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

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Electrophoresis ; 45(9-10): 885-896, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38356010

RESUMEN

Nanopore sequencing technology has broad application prospects in forensic medicine due to its small size, portability, fast speed, real-time result analysis capabilities, single-molecule sequencing abilities, and simple operation. Here, we demonstrate for the first time that nanopore sequencing platforms can be used to identify individuals in the field. Through scientific and reasonable design, a nanopore MinION MK1B device and other auxiliary devices are integrated into a portable detection box conducive to individual identification at the accident site. Individual identification of 12 samples could be completed within approximately 24 h by jointly detecting 23 short tandem repeat (STR) loci. Through double-blinded experiments, the genotypes of 49 samples were successfully determined, and the accuracy of the STR genotyping was verified by the gold standard. Specifically, the typing success rate for 1150 genotypes was 95.3%, and the accuracy rate was 86.87%. Although this study focused primarily on demonstrating the feasibility of full-process testing, it can be optimistically predicted that further improvements in bioinformatics workflows and nanopore sequencing technology will help enhance the feasibility of Oxford Nanopore Technologies equipment for real-time individual identification at accident sites.


Asunto(s)
Repeticiones de Microsatélite , Secuenciación de Nanoporos , Humanos , Repeticiones de Microsatélite/genética , Secuenciación de Nanoporos/métodos , Genética Forense/métodos , Proyectos Piloto , Reproducibilidad de los Resultados , Genotipo , Análisis de Secuencia de ADN/métodos , Dermatoglifia del ADN/métodos , Diseño de Equipo
2.
Zhongguo Zhong Yao Za Zhi ; 49(10): 2745-2753, 2024 May.
Artículo en Zh | MEDLINE | ID: mdl-38812175

RESUMEN

This study investigated the protective effect of ginsenoside Rg_1(GRg_1) on oxygen and glucose deprivation/reoxygenation(OGD/R)-injured rat adrenal pheochromocytoma(PC12) cells and whether the underlying mechanism was related to the regulation of inositol-requiring enzyme 1(IRE1)-c-Jun N-terminal kinase(JNK)-C/EBP homologous protein(CHOP) signaling pathway. An OGD/R model was established in PC12 cells, and PC12 cells were randomly classified into control, model, OGD/R+GRg_1(0.1, 1, 10 µmol·L~(-1)), OGD/R+GRg_1+rapamycin(autophagy agonist), OGD/R+GRg_1+3-methyladenine(3-MA,autophagy inhibitor), OGD/R+GRg_1+tunicamycin(endoplasmic reticulum stress agonist), OGD/R+GRg_1+4-phenylbutyric acid(4-PBA, endoplasmic reticulum stress inhibitor), and OGD/R+GRg_1+3,5-dibromosalicylaldehyde(DBSA, IRE1 inhibitor) groups. Except the control group, the other groups were subjected to OGD/R treatment, i.e., oxygen and glucose deprivation for 6 h followed by reoxygenation for 6 h. Cell viability was detected by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl tetrazolium bromide(MTT) assay. Apoptosis was detected by Hoechst 33342 staining, and the fluorescence intensity of autophagosomes by the monodansylcadaverine(MDC) assay. Western blot was employed to determine the expression of autophagy-related proteins(Beclin1, LC3-Ⅱ, and p62) and the pathway-related proteins [IRE1, p-IRE1, JNK, p-JNK, glucose-regulated protein 78(GRP78), and CHOP]. The results showed that GRg_1 dose-dependently increased the viability of PC12 cells and down-regulated the expression of Beclin1, LC3-Ⅱ, p-IRE1, p-JNK, GRP78, and CHOP, compared with the model group. Furthermore, GRg_1 decreased the apoptosis rate and MDC fluorescence intensity and up-regulated the expression of p62 protein. Compared with the OGD/R+GRg_1(10 µmol·L~(-1)) group, OGD/R+GRg_1+rapamycin and OGD/R+GRg_1+tunicamycin groups showed increased apoptosis rate and MDC fluorescence intensity, up-regulated protein levels of Beclin1, LC3-Ⅱ, p-IRE1, p-JNK, GRP78, and CHOP, decreased relative cell survival rate, and down-regulated protein level of p62. The 3-MA, 4-PBA, and DBSA groups exerted the opposite effects. Taken together, GRg_1 may ameliorate OGD/R-induced PC12 cell injury by inhibiting autophagy via the IRE1-JNK-CHOP pathway.


Asunto(s)
Apoptosis , Ginsenósidos , Glucosa , Proteínas Serina-Treonina Quinasas , Factor de Transcripción CHOP , Animales , Ratas , Células PC12 , Factor de Transcripción CHOP/metabolismo , Factor de Transcripción CHOP/genética , Glucosa/metabolismo , Ginsenósidos/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Apoptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Autofagia/efectos de los fármacos , Endorribonucleasas/metabolismo , Endorribonucleasas/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Oxígeno/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Complejos Multienzimáticos
3.
Neuropathol Appl Neurobiol ; 49(5): e12934, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37705167

RESUMEN

BACKGROUND AND PURPOSE: Hyperphosphorylation of Tau is one of the important pathological features of Alzheimer's disease (AD). Therefore, studying the mechanisms behind Tau hyperphosphorylation is crucial in exploring the pathogenesis of neurological damage in AD. METHODS: In this study, after the establishment of rat models of AD, quantitative phosphoproteomics and proteomics were performed to identify proteins, showing that phosphorylation of microtubule associated protein 1A (MAP 1A) was lower in the model group. Western blot confirmed the changes of MAP 1A in the SD rats, APP/PS1 transgenic mice and cell AD models. To further study the molecular mechanism of recombinant MAP 1A phosphorylation affecting Tau phosphorylation, interfering siRNA-MAP 1A and protein immunoprecipitation reaction analysis were performed in AD cell models. RESULTS: Cyclin-dependent kinase 5 (CDK5) showed reduced binding to MAP 1A and increased binding to Tau, resulting in a decrease in phosphorylated MAP 1A (p-MAP 1A) and an increase in phosphorylated Tau (p-Tau), and MAP 1A silencing promoted binding of CDK5-Tau and increased Tau phosphorylation, thereby reducing the cell survival rate. CONCLUSIONS: In summary, we found that p-MAP 1A downregulation associated with p-Tau upregulation was due to their altered binding forces to CDK5, and MAP 1A could enhance autophosphorylation by competitive binding to CDK5 and antagonise Tau phosphorylation. This leads to neuronal protection and reducing tissue damage levels in AD, which can help better understand the mechanisms of AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer , Animales , Ratones , Ratas , Enfermedad de Alzheimer/metabolismo , Ratones Transgénicos , Fosforilación , Ratas Sprague-Dawley , Proteínas tau/metabolismo , Regulación hacia Arriba
4.
Zhongguo Zhong Yao Za Zhi ; 47(19): 5274-5283, 2022 Oct.
Artículo en Zh | MEDLINE | ID: mdl-36472034

RESUMEN

To investigate the protective effect of Tongqiao Huoxue Decoction containing cerebrospinal fluid(TQHXD-CSF) on HT22 cells damaged by oxygen-glucose deprivation/reoxygenation(OGD/R) and whether the mechanism is related to the regulation of ASK1/MKK4/JNK signaling pathway. HT22 cells were subjected to OGD/R to simulate cerebral ischemia-reperfusion injury(CIRI). Then the cells were randomly divided into five groups: blank cerebrospinal fluid(control group), OGD/R group, TQHXD-CSF group, Z-VAD-FMK group(20 µmol·L~(-1)) and TQHXD-CSF+Z-VAD-FMK group. Except the control group, cells in the other groups were reoxygenated for 12 h after 6 h of oxygen and glucose deprivation for modeling OGD/R, and group administration was performed. Cell viability and cytotoxicity were detected by CCK8 and LDH assay kit, respectively and the morphology of HT22 cells was observed by inverted microscope. Western blot and qRT-PCR were employed to detect the protein and mRNA expression levels of Bax, Bcl-2 and caspase-3, respectively. Then HT22 cells were assigned into the control group, OGD/R group, si-NC group, si-ASK1 group, TQHXD-CSF group and TQHXD-CSF+si-ASK1 group. Cell viability, proliferation and apoptosis were determined by CCK8, electric cell-substrate impedance sensing(ECIS), and Hoechst staining and flow cytometry, respectively. The protein expression of MKK4, p-MKK4, JNK, p-JNK, c-Jun, p-c-Jun, Cyt C, Bax, Bcl-2 and caspase-3 was tested by Western blot. The results showed that compared with OGD/R group, TQHXD-CSF significantly enhanced cell viability, improved cell morphology and reduced the protein and mRNA expression levels of Bax, Bcl-2 and caspase-3. In addition, when ASK1 was silenced, compared with OGD/R group, TQHXD-CSF remarkably improved cell viability, and decreased apoptosis rate and the protein expression levels of p-MKK4, p-JNK, p-c-Jun, Cyt C, Bax/Bcl-2 and caspase-3, but the effect was not as good as that of TQHXD-CSF+si-ASK1 group. In conclusion, TQHXD-CSF can inhibit apoptosis mediated by ASK1/MKK4/JNK signaling pathway in OGD/R-damaged HT22 cells, and has protective effect on ischemia-reperfusion injury.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Daño por Reperfusión , Humanos , Apoptosis , Proteína X Asociada a bcl-2/metabolismo , Caspasa 3/metabolismo , Glucosa , Oxígeno/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Daño por Reperfusión/metabolismo , ARN Mensajero/metabolismo
5.
J Cardiovasc Pharmacol ; 73(5): 316-325, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30855407

RESUMEN

Chuanxiong rhizome has been widely used for the treatment of cerebral vascular disease in traditional Chinese medicine. The integrity of blood-brain barrier (BBB) is closely linked to the cerebral vascular disease. The protective effects of ligustilide, the major bioactive component in Chuanxiong rhizome, on cerebral blood vessels have been reported previously, but its effects and potential mechanism on BBB have not been entirely clarified. In the current work, the effects of ligustilide on BBB permeability and the underlying molecular mechanisms had been investigated using the model of BBB established by coculturing astrocytes and brain microvascular endothelial cells isolated from the rat brain. The ischemia-damaged model of BBB has been established with oxygen and glucose deprivation (OGD). Our results indicated that OGD significantly increased the permeability in the coculture BBB model. This OGD-induced increase in permeability could suppress by ligustilide in a concentration-dependent manner. Also, ligustilide promoted both gene and protein expression of tight junction proteins. Ligustilide suppressed the upregulation of HIF-1α, vascular endothelial growth factor, and AQP-4 in the BBB model induced by OGD. Collectively, all results have demonstrated that ligustilide is capable of reducing the permeability of BBB in vitro model induced by OGD through HIF-1α/vascular endothelial growth factor pathway and AQP-4, which provide a new target for the clinical application of ligustilide on BBB after stroke in future.


Asunto(s)
4-Butirolactona/análogos & derivados , Astrocitos/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Glucosa/deficiencia , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , 4-Butirolactona/farmacología , Animales , Acuaporina 4/metabolismo , Astrocitos/metabolismo , Astrocitos/patología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Hipoxia de la Célula , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Ratas Sprague-Dawley , Transducción de Señal , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
6.
Molecules ; 24(19)2019 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-31597329

RESUMEN

The neuroprotective role of schizandrin (SA) in cerebral ischemia-reperfusion (I/R) was recently highlighted. However, whether SA plays a regulatory role on autophagy in cerebral I/R injury is still unclear. This study aimed to explore whether the neuroprotective mechanisms of SA were linked to its regulation of AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR)/autophagy pathway in vivo and in vitro. The present study confirmed that SA significantly improved oxygen-glucose deprivation/re-oxygenation (OGD/R)-induced PC12 cells injury. The results of immunoblotting and confocal microscope showed that SA decreased autophagy in OGD/R-injured PC12 cells, which was reflected by the decreased Beclin-1 and LC3-II expression, autophagy flux level, and LC3 puncta formation. In addition, the autophagy inducer rapamycin partially prevented the effects of SA on cell viability and autophagy after OGD/R, whereas the autophagy inhibitor 3-methyladenine (3-MA) exerted the opposite effect. The results of Western blotting showed that SA markedly decreased the phosphorylation of AMPK (p-AMPK), whereas the phosphor-mTOR (p-mTOR) levels increased in the presence of OGD/R insult. Furthermore, pretreatment with the AMPK inducer AICAR partially reversed the protective effects and autophagy inhibition of SA. However, AMPK inhibitor Compound C pretreatment further promoted the inhibition of SA on autophagy induction and cell damage induced by OGD/R. Taken together, these findings demonstrate that SA protects against OGD/R insult by inhibiting autophagy through the regulation of the AMPK-mTOR pathway and that SA may have therapeutic value for protecting neurons from cerebral ischemia.


Asunto(s)
Autofagia/efectos de los fármacos , Ciclooctanos/farmacología , Glucosa/metabolismo , Lignanos/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Oxidación-Reducción , Oxígeno/metabolismo , Compuestos Policíclicos/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Ciclooctanos/química , Modelos Animales de Enfermedad , Lignanos/química , Ratones , Modelos Biológicos , Estructura Molecular , Fármacos Neuroprotectores/química , Compuestos Policíclicos/química , Ratas , Daño por Reperfusión/etiología , Daño por Reperfusión/metabolismo , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
7.
Cell Mol Neurobiol ; 37(4): 619-633, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27380043

RESUMEN

Tetramethylpyrazine (TMP, also known as Ligustrazine), which is isolated from Chinese Herb Medicine Ligustium wollichii Franchat (Chuan Xiong), has been widely used in China for the treatment of ischemic stroke by Chinese herbalists. Brain microvascular endothelial cells (BMECs) are the integral parts of the blood-brain barrier (BBB), protecting BMECs against oxygen-glucose deprivation (OGD) which is important for the treatment of ischemic stroke. Here, we investigated the protective mechanisms of TMP, focusing on OGD-injured BMECs and the Rho/Rho-kinase (Rho-associated kinases, ROCK) signaling pathway. The model of OGD-injured BMECs was established in this study. BMECs were identified by von Willebrand factor III staining and exposed to fasudil, or TMP at different concentrations (14.3, 28.6, 57.3 µM) for 2 h before 24 h of OGD injury. The effect of each treatment was examined by cell viability assays, measurement of intracellular reactive oxygen species (ROS), and transendothelial electric resistance and western blot analysis (caspase-3, endothelial nitric oxide synthase (eNOS), RhoA, Rac1). Our results show that TMP significantly attenuated apoptosis and the permeability of BMECs induced by OGD. In addition, TMP could notably down-regulate the characteristic proteins in Rho/ROCK signaling pathway such as RhoA and Rac1, which triggered abnormal changes of eNOS and ROS, respectively. Altogether, our results show that TMP has a strong protective effect against OGD-induced BMECs injury and suggest that the mechanism might be related to the inhibition of the Rho/ROCK signaling pathway.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Células Endoteliales/metabolismo , Microvasos/efectos de los fármacos , Pirazinas/farmacología , Transducción de Señal/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Glucosa/metabolismo , Masculino , Microvasos/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Oxígeno/metabolismo , Sustancias Protectoras/farmacología , Ratas Sprague-Dawley
8.
Acta Pharmacol Sin ; 37(9): 1218-28, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27498779

RESUMEN

AIM: Sweroside is an iridoid glycoside with diverse biological activities. In the present study we investigated the effects of sweroside on α-naphthylisothiocyanate (ANIT)-induced cholestatic liver injury in mice. METHODS: Mice received sweroside (120 mg·kg(-1)·d(-1), ig) or a positive control INT-747 (12 mg·kg(-1)·d(-1), ig) for 5 d, and ANIT (75 mg/kg, ig) was administered on d 3. The mice were euthanized on d 5, and serum biochemical markers, hepatic bile acids and histological changes were analyzed. Hepatic expression of genes related to pro-inflammatory mediators and bile acid metabolism was also assessed. Primary mouse hepatocytes were exposed to a reconstituted mixture of hepatic bile acids, which were markedly elevated in the ANIT-treated mice, and the cell viability and expression of genes related to pro-inflammatory mediators were examined. RESULTS: Administration of sweroside or INT-747 effectively ameliorated ANIT-induced cholestatic liver injury in mice, as evidenced by significantly reduced serum biochemical markers and attenuated pathological changes in liver tissues. Furthermore, administration of sweroside or INT-747 significantly decreased ANIT-induced elevation of individual hepatic bile acids, such as ß-MCA, CA, and TCA, which were related to its effects on the expression of genes responsible for bile acid synthesis and transport as well as pro-inflammatory responses. Treatment of mouse hepatocytes with the reconstituted bile acid mixture induced significant pro-inflammatory responses without affecting the cell viability. CONCLUSION: Sweroside attenuates ANIT-induced cholestatic liver injury in mice by restoring bile acid synthesis and transport to their normal levels, as well as suppressing pro-inflammatory responses.


Asunto(s)
1-Naftilisotiocianato/toxicidad , Antiinflamatorios/uso terapéutico , Ácidos y Sales Biliares/biosíntesis , Colestasis/tratamiento farmacológico , Glucósidos Iridoides/uso terapéutico , Hígado/efectos de los fármacos , Animales , Antiinflamatorios/administración & dosificación , Ácidos y Sales Biliares/farmacología , Biomarcadores/sangre , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Colestasis/inducido químicamente , Colestasis/inmunología , Colestasis/metabolismo , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/inmunología , Factores Inmunológicos/genética , Glucósidos Iridoides/administración & dosificación , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Cultivo Primario de Células
9.
Int J Mol Sci ; 17(9)2016 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-27589720

RESUMEN

Ruscogenin, an important steroid sapogenin derived from Ophiopogon japonicus, has been shown to inhibit cerebral ischemic injury. However, its potential molecular action on blood-brain barrier (BBB) dysfunction after stroke remains unclear. This study aimed to investigate the effects of ruscogenin on BBB dysfunction and the underlying mechanisms in middle cerebral artery occlusion/reperfusion (MCAO/R)-injured mice and oxygen-glucose deprivation/reoxygenation (OGD/R)-injured mouse brain microvascular endothelial cells (bEnd.3). The results demonstrated that administration of ruscogenin (10 mg/kg) decreased the brain infarction and edema, improved neurological deficits, increased cerebral brain flow (CBF), ameliorated histopathological damage, reduced evans blue (EB) leakage and upregulated the expression of tight junctions (TJs) in MCAO/R-injured mice. Meanwhile, ruscogenin (0.1-10 µM) treatment increased cell viability and trans-endothelial electrical resistance (TEER) value, decreased sodium fluorescein leakage, and modulated the TJs expression in OGD/R-induced bEnd.3 cells. Moreover, ruscogenin also inhibited the expression of interleukin-1ß (IL-1ß) and caspase-1, and markedly suppressed the expression of Nucleotide-binding domain (NOD)-like receptor family, pyrin domain containing 3 (NLRP3) and thiredoxin-interactive protein (TXNIP) in vivo and in vitro. Furthermore, ruscogenin decreased reactive oxygen species (ROS) generation and inhibited the mitogen-activated protein kinase (MAPK) pathway in OGD/R-induced bEnd.3 cells. Our findings provide some new insights into its potential application for the prevention and treatment of ischemic stroke.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Inflamasomas/metabolismo , Sistema de Señalización de MAP Quinasas , Espirostanos/farmacología , Animales , Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Barrera Hematoencefálica/metabolismo , Proteínas Portadoras/metabolismo , Caspasa 1/genética , Caspasa 1/metabolismo , Hipoxia de la Célula , Línea Celular , Infarto de la Arteria Cerebral Media/metabolismo , Inflamasomas/efectos de los fármacos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ophiopogon/química , Especies Reactivas de Oxígeno/metabolismo , Espirostanos/uso terapéutico , Tiorredoxinas/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo
10.
Cell Mol Neurobiol ; 35(8): 1105-15, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25976179

RESUMEN

Muscone is the main chemical ingredient in Musk which is main crude drug in Tongqiaohuoxue decoction (TQHXD), and TQHXD has a protective effect on damaged neurons, so we hypothesize that muscone can alter blood-brain barrier (BBB) permeability via the modulation of P-glycoprotein (P-gp) and matrix metalloproteinase-9 (MMP-9) expression. In this study, astrocytes (AC) and human umbilical vein endothelial cells (ECV304) were co-cultured to simulate the BBB model in vitro. Leak testing, transmembrane resistance experiments, and BBB-specific enzyme testing were used to test whether the model was successful. Different concentrations of muscone permeating the BBB were detected by gas chromatography (GC). The change of the transendothelial electrical resistance (TEER) on the BBB in vitro after treating with muscone was detected by Millicell-ERS. The protein expression of P-gp, MMP-9 in normal, and oxygen/glucose deprivation (OGD) BBB model was determined by western blotting to inquire that the mechanism of muscone penetrates the BBB model in vitro. The results show that muscone was detected in the lower medium of the BBB model by GC; the values of TEER were no significant difference before and after muscone (8 µM) was added to the BBB model; the expression of P-gp significantly decreased after the BBB model treatment with muscone (4, 8, and 16 µM) for 24 h; the expression of P-gp and MMP-9 in different concentrations of muscone groups had different degrees of reduction compared with the BBB in the state of OGD. In conclusion, muscone could permeate the BBB model, and it was associated with the inhibition of P-gp and MMP-9 expression. An understanding of the mechanisms of muscone across the BBB is crucial to the development of therapeutic modalities for cerebral vascular diseases.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Barrera Hematoencefálica/metabolismo , Cicloparafinas/metabolismo , Cicloparafinas/farmacología , Metaloproteinasa 9 de la Matriz/biosíntesis , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Ratas , Ratas Sprague-Dawley
11.
Yao Xue Xue Bao ; 50(2): 162-8, 2015 Feb.
Artículo en Zh | MEDLINE | ID: mdl-25975022

RESUMEN

To investigate the neuroprotective of ligustilide (LIG) against glutamate-induced apoptosis of PC12 cells, cell viability were examined by MTT assay. Flow cytometry was applied to assay cell apoptosis rate. Intracellular calcium concentration was measured by using fluorescent dye Fluo-3/AM. Cytochrome C (Cyt C), Caspase-3, Bax and Bcl-2 protein expression were assayed by western blot. The results showed that glutamate is cytotoxic with an inhibitory concentration 50 (ID50) of 15 mmol · L(-1). Pretreatment with LIG (1, 5, 15 µmol · L(-1)) significantly improved cell viability. The apoptosis rate in glutamate-induced PC12 cells was 13.39%, and decreased in the presence of LIG (1, 5, 15 µmol · L(-1)) by 9.06%, 6.48%, 3.82%, separately. Extracellular accumulation of Ca2+ induced by glutamate were significantly reduced by LIG. The results of western blot manifested that pretreatment LIG could decrease the release of Cyt C from mitochondria, down-regulate Caspase-3 protein expression and up-regulate Bcl-2/Bax ratio, thereby protects PC12 cells from apoptosis. In summary, LIG had protective effect on glutamate-induced apoptosis in PC12 cells through attenuating the increase in intracellular Ca2+ concentration, and inhibiting the release of Cyt C from mitochondria to cytoplasm.


Asunto(s)
4-Butirolactona/análogos & derivados , Apoptosis/efectos de los fármacos , 4-Butirolactona/farmacología , Compuestos de Anilina , Animales , Proteínas Reguladoras de la Apoptosis , Calcio/metabolismo , Caspasa 3/metabolismo , Supervivencia Celular , Citocromos c/metabolismo , Ácido Glutámico/efectos adversos , Mitocondrias/metabolismo , Células PC12/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Xantenos , Proteína X Asociada a bcl-2/metabolismo
12.
Wei Sheng Wu Xue Bao ; 52(1): 96-103, 2012 Jan.
Artículo en Zh | MEDLINE | ID: mdl-22489466

RESUMEN

OBJECTIVE: To provide effective microorganisms for the treatment of water polluted by microcystins (MCs), the strains capable of biodegrading microcystin LR (MC-LR) were isolated from the sediment of Chaohu Lake. The degradation of microcystin-LR by the intracellular extracts of the strains were studied. METHODS: The enrichment culture using MC-LR as the sole carbon source was utilized to isolate the microcystin-degrading strains. The isolated strain was identified according to the observation of morphology, the physiological and chemical tests and the analysis of 16S rRNA gene sequences. The degradation of MC-LR by the intracellular extracts was studied. RESULTS: Strain M6 effectively degrading MC-LR was isolated and the strain M6 could grow utilizing MC-LR as the sole carbon source. Phylogenetic analysis based on 16S rRNA gene showed the similarity of 99% between strain M6 and Bacillus cereus. The experimental results suggest that the active substances for degrading MC-LR were the intracellular extracts which were the tissue enzymes of cells rather than induced enzymes. The degradation of MC-LR may be due to the catalytic effects of three enzymes. The degradation rate of 98.7% could be reached under the following conditions: pH 8.0, 404.9 mg/L of intracellular extracts, and 10 mg/L of the initial concentration of MC-LR. CONCLUSION: Strain M6 which could biodegrade MC-LR efficiently was identified as Bacillus cereus. The influences of pH, the concentration of intracellular extracts and the initial concentration of MC-LR on the enzymatic reaction were obvious.


Asunto(s)
Bacillus cereus/metabolismo , Microcistinas/metabolismo , Concentración de Iones de Hidrógeno , Toxinas Marinas
13.
Phytomedicine ; 101: 154111, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35512628

RESUMEN

BACKGROUND: Mitophagy plays a critical role in cerebral ischemia/reperfusion by timely removal of dysfunctional mitochondria. In mammals, PINK1/Parkin is the most classic pathway mediating mitophagy. And the activation of PINK1/Parkin mediated mitophagy exerts neuroprotective effects during cerebral ischemia reperfusion injury (CIRI). Ligustilide (LIG) is a natural compound extracted from ligusticum chuanxiong hort and angelica sinensis (Oliv.) diels that exerts neuroprotective activity after cerebral ischemia reperfusion injury (CIRI). However, it still remains unclear whether LIG could attenuates cerebral ischemia reperfusion injury (CIRI) through regulating mitophagy mediated by PINK1/Parkin. PURPOSE: To explore the underlying mechanism of LIG on PINK1/Parkin mediated mitophagy in the hippocampus induced by ischemia reperfusion. METHODS: This research used the middle cerebral artery occlusion and reperfusion (MCAO/R) animal model and oxygen-glucose deprivation and reperfusion (OGD/R) as in vitro model. Neurological behavior score, 2, 3, 5-triphenyl tetrazolium chloride (TTC) staining and Hematoxylin and Eosin (HE) Staining were used to detect the neuroprotection of LIG in MCAO/R rats. Also, the levels of ROS, mitochondrial membrane potential (MMP) and activities of Na+-K+-ATPase were detected to reflect mitochondrial function. Moreover, transmission electron microscope (TEM) and fluorescence microscope were used to observe mitophagy and the western blot was performed to explore the changes in protein expression in PINK1/Parkin mediated mitophagy. Finally, exact mechanism between neuroprotection of LIG and mitophagy mediated by PINK1/Parkin was explored by cell transfection. RESULTS: The results show that LIG improved mitochondrial functions by mitophagy enhancement in vivo and vitro to alleviate CIRI. Whereas, mitophagy enhanced by LIG under CIRI is abolished by PINK1 deficiency and midivi-1, a mitochondrial division inhibitor which has been reported to have the function of mitophagy, which could further aggravate the ischemia-induced brain damage, mitochondrial dysfunction and neuronal injury. CONCLUSION: LIG could ameliorate the neuronal injury against ischemia stroke by promoting mitophagy via PINK1/Parkin. Targeting PINK1/Parkin mediated mitophagy with LIG treatment might be a promising therapeutic strategy for ischemia stroke.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Daño por Reperfusión , 4-Butirolactona/análogos & derivados , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Hipocampo/metabolismo , Infarto de la Arteria Cerebral Media , Mamíferos/metabolismo , Mitofagia , Proteínas Quinasas/metabolismo , Ratas , Reperfusión , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
14.
J Pharm Pharmacol ; 74(1): 32-40, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34791341

RESUMEN

OBJECTIVES: Chrysophanol (CHR), also well-known as Rhei radix et rhizome, is a crucial component in traditional Chinese medicine. It has been widely studied as a potential treatment for many diseases due to its anti-inflammatory effects. However, there are very few studies to establish the potential therapeutic effect of CHR in cell and animal models of Alzheimer's disease (AD). Therefore, we aim to investigate whether CHR could be used as a potential therapeutic approach to patients with AD and further disclose the underlying mechanism. Increasing studies have shown that endoplasmic reticulum (ER) calcium (Ca2+) homeostasis emerges as a central player in AD pathogenesis. Moreover, augmentation of ER stress (ERS) promotes neuronal apoptosis, and excessive oxidative stress is an inducer of ERS. Therefore, we believe that ERS-mediated apoptosis may be one of the causes of AD. METHODS: This study examined the neuroprotective effects of CHR on AD rats and AD cell models and explored its potential mechanism. KEY FINDINGS: CHR could reduce the damage of neurons. In AD cell models, CHR significantly inhibited Aß 25-35-induced neuronal damage, reduced the number of apoptotic cells and improved cell survival rate. Western blot showed that the expression of caspases 3, 9 and 12 was decreased after CHR treatment, and CHR also affected the ERS signalling pathway. In addition, the higher expression of pro-apoptotic proteins in the AD cell model was reduced after CHR treatment by inhibiting GRP78 signalling. Further studies have shown that overexpressed protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) inhibited the regulatory effect of CHR on PERK and weakened the neuroprotective effect of CHR on the AD cell model. CONCLUSIONS: This study revealed a novel mechanism through which CHR plays a neuroprotective role by regulating ERS when it comes to the therapy of AD.


Asunto(s)
Antraquinonas , Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Antraquinonas/metabolismo , Antraquinonas/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Ratas , Transducción de Señal/efectos de los fármacos , eIF-2 Quinasa/metabolismo
15.
Phytomedicine ; 95: 153884, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34929562

RESUMEN

BACKGROUND: Ischemic stroke is a major global cause of death and permanent disability. Studies have suggested that mitochondria play a critical role in maintaining cellular energy homeostasis and inevitably involved in neuronal damage during cerebral ischemic. Ligustilide is the main active ingredient of Angelica sinensis and Ligusticum chuanxiongs with neuroprotective activity. PURPOSE: These study sought to exlopre the role of LIG in improving mitochondrial function and the relationship between LIG induced mitochondrial fission and mitophagy in ischemic stroke. METHODS: Cerebral I/R injury was established by the model of Oxygen-glucose deprivation/reperfusion (OGD/R) in HT22 cells and middle cerebral artery occlusion (MCAO) in rats. Mitochondrial functions of were detected by flow cytometry and immunofluorescence, and mitochondrial fission were detected by western blots. Furthermore, we studied the role of AMPK pathway in the neuroprotective effect of LIG. RESULTS: LIG treatment significantly increased the MMP and ATP production, decreased the reactive oxygen species (ROS) generation and Ca2+ overload, and further induced mitochondrial fission and mitophagy. Moreover, we found that blocking mitochondrial fission by mdivi-1 resulted in accumulation of damaged mitochondria mainly through selectively blocking mitophagy, thereby inhibiting viability of HT-22 cells after OGD/R. Also, Drp-1 inhibitor mdivi-1 increased the infarct volume and aggravated the neurological deficits after MCAO operation in vivo. Additionally, LIG triggered AMP-activated protein kinase (AMPK) pathway. AMPKα2 knockdown attenuated LIG-induced mitochondrial fission through inhibiting the expression of Drp1 and Fis1, and led to nerve cell apoptosis. CONCLUSION: Our study indicate that LIG attenuated the injury of ischemic stroke by improving mitochondrial function and highlight the critical role of LIG in the regulation of LIG-induced mitochondrial fission and mitophagy via an AMPK-dependent manner. These findings indicate that LIG protects nerve damage against ischemic stroke by inducing Drp1-mediated mitochondrial fission via activation of AMPK signaling pathway in vivo and in vitro.


Asunto(s)
4-Butirolactona/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Dinámicas Mitocondriales , 4-Butirolactona/análogos & derivados , Animales , Apoptosis , Isquemia Encefálica/tratamiento farmacológico , Dinaminas , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Dinámicas Mitocondriales/efectos de los fármacos , Ratas
16.
J Ethnopharmacol ; 298: 115585, 2022 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-35921993

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Tong-Qiao-Huo-Xue Decoction (TQHXD) is a traditional classic Chinese Medicinal Formula (CMF) used for clinical treatment of ischemic stroke. TQHXD leads to improvement in the symptoms of the acute period of cerebral infarction and recovery period after stroke. Our previous studies also showed that TQHXD produced a significant protective effect on the brain after cerebral ischemia-reperfusion (I/R) injury. It is reported that autophagy is closely related to ischemic brain injury; however, the functional contribution of TQHXD to brain microvascular endothelial cell (BMEC) autophagy and its underlying mechanism remains unclear. AIM OF THE STUDY: The purpose of this study was to investigate the effects and mechanism of TQHXD in inhibiting cerebral ischemia-induced endothelial autophagy. MATERIALS AND METHODS: The high-performance liquid chromatography (HPLC) fingerprint of the chemical constituents from TQHXD was established for the quality control, and the Longa method was used to evaluate the efficacy of TQHXD in rats with middle cerebral artery occlusion (MCAO). The expression of LC3 was determined by immunofluorescence double staining. To evaluate the protective effects of TQHXD-containing cerebrospinal fluid (CSF) on BMECs injured by oxygen-glucose deprivation and reperfusion, cell survival rate was determined using the CCK-8 assay and cell apoptosis was determined by fluorescein isothiocyanate (FITC)-Annexin V/PI. Autophagy was detected using transmission electron microscopy. RESULTS: The results showed that TQHXD-CSF significantly ameliorated oxygen-glucose deprivation/reperfusion (OGD/R)-induced injury in BMECs. Confocal microscopy and Western blot results showed that TQHXD-CSF reduced autophagy-related protein expression and autophagosome number. The results of the western blotting indicated that TQHXD-CSF caused a marked increase in the phosphorylation of protein kinase B and phosphoinsotide-3 kinase (Akt/p-Akt and PI3K/p-PI3K, respectively) and their expression levels were down-regulated after treatment with pathway inhibitor, ZSTK474. Furthermore, in a MCAO model in rats, TQHXD markedly increased p-PI3K, p-Akt and p-mTOR, whereas the autophagy related proteins decreased. CONCLUSIONS: Taken together, these findings demonstrate that TQHXD protects against ischemic insult by inhibiting autophagy through the regulation of the PI3K/Akt/mammalian target of rapamycin (mTOR) pathway and that TQHXD may have therapeutic value for protecting BMECs from cerebral ischemia.


Asunto(s)
Isquemia Encefálica , Daño por Reperfusión , Animales , Autofagia , Isquemia Encefálica/tratamiento farmacológico , Medicamentos Herbarios Chinos , Glucosa/metabolismo , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Oxígeno/metabolismo , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Daño por Reperfusión/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
17.
Phytomedicine ; 106: 154437, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36099654

RESUMEN

BACKGROUND: Activation of blood stasis is a crucial aspect of stroke treatment, and the Tong-Qiao-Huo-Xue-Decoction (TQHXD) formula is commonly utilized for this purpose. However, the mechanism underlying the protective effects of TQHXD against cerebral ischemia-reperfusion (I/R) injury is unclear. PURPOSE: Identification of the TQHXD components responsible for its protective effects and determination of their mode of action against cerebral I/R injury. METHODS: Gas chromatography (GC) and high-performance liquid chromatography (HPLC) were carried out to determine the active aspects of TQHXD. The active components and targets of TQHXD were looked up in the TCMSP and HERB databases; the Genecards, OMIM, TTD, and DrugBank databases were used to identify targets related to cerebral infarction; and the intersecting targets were obtained. The drug-ingredient-target-disease network and PPI network were subsequently built using Cytoscape 3.7.1 and STRING websites. Autodock VINA was used to perform molecular docking between the core target ASK1 and the active components of TQHXD detected by HPLC and GC. After successfully creating a rat model of middle cerebral artery occlusion (MCAO), the therapeutic effect of TQHXD was observed using triphenyltetrazolium and hematoxylin-eosin staining. We used Tunel-NeuN staining and transmission electron microscopy (TEM) to quantify hippocampal apoptosis. RT-qPCR and western blotting were used to detect protein and mRNA expression, respectively. RESULTS: HPLC and GC identified six active ingredients. Network pharmacology analyses were performed to test 66 intersection targets, including ASK1, MKK4, and JNK. Ferulic acid, HSYA, ligustilide, paeoniflorin, and muscone all displayed high binding affinity with ASK1 in molecular docking studies. The neuroprotective effects of TQHXD in I/R rats were demonstrated in the experimental models. In comparison with the model group, TQHXD decreased the apoptosis rate and reduced the protein levels of p-ASK1, caspase 3, p-MKK4, CytC, p-c-Jun, Bax/Bcl-2, and p-JNK, while considerably increasing the mRNA levels of Bcl-2 and decreasing those of Bax. CONCLUSION: By controlling the ASK1/MKK4/JNK pathway, TQHXD protects neurons from I/R damage and prevents apoptosis. Thus, TQHXD may be effective for the treatment of ischemic stroke. And the mechanism behind these therapeutic actions of TQHXD is supported by this research.


Asunto(s)
Fármacos Neuroprotectores , Daño por Reperfusión , Animales , Apoptosis , Caspasa 3/metabolismo , Medicamentos Herbarios Chinos , Eosina Amarillenta-(YS)/farmacología , Eosina Amarillenta-(YS)/uso terapéutico , Hematoxilina/farmacología , Hematoxilina/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas , Simulación del Acoplamiento Molecular , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/metabolismo , Ratas , Daño por Reperfusión/tratamiento farmacológico , Proteína X Asociada a bcl-2/metabolismo
18.
Phytomedicine ; 95: 153882, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34968897

RESUMEN

BACKGROUND: YiQiFuMai lyophilized injection (YQFM) is derived from a traditional Chinese medicine prescription termed Shengmai San.YQFM is clinically applied to the treatment of cardiovascular and cerebrovascular diseases. It has been found that critical components of YQFM affect non-muscle myosin heavy chain IIA (NMMHC IIA), but its regulation in the excessive autophagy and the underlying mechanism has yet to be clarified. PURPOSE: To evaluate whether YQFM has neuroprotective effects on cerebral ischemia/reperfusion-induced injury by inhibiting NMMHC IIA-actin-ATG9A interaction for autophagosome formation. METHODS: The neuroprotective effects of YQFM were investigated in vivo in mice with middle cerebral artery occlusion/reperfusion (MCAO/R) (n = 6) by detecting neurological deficits, infarct volume, and histopathological changes. The NMMHC IIA-actin-ATG9A interaction was determined using immunofluorescence co-localization, co-immunoprecipitation, and proximity ligation assay. Rat pheochromocytoma (PC12) cells subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) were used to mimic neurons in in vitro experiments. RESULTS: In MCAO/R model mice, YQFM (1.342 g/kg) attenuated brain ischemia/reperfusion-induced injury by regulating NMMHC IIA-actin-mediated ATG9A trafficking. YQFM (400 µg/ml) also exerted similar effects on OGD/R-induced PC12 cells. Furthermore, RNAi of NMMHC IIA weakened the NMMHC IIA-F-actin-dependent ATG9A trafficking and, therefore, attenuated the neuroprotective activities of YQFM in vitro. CONCLUSION: These findings demonstrated that YQFM exerted neuroprotective effects by regulating the NMMHC IIA-actin-ATG9A interaction for autophagosome formation. This evidence sheds new light on the potential mechanism of YQFM in the treatment of cerebral ischemia/reperfusion.


Asunto(s)
Isquemia Encefálica , Medicamentos Herbarios Chinos , Fármacos Neuroprotectores , Daño por Reperfusión , Actinas , Animales , Autofagia , Proteínas Relacionadas con la Autofagia , Isquemia Encefálica/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Proteínas de la Membrana , Ratones , Fármacos Neuroprotectores/farmacología , Ratas , Daño por Reperfusión/tratamiento farmacológico , Proteínas de Transporte Vesicular
19.
Photodiagnosis Photodyn Ther ; 34: 102202, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33556618

RESUMEN

Keloids are characterized by abnormal proliferation of fibroblasts and continuous deposition of extracellular matrix (ECM) components. In the field of dermopathy, photodynamic therapy (PDT) with visible light has been increasingly investigated. The natural photosensitizer Hypocrellin A (HA) was shown to have excellent light induced anticancer, antimicrobial and antiviral activities. In this experiment, we investigated the impacts of HA united light-emitting diode (LED) red light irradiation on human keloid fibroblast cells (KFs). Our results showed that HA combined with red light irradiation treatment (HA-R-PDT) decreased KF viability, reduced KF collagen production and ECM accumulation, inhibited cell proliferation, suppressed cell invasion and induced cell apoptosis. Moreover, our observations demonstrated that the TGF-ß/Smad signalling pathway and autophagy were restrained by HA-R-PDT. TGF-ß1 could promote autophagy in KFs through both the Smad and ERK pathways, while inhibition of autophagy altered the TGF-ß1 levels through negative feedback. Therefore, HA-R-PDT suppressed cell hyperproliferation, collagen synthesis and ECM accumulation of KFs by regulating the TGF-ß1-ERK-autophagy-apoptosis signalling pathway. HA-R-PDT deserves systematic investigation as a potential therapeutic strategy for keloids, and autophagy might be a promising candidate in the treatment of KFs.


Asunto(s)
Queloide , Fotoquimioterapia , Apoptosis , Autofagia , Proliferación Celular , Células Cultivadas , Fibroblastos/patología , Humanos , Queloide/radioterapia , Luz , Perileno/análogos & derivados , Fenol , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Quinonas
20.
Cell Signal ; 69: 109550, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32007528

RESUMEN

Cutaneous squamous cell carcinoma (cSCC) is a type of malignant skin tumor derived from epidermal Malpighian cells. Photodynamic therapy is regarded as a crucial method in oncology. Hypocrellin A (HA), an efficient natural photosensitizer, has been reported to exert excellent light induced antiviral, antimicrobial and anticancer activity through mediating multiple signaling pathways. The purpose of the present study is to examine the effects of HA united red light irradiation on human squamous carcinoma A431 cells and further reveal the underlying regulatory mechanisms. The results showed that synergistic treatment of HA and red light irradiation inhibited cell proliferation and induced cell apoptosis and autophagy. Moreover, HA united red light irradiation caused a significant accumulation of reactive oxygen species (ROS), and induced the activation of c-Jun NH 2 terminal kinases (JNKs) which was inhibited by the antioxidant N-Acetyl-cysteine (NAC). Furthermore, HA united red light irradiation activated the nuclear factor-kappa B (NF-κB) pathway, and inhibition of NF-κB activity exacerbated HA united red light irradiation-induced apoptosis but suppressed cell autophagy. In addition, the inhibition of autophagy promoted HA united red light irradiation-induced apoptosis and facilitated the NF-κB activity. Over all, our results revealed that HA united red light irradiation could inhibit A431 cell proliferation by inducing apoptosis and autophagy via the activation of the ROS mediated JNK and NF-κB pathways, providing prospective for HA as a potential therapeutic for the treatment of cSCC.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Carcinoma de Células Escamosas/terapia , Perileno/análogos & derivados , Fenol/farmacología , Fármacos Fotosensibilizantes/farmacología , Quinonas/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , FN-kappa B/metabolismo , Perileno/farmacología , Fotoquimioterapia , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Cutáneas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA