Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
BMC Complement Med Ther ; 24(1): 162, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38632534

RESUMEN

The incidence of dementia is rising, with neuronal cell death from oxidative stress and apoptosis recognized as a significant contributor to its development. However, effective strategies to combat this condition are lacking, necessitating further investigation. This study aimed to assess the potential of an anthocyanin-rich extract from Zea mays L. var. ceratina (AZC) in alleviating neuronal cell death.Neurotoxicity was induced in SH-SY5Y cells using hydrogen peroxide (H2O2) at a concentration of 200 µM. Cells were pretreated with varying doses (31.25 and 62.5 µg/mL) of AZC. Cell viability was assessed using the MTT assay, and molecular mechanisms including reactive oxygen species (ROS) levels, antioxidant enzyme activities (catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px)), malondialdehyde (MDA) levels for oxidative stress, and the activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2), cAMP response element-binding protein (CREB), and apoptotic factors (B-cell lymphoma 2 (Bcl-2), caspase 3) were explored.Results showed that AZC significantly improved cell viability, reduced ROS production and MDA levels, and downregulated caspase 3 expression. It enhanced CAT, SOD, and GSH-Px activities, activated ERK1/2 and CREB, and upregulated Bcl-2 expression. These findings support the neuroprotective effects of AZC, suggesting it activates ERK1/2, leading to CREB activation and subsequent upregulation of Bcl-2 expression while suppressing caspase 3. AZC may mitigate neuronal cell death by reducing ROS levels through enhanced scavenging enzyme activities.In conclusion, this study underscores the potential of AZC as a neuroprotective agent against neuronal cell death. However, further investigations including toxicity assessments, in vivo studies, and clinical trials are necessary to validate its benefits in neuroprotection.


Asunto(s)
Neuroblastoma , Fármacos Neuroprotectores , Humanos , Animales , Abejas , Peróxido de Hidrógeno/farmacología , Especies Reactivas de Oxígeno/metabolismo , Caspasa 3/metabolismo , Antocianinas , Zea mays/metabolismo , Línea Celular Tumoral , Muerte Celular , Antioxidantes/farmacología , Fármacos Neuroprotectores/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Superóxido Dismutasa/metabolismo
2.
BMC Complement Med Ther ; 23(1): 117, 2023 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-37055744

RESUMEN

The prevalence of dementia is increasing, and most of the causes are related to neuronal cell death. Unfortunately, no effective strategy is available for protecting against this condition. Based on the use of the synergistic concept together with the positive modulation effect of both mulberry fruit and mulberry leaf on dementia, we hypothesized that the combined extract of mulberry fruit and mulberry leaf (MFML) should mitigate neuronal cell death. Neuronal cell damage was induced in SH-SY5Y cells by exposure to hydrogen peroxide at a dose of 200 µM. SH-SY5Y cells were given MFML at doses of 62.5 and 125 µg/mL before induced cytotoxicity. Then, the cell viability was determined via MTT assay, and the possible underlying mechanisms were investigated via the alterations of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), malondialdehyde (MDA), nuclear factor-κB (NF-κB), and tumor necrosis factor-alpha (TNF-α), together with apoptotic factors including (B-cell lymphoma 2) BCL2, Casapase-3 and Caspase-9. The results showed that MFML significantly enhanced cell viability. It also significantly decreased MDA level, NF-κB, TNF-α, Casapase-3, Caspase-9, but increased SOD, GSH-Px and BCL2. These data demonstrated the neuroprotective effect of MFML. The possible underlying mechanisms might occur partly via the improvement of the inappropriate apoptotic mechanisms via BCL2, Casapase-3 and Caspase-9 together with the decrease in neurodegeneration induced by the reduction of inflammation and oxidative stress. In conclusion, MFML is a potential neuroprotectant candidate against neuronal cell injury. However, toxicity, animal studies, and clinical trials are essential to confirm these benefits.


Asunto(s)
Demencia , Morus , Neuroblastoma , Fármacos Neuroprotectores , Animales , Humanos , Fármacos Neuroprotectores/farmacología , Caspasa 9/metabolismo , Apoptosis , Peróxido de Hidrógeno/toxicidad , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Frutas , Línea Celular Tumoral , Caspasa 3/metabolismo , Neuroblastoma/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Superóxido Dismutasa/metabolismo , Hojas de la Planta
3.
Mol Med Rep ; 20(2): 1933-1942, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31257498

RESUMEN

It has been reported that amyloid ß peptide, the major component of senile plaques, serves a critical role in the development and progression of Alzheimer's disease (AD) by generating reactive oxygen species (ROS), leading to oxidative stress. The aim of the present study was to investigate the protective effect of Oroxylum indicum (L.) extract against Aß25­35­induced oxidative stress and cell injury using SH­SY5Y cells as a model, and at exploring the underlying mechanisms. The results revealed that the exposure of cells to 20 µM Aß25­35 significantly increased cellular oxidative stress, as evidenced by the increased ROS levels. Aß25­35 treatment also increased caspase­3/7 activity and lactate dehydrogenase (LDH) release, and caused viability loss. Oroxylum indicum treatment not only attenuated the generation of ROS and suppressed caspase­3/7 activity but also reduced the neurotoxicity of Aß25­35 in a concentration­dependent manner, as evidenced by the increased cell viability and decreased LDH release. Treatment with Oroxylum indicum also increased superoxide dismutase (SOD) and catalase (CAT) activity, increased the phosphorylation of Akt and cAMP­responsive element binding protein (CREB), and contributed to the upregulation of Bcl­2 protein. In combination, these results indicated that Oroxylum indicum extract could protect SH­SY5Y cells against Aß25­35­induced cell injury, at least partly, by inhibiting oxidative stress, increasing SOD and CAT activity, attenuating caspase 3/7 activity and promoting the cell survival pathway, Akt/CREB/Bcl­2. The approach used in the present study may also be useful for preventing the neurotoxicity induced by Aß in AD and related neurodegenerative diseases. Further studies investigating the activity of Oroxylum indicum extract in vivo are now required.


Asunto(s)
Bignoniaceae/química , Neuroblastoma/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Apoptosis/efectos de los fármacos , Catalasa/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Neuroblastoma/patología , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacología , Extractos Vegetales/química , Proteínas Proto-Oncogénicas c-akt/genética , Especies Reactivas de Oxígeno , Superóxido Dismutasa/genética
4.
Biomed Rep ; 8(4): 359-364, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29541456

RESUMEN

Iron accumulation in activated microglia has been consistently reported in neurodegenerative diseases. Previous results suggest that these cells facilitate neuroinflammation leading to neuronal cell death. Therefore, chemical compounds that alleviate the activation of iron-rich microglia may result in neuroprotection. In the present study, the effect of valproic acid (VPA) on microglial activation under iron-rich conditions was investigated. BV-2 microglial cells were exposed to lipopolysaccharide (LPS; 1 µg/ml) and iron (300 µg/ml) with or without VPA (1.6 mM). The results demonstrated that VPA attenuated the activation of iron-rich BV2 cells induced by LPS by down-regulating the mRNA expression of inducible nitric oxide (NO) synthase and interleukin 1ß (IL-1ß; P<0.01), to ultimately reduce the production of NO and IL-1ß (P<0.01). These events were accompanied by an attenuation in the nuclear translocation of nuclear factor-κB p65 subunit (P<0.01). These findings suggest that VPA may be therapeutically useful for attenuating the activation of iron-rich microglia.

5.
Biomed Rep ; 6(4): 441-448, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28413643

RESUMEN

The aim of the present study was to investigate the molecular mechanisms underlying Cratoxylum formosum (CF) Dyer-induced cancer cell death and antimigratory effects in HepG2 liver cancer cells. The cytotoxic, antiproliferative and antimigratory effects of CF leaf extract on human liver cancer HepG2 cell lines were evaluated using sulforhodamine B, colony formation, and wound healing assays. In addition, apoptosis induction mechanisms were investigated via reactive oxygen species (ROS) formation, caspase 3 activities, and mitochondrial membrane potential (ΔΨm) disruption. Gene expression and apoptosis-associated protein levels were measured by reverse transcription-quantitative polymerase chain reaction and western blotting. CF induced HepG2 cell death in a time- and dose-dependent manner with half maximal inhibitory concentration values of 219.03±9.96 and 124.90±6.86 µg/ml at 24 and 48 h, respectively. Treatment with CF caused a significant and dose-dependent decrease in colony forming ability and cell migration. Furthermore, the present study demonstrated that CF induced ROS formation, increased caspase 3 activities, decreased the ΔΨm, and caused HepG2 apoptosis. CF marginally decreased the expression level of the cell cycle regulatory protein, ras-related C3 botulinum toxin substrate 1 (rho family, small GTP binding protein Rac1) and the downstream protein, cyclin dependent kinase 6. Additionally, CF significantly enhanced p21 levels, reduced cyclin D1 protein levels and triggered cancer cell death. CF leaf extracts induced cell death, stimulated apoptosis and inhibited migration in HepG2 cells. Thus, CF may be useful for developing an anticancer drug candidate for the treatment of liver cancer.

6.
Biomed Pharmacother ; 90: 77-84, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28343074

RESUMEN

In this study we investigated how Cratoxy formosum (CF) leaf extract affects the viability and migration of human breast cancer cells including the mechanism(s) responsible. Our results showed that CF leaf extract strongly induced MCF-7 cell death in a concentration- and time-dependent manner, with IC50 values of 85.70±4.52µg/mL and 53.74±3.02µg/mL at 24h and 48h, respectively. Additionally, CF leaf extract potentiated the activity of 4 anticancer drugs with the greatest synergy occurring between CF and 5-FU. CF leaf extract also caused a dose-dependent decrease in colony forming ability with IC50 values of 36.37+1.80 µg/mL and cell migration, with IC50 values of 43.68±0.86µg/mL. Moreover, CF significantly induced ROS formation, increased caspase 3 activities, and reduced the mitochondrial membrane potential, leading to cancer cell apoptosis and cell death. In addition, the extract inhibited cancer cell migration at 25µg/mL by reducing MMP 2 and MMP 9 protein expression. Moreover, CF leaf extracts strongly decreased expression of the cell cycle regulatory protein Rac1 and downstream protein, cdk6. CF leaf extract significantly stimulated p21 and this correlated with a reduction in cyclin D1 protein levels. In summary, CF leaf extract can inhibit cell proliferation, induce cell apoptosis, and reduce cell migration in the MCF-7 cell line. It could also be beneficial for enhancing the activity of anticancer drugs used to treat breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Extractos Vegetales/farmacología , Hojas de la Planta/química , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Caspasa 3/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Ciclina D1/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Fluorouracilo/farmacología , Humanos , Células MCF-7 , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos
7.
Neurosci Lett ; 603: 6-11, 2015 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-26170247

RESUMEN

It has been reported that persons carrying the H63D variant in their hemochromatosis (HFE) gene are at increased risk of Alzheimer's disease (AD). We investigated the possibility that okra (Abelmoschus esculentus) and quercetin could mitigate this risk factor by examining its effect on AD-associated cellular events in HFE stably expressing SH-SY5Y cells. Treatment of H63D HFE cells either with okra or quercetin significantly decreased reactive oxygen species (ROS), hydrogen peroxide (H2O2), and protein oxidation compared to untreated cells. The levels of tau phosphorylation at serine-199, serine-202, and serine-396 sites were also significantly decreased when cells were treated with okra. Exposure of the H63D and wild type (WT) cells to iron increased tau phosphorylation, but this response was decreased significantly when cells were treated with okra. The mechanism responsible for these changes appears to be related to decreased glycogen synthase kinase (GSK)-3ß activity, an upstream signaling kinase of tau phosphorylation. We also established that okra treatment dramatically decreases intracellular iron levels in H63D cells compared to untreated cells. Our results provide important in vitro data on the effects of okra on various AD-associated cellular processes in H63D variant HFE cells. These results suggest okra may be beneficial in people expressing the H63D variant to reduce the risk of AD and other neurodegenerative diseases related to oxidative stress. Further in vivo studies would help confirm this.


Asunto(s)
Abelmoschus/química , Enfermedad de Alzheimer/metabolismo , Antioxidantes/farmacología , Antígenos de Histocompatibilidad Clase I/metabolismo , Proteínas de la Membrana/metabolismo , Extractos Vegetales/farmacología , Enfermedad de Alzheimer/genética , Línea Celular Tumoral , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Proteína de la Hemocromatosis , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Peróxido de Hidrógeno/metabolismo , Espacio Intracelular/metabolismo , Hierro/metabolismo , Proteínas de la Membrana/genética , Mutación , Neuroblastoma , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Carbonilación Proteica , Quercetina/análogos & derivados , Quercetina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Proteínas tau/metabolismo
8.
Neurosci Lett ; 500(1): 36-40, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21683124

RESUMEN

Activation of microglia could be beneficial and yet simultaneously harmful depending upon nature of pathological milieu. Regardless of disease-specific etiology, iron accumulation, particularly in activated microglia, is a notable feature associated with a series of neuropathologies, including Alzheimer's diseases. Although mounting evidence supports the role of iron in oxidative brain injury, knowledge on its regulatory role in neuroinflammation is still scarce. Here, we hypothesize that cellular iron status may be involved in determining the roles of activated microglia in neuroinflammatory processes. In this study, we examined effects of iron on expression of MMPs known to be involved in nervous system inflammation and degeneration using rat microglial cell line (HAPI). Stimulation experiments were performed using lipopolysaccharide (LPS). We demonstrated by RT-PCR that increased cellular iron levels enhanced the expression of MMP-9 in activated microglia, but had no effect on MMP-1. Studies using western blot and gelatin zymography analyses demonstrated that increased cellular iron levels in activated microglia enhanced the secretion of MMP-9 and MMP-1. Taken together, these results demonstrated regulatory roles of iron in the expression of MMPs by activated microglia at the transcription and translation levels. Using a colorimetric NBT reduction assay, we showed that increased cellular iron levels impaired zymosan phagocytic activity in activated microglia. Thus, these findings further our understanding toward the consequences of iron accumulation by activated microglia in neurodegeneration and suggest a possible link between iron metabolism in activated microglia and neuroinflammation.


Asunto(s)
Hierro/metabolismo , Metaloproteinasas de la Matriz/biosíntesis , Microglía/metabolismo , Fagocitosis , Animales , Línea Celular , Medio de Cultivo Libre de Suero , Encefalitis/inmunología , Encefalitis/metabolismo , Compuestos Férricos/farmacología , Ferritinas/metabolismo , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Microglía/efectos de los fármacos , Compuestos de Amonio Cuaternario/farmacología , Ratas , Receptores de Transferrina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Neurobiol Aging ; 32(8): 1409-19, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19775775

RESUMEN

A number of genetic association studies have appeared that address HFE gene variants in neurodegenerative disorders. However, the cellular impact of HFE in the nervous system has received little attention. To begin to address the role of the HFE allelic variants on cellular events associated with neurodegeneration, we examined the hypothesis that HFE polymorphisms are associated with alterations in tau phosphorylation in a human neuroblastoma cell line (SH-SY5Y). The results show that in a cell culture model, the H63D allele is associated with increased tau phosphorylation. The mechanisms responsible for these changes appear related to increased glycogen synthase kinase (GSK)-3ß activity. GSK-3ß activity is up-regulated in the cells expressing H63D HFE and can be modified by the addition of iron or treatment with an iron chelator in SH-SY5Y cells expressing wild-type HFE. Oxidative stress, also associated with elevated cellular iron, is associated with increased tau phosphorylation at the same sites as seen in H63D cells and treatment with Trolox, an anti-oxidant, lowered tau phosphorylation. These results suggest H63D HFE increases tau phosphorylation via GSK-3ß activity and iron-mediated oxidative stress.


Asunto(s)
Alelos , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Serina/metabolismo , Proteínas tau/metabolismo , Antioxidantes/farmacología , Línea Celular Tumoral , Cromanos/farmacología , Variación Genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3/fisiología , Glucógeno Sintasa Quinasa 3 beta , Proteína de la Hemocromatosis , Humanos , Hierro/química , Hierro/fisiología , Neuroblastoma , Enfermedades Neurodegenerativas/genética , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Fosforilación/efectos de los fármacos , Fosforilación/genética
10.
J Neurosci Res ; 88(14): 3079-89, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20734416

RESUMEN

Numerous epidemiological studies suggest that the expression of the HFE allelic variant H63D may be a risk factor or genetic modifier for Alzheimer's disease (AD). The H63D variant alters cellular iron homeostasis and increases baseline oxidative stress. The elevated cellular stress milieu, we have proposed, may alter cellular responses to genetic and environmental determinants of AD. Accumulation of ß-amyloid peptides (Aß) is one of the most prominent pathogenic characteristics of AD. Several studies have demonstrated that Aß can induce neuronal cell death through apoptosis. In this study, we provide evidence that an Aß(25-35) fragment, which contains the cytotoxic sequence of the amyloid peptide, activates the intrinsic apoptotic pathway in SH-SY5Y human neuroblastoma cells expressing the HFE allelic variant H63D to a greater extent than in cells with wild-type (WT) HFE. Specifically, Aß(25-35) peptide exposure significantly induced Bax translocation from the cytosol to the mitochondria in H63D-expressing cells compared with WT cells. This translocation was associated with increased cytochrome c release from mitochondria and an increase in active caspase-9 and caspase-3 activity in H63D cells. Consequently, there is increased apoptosis in cells expressing the H63D variant as opposed to cells expressing WT HFE. We also found increased amyloid precursor protein (APP) and Aß(1-42) peptide in the mitochondrial compartment as well as increased mitochondrial stress in H63D-expressing cells compared with WT. These findings support our hypothesis that the presence of the HFE H63D allele enables factors that trigger neurodegenerative processes associated with AD and predisposes cells to cytotoxcity.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/toxicidad , Apoptosis/genética , Antígenos de Histocompatibilidad Clase I/genética , Proteínas de la Membrana/genética , Mitocondrias/metabolismo , Neuronas/metabolismo , Alelos , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/biosíntesis , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Línea Celular Tumoral , Predisposición Genética a la Enfermedad/genética , Variación Genética , Proteína de la Hemocromatosis , Humanos , Mitocondrias/patología , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/biosíntesis , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA