein precursor and mutant presenilin (M146L). The MTT assay was used to test cytotoxicity of protocatechuic acid, and reverse transcription polymerase chain reaction (RT-PCR) was carried out to determine the mRNA expression level of APP. RESULTS: The MTT assay showed that protocatechuic acid at suitable concentrations didn't have cytotoxicity on M146L cell survival. Protocatechuic acid at the concentration of 0.25 mmol/L, 0.5 mmol/L and 1.0 mmol/L significantly inhibited the mRNA expression of APP. CONCLUSION: The study suggests that the suitable dose of protocatechuic acid could inhibit the mRNA expression of APP in M146L cell,and its mechanism needs further study.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Araceae/química , Hidroxibenzoatos/farmacología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/efectos de los fármacos , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cricetinae , Relación Dosis-Respuesta a Droga , Hidroxibenzoatos/administración & dosificación , Presenilina-1/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección
15.
Medicine (Baltimore) ; 101(34): e30296, 2022 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-36042656

RESUMEN

This study aimed to in the management of Kasabach-Merritt phenomenon (KMP), a severe thrombocytopenic coagulopathy that occurs in the presence of an enlarging vascular tumor. Here, we retrospectively evaluated 12 patients with KMP in Guangzhou Women and Children's Medical Center, Guangzhou Medical University, from 2017 to 2021. 12 patients, including 7 females and 5 males, were identified. Tumors were located in the leg (n = 4), neck (n = 1), face (n = 3), chest wall (n = 1), back (n = 2), and retroperitoneum (n = 1). A plaque-like lesion with ecchymosis was the most common cutaneous manifestation. All the patients underwent embolization therapy. Nine patients received steroid treatment and 7 patients were administered with sirolimus. The mean duration of treatment was 1.6 months. All the patients reported in this study were alive when discharged. Embolization combined with steroid and sirolimus appears effective in patients with KMP, as well as in those who experienced disease recurrence. However, a long-term follow-up of the children cured of KMP will be necessary to monitor its recurrence and improve the outcome.


Asunto(s)
Hemangioendotelioma , Síndrome de Kasabach-Merritt , Sarcoma de Kaposi , Niño , Terapia Combinada , Femenino , Humanos , Lactante , Síndrome de Kasabach-Merritt/tratamiento farmacológico , Síndrome de Kasabach-Merritt/patología , Masculino , Recurrencia Local de Neoplasia , Estudios Retrospectivos , Sirolimus
16.
Zhong Yao Cai ; 34(4): 572-5, 2011 Apr.
Artículo en Zh | MEDLINE | ID: mdl-21809545

RESUMEN

OBJECTIVE: To explore the effect and mechanism of ferulic acid on differentiation in bFGF-treated PC12 cells. METHODS: The length of neurite outgrowth and the percentage of PC12 cells induced in the presence of 0 ng/mL or 1 ng/mL bFGF were assayed. RESULTS: Compared with that of control group,ferulic acid could enhance the differentiation effect of bFGF (1 ng/mL) in PC12 cells (P < 0.01) and the enhancing effect could be blocked by the specific MAPK kinase inhibitor, PD98059. CONCLUSION: Ferulic acid potentiates neurite outgrowth in bFGF-treated PC12 cells by MAPK-dependent signaling pathway.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Ácidos Cumáricos/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Flavonoides/farmacología , Neuritas/efectos de los fármacos , Animales , Ácidos Cumáricos/administración & dosificación , Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Neuritas/fisiología , Células PC12/efectos de los fármacos , Plantas Medicinales/química , Ratas , Transducción de Señal/efectos de los fármacos
17.
Zhong Yao Cai ; 34(4): 567-72, 2011 Apr.
Artículo en Zh | MEDLINE | ID: mdl-21809544

RESUMEN

OBJECTIVE: To study the neurotrophic effects of protocatechuic acid on neurite outgrowth and survival in cultured cerebral cortical neurons. METHODS: The newborn rat cerebral cortex neurons were cultured in vitro. The convert phase microscope was used to count the survival neurons with neurites and measure the average length of neurites. MTT and LDH assay were carried out to investigate the effect of PCA on the neuronal viability. RESULTS: Compared with control group, different concentration of PCA could increase the number of survival neurons with neurites and the average length of neurites. MTT and LDH assay showed that PCA promoted neuron survival in a dose-dependent manner. CONCLUSION: PCA can enhance the survival of rat cortical neurons with neurites and promote the neurite outgrowth of rat cortical neurons.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Corteza Cerebral/citología , Hidroxibenzoatos/farmacología , Neuritas/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Animales Recién Nacidos , Recuento de Células , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/embriología , Relación Dosis-Respuesta a Droga , Femenino , Hidroxibenzoatos/administración & dosificación , Lactato Deshidrogenasas/metabolismo , Masculino , Neuritas/fisiología , Neuronas/citología , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley
18.
J Agric Food Chem ; 69(42): 12456-12464, 2021 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-34647728

RESUMEN

Neural stem cell (NSC) differentiation and proliferation are important biological processes in the cerebral neural network. However, these two abilities of NSCs are limited. Thus, the induction of differentiation and/or proliferation through the administration of plant-derived small-molecule compounds could be used to repair damaged neural networks. The present study reported that gallic acid (GA), an important phenolic acid found in tea, selectively caused NSCs to differentiate into immature neurons and promoted NSC proliferation by activating the mitogen-activated protein kinase/extracellular-regulated kinase (MAPK/ERK) pathway. In addition, it was found that 3,4-dihydroxybenzoic acid was the main active structure exhibiting neurotrophic activity. The substitution of the carboxyl group on the benzene ring with the ester group may promote differentiation based on the structure of 3,4-dihydroxybenzoic acid. Furthermore, the introduction of the 5-hydroxyl group may promote proliferation. The present study identified that GA can promote the differentiation and proliferation of NSCs in vitro and exert pharmacological activity on NSCs.


Asunto(s)
Quinasas de Proteína Quinasa Activadas por Mitógenos , Células-Madre Neurales , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Ácido Gálico/farmacología , Sistema de Señalización de MAP Quinasas , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Ratas
19.
Ann Med ; 53(1): 1461-1469, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34435521

RESUMEN

BACKGROUND: Acute lymphoblastic leukaemia (ALL) is often characterized by broad clinical and biological heterogeneity, as well as recurrent genetic aberrations. Despite remarkable improvements in the treatment outcome in paediatric ALL over the past several decades, it remains a leading cause of morbidity and mortality among children. Cytokines have been extensively studied in haematologic diseases; however, the mechanisms by which cytokines contribute to ALL pathogenesis remain poorly understood. METHODS: IL-33 levels were measured by enzyme-linked immunosorbent assay (ELISA). IL1RL1 expression on ALL cell surface was accessed by flow cytometry. Expression of phosphorylated p38 MAPK, p38, pAKT, AKT and GAPDH were quantified by western blot. Cell survival signals were evaluated by apoptosis using flow cytometry. RESULTS: BM samples from ALL patients at diagnosis upregulated their cell surface expression of IL1RL1, and a higher interleukin (IL)-33 level in the serum was observed as compared to the healthy individuals. Moreover, exogenous IL-33 treatment significantly inhibited apoptosis by activating p38 mitogen-activated protein kinase (MAPK) and AKT pathway, while the inhibitor for p38 MAPK, SB203580, counteracted IL-33-induced anti-apoptosis via inactivation of p38 MAPK and AKT. Furthermore, IL-33 negatively regulates cyclin B1 protein level while increasing the expression of CDK1, with SB203580 inhibiting the effect. CONCLUSION: Our study reveals an important role for IL-33/IL1RL1 axis in supporting ALL which may represent a novel treatment for paediatric patients.KEY MESSAGESBoth IL-33 and IL1RL1 levels are upregulated in primary ALL samples.IL-33 increased both p38 MAPK and AKT activation in ALL.IL-33 promotes survival and cell cycle progression of ALL cells via activating p38 MAPK.


Asunto(s)
Apoptosis/efectos de los fármacos , Inflamación/metabolismo , Interleucina-33/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Proteínas Proto-Oncogénicas c-akt , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Niño , Ensayo de Inmunoadsorción Enzimática , Humanos , Interleucina-33/sangre , Recurrencia Local de Neoplasia , Fosforilación , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo
20.
PLoS Negl Trop Dis ; 15(8): e0009633, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34347790

RESUMEN

Dengue fever virus (DENV) is a global health threat that is becoming increasingly critical. However, the pathogenesis of dengue has not yet been fully elucidated. In this study, we employed bioinformatics analysis to identify potential biomarkers related to dengue fever and clarify their underlying mechanisms. The results showed that there were 668, 1901, and 8283 differentially expressed genes between the dengue-infected samples and normal samples in the GSE28405, GSE38246, and GSE51808 datasets, respectively. Through overlapping, a total of 69 differentially expressed genes (DEGs) were identified, of which 51 were upregulated and 18 were downregulated. We identified twelve hub genes, including MX1, IFI44L, IFI44, IFI27, ISG15, STAT1, IFI35, OAS3, OAS2, OAS1, IFI6, and USP18. Except for IFI44 and STAT1, the others were statistically significant after validation. We predicted the related microRNAs (miRNAs) of these 12 target genes through the database miRTarBase, and finally obtained one important miRNA: has-mir-146a-5p. In addition, gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were carried out, and a protein-protein interaction (PPI) network was constructed to gain insight into the actions of DEGs. In conclusion, our study displayed the effectiveness of bioinformatics analysis methods in screening potential pathogenic genes in dengue fever and their underlying mechanisms. Further, we successfully predicted IFI44L and IFI6, as potential biomarkers with DENV infection, providing promising targets for the treatment of dengue fever to a certain extent.


Asunto(s)
Biología Computacional , Dengue/genética , Biomarcadores , Redes Reguladoras de Genes , Humanos , Mapas de Interacción de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA