Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 929
Filtrar
Más filtros

Medicinas Complementárias
Tipo del documento
Intervalo de año de publicación
1.
J Mol Neurosci ; 74(2): 44, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38630337

RESUMEN

Plants are a valuable source of information for pharmacological research and new drug discovery. The present study aimed to evaluate the neuroprotective potential of the leaves of the medicinal plant Sterculia setigera. In vitro, the effect of Sterculia setigera leaves dry hydroethanolic extract (SSE) was tested on cultured cerebellar granule neurons (CGN) survival when exposed to hydrogen peroxide (H2O2) or 6-hydroxydopamine (6-OHDA), using the viability probe fluorescein diacetate (FDA), a lactate dehydrogenase (LDH) activity assay, an immunocytochemical staining against Gap 43, and the quantification of the expression of genes involved in apoptosis, necrosis, or oxidative stress. In vivo, the effect of intraperitoneal (ip) injection of SSE was assessed on the developing brain of 8-day-old Wistar rats exposed to ethanol neurotoxicity by measuring caspase-3 activity on cerebellum homogenates, the expression of some genes in tissue extracts, the thickness of cerebellar cortical layers and motor coordination. In vitro, SSE protected CGN against H2O2 and 6-OHDA-induced cell death at a dose of 10 µg/mL, inhibited the expression of genes Casp3 and Bad, and upregulated the expression of Cat and Gpx7. In vivo, SSE significantly blocked the deleterious effect of ethanol by reducing the activity of caspase-3, inhibiting the expression of Bax and Tp53, preventing the reduction of the thickness of the internal granule cell layer of the cerebellar cortex, and restoring motor functions. Sterculia setigera exerts neuroactive functions as claimed by traditional medicine and should be a good candidate for the development of a neuroprotective treatment against neurodegenerative diseases.


Asunto(s)
Muerte Celular , Etanol , Neuronas , Fármacos Neuroprotectores , Extractos Vegetales , Hojas de la Planta , Sterculia , Animales , Ratas , Caspasa 3/metabolismo , Etanol/administración & dosificación , Etanol/química , Etanol/toxicidad , Peróxido de Hidrógeno/toxicidad , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacología , Oxidopamina/toxicidad , Ratas Wistar , Sterculia/química , Hojas de la Planta/química , Plantas Medicinales/química , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neuronas/patología , Lactato Deshidrogenasas/metabolismo , Proteína GAP-43/análisis , Apoptosis/genética , Estrés Oxidativo/genética , Cerebelo/citología , Cerebelo/efectos de los fármacos , Cerebelo/patología , Cerebelo/fisiología , Masculino , Femenino , Células Cultivadas , Muerte Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Fitoquímicos/administración & dosificación , Fitoquímicos/análisis , Fitoquímicos/química , Fitoquímicos/farmacología , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Extractos Vegetales/farmacología , Antioxidantes/análisis , Antioxidantes/química , Antioxidantes/farmacología , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem , Cromatografía Líquida con Espectrometría de Masas , Metabolismo Secundario
2.
Int J Dev Neurosci ; 84(3): 251-261, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38469915

RESUMEN

OBJECTIVE: The aim of this study is to evaluate whether exogenous melatonin (MEL) mitigates the deleterious effects of high-dose caffeine (CAF) administration in pregnant rats upon the fetal hippocampus. MATERIALS AND METHODS: A total of 32 adult Wistar albino female rats were divided into four groups after conception (n = 8). At 9-20 days of pregnancy, intraperitoneal (i.p.) MEL was administered at a dose of 10 mg/kg/day in the MEL group, while i.p. CAF was administered at a dose of 60 mg/kg/day in the CAF group. In the CAF plus MEL group, i.p. CAF and MEL were administered at a dose of 60 and 10 mg/kg/day, respectively, at the same period. Following extraction of the brains of the fetuses sacrificed on the 21st day of pregnancy, their hippocampal regions were analyzed by hematoxylin and eosin and Cresyl Echt Violet, anti-GFAP, and antisynaptophysin staining methods. RESULTS: While there was a decrease in fetal and brain weights in the CAF group, it was found that the CAF plus MEL group had a closer weight average to that of the control group. Histologically, it was observed that the pyramidal cell layer consisted of 8-10 layers of cells due to the delay in migration in hippocampal neurons in the CAF group, while the MEL group showed similar characteristics with the control group. It was found that these findings decreased in the CAF plus MEL group. CONCLUSION: It is concluded that high-dose CAF administration causes a delay in neurogenesis of the fetal hippocampus, and exogenous MEL is able to mitigate its deleterious effects.


Asunto(s)
Cafeína , Hipocampo , Melatonina , Fármacos Neuroprotectores , Ratas Wistar , Animales , Femenino , Melatonina/farmacología , Melatonina/administración & dosificación , Hipocampo/efectos de los fármacos , Embarazo , Cafeína/administración & dosificación , Cafeína/farmacología , Ratas , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/administración & dosificación , Estimulantes del Sistema Nervioso Central/toxicidad , Estimulantes del Sistema Nervioso Central/administración & dosificación , Relación Dosis-Respuesta a Droga
3.
Naunyn Schmiedebergs Arch Pharmacol ; 395(3): 305-313, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35024909

RESUMEN

We investigated the effect of Pycnogenol as an antioxidant on improving motor function, depression, and the expression of NF-ƙB and Nrf2 genes in the experimental model of Parkinson's disease. Forty adult male NMRI mice weighing about 30 g were randomly divided into five groups of eight. Saline group: received 3 µl of saline, as 6-hydroxydopamine (6-OHDA) solvent, unilaterally in the left striatum, treatment groups: first received 3 µl 6-OHDA unilaterally inside the ipsilateral striatum and then divided into subgroup A: received distilled water, Pycnogenol solvent, by gavage for 7 days (lesion group), and subgroup B: received Pycnogenol at doses of 10, 20, and 30 mg/kg by gavage for 7 days. Seven days after Parkinson's model induction, the apomorphine test, the degree of catalepsy by bar test, the duration of immobility (depression) by forced swimming test (FST) were measured. In addition, the expression of NF-ƙB and Nrf2 genes was measured using the real-time PCR technique. The total number of rotations in the apomorphine test decreased significantly in the groups receiving Pycnogenol. Administration of Pycnogenol significantly reduced catalepsy. The study of depression in the group receiving Pycnogenol showed a significant reduction. Also, Pycnogenol increased the expression of the Nrf2 anti-inflammatory gene, but it had no significant difference in the expression of NF-ƙB gene. Pycnogenol, presumably with its antioxidative and genomic effects, improves the expression of the anti-inflammatory gene and found that neuroprotection effect in the brain.


Asunto(s)
Antioxidantes/farmacología , Flavonoides/farmacología , Trastornos Parkinsonianos/tratamiento farmacológico , Extractos Vegetales/farmacología , Animales , Antioxidantes/administración & dosificación , Apomorfina/administración & dosificación , Relación Dosis-Respuesta a Droga , Flavonoides/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Factor 2 Relacionado con NF-E2/genética , FN-kappa B/genética , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Oxidopamina , Trastornos Parkinsonianos/fisiopatología , Extractos Vegetales/administración & dosificación
4.
Brain Res Bull ; 180: 108-117, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35026347

RESUMEN

BACKGROUND: Metabolic syndrome patients are commonly prone to major health problems as cardiovascular diseases, diabetes mellitus, chronic kidney disease, cancer and neuropsychological complications including dementia. OBJECTIVES: This research investigates mechanisms linking metabolic syndrome to cognitive impairment and possible impact of vitamin D supplementation. METHODS: Forty male Wistar rats were divided into 4 groups. Control, metabolic syndrome (20% fructose solution in drinking water for 12 weeks, vitamin D supplemented (500 IU/kg/day)) and metabolic syndrome supplemented with vitamin D. Animals were assessed for spatial memory, hippocampal expression of SNAP 25, VAMP and mGlut2 receptor and hippocampus histological examination. Animals with metabolic syndrome showed prolonged acquisition and retention latencies in morris water maze, decreased hippocampal expression of SNAP 25 and VAMP and increased mGlut2 expression. Histologically CA1, CA3 regions and dentate nucleus revealed increase in degenerated neurons and glia cells with decreased pyramidal cell layer thickness. Vitamin D supplementation mitigated alterations induced by metabolic syndrome. CONCLUSIONS: Metabolic syndrome decreased hippocampal synaptic proteins and altered glutamatergic transmission and increased hippocampal neuronal degeneration. Vitamin D supplementation offered neuroprotective effects.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Hipocampo/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Síndrome Metabólico/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Receptores de Glutamato Metabotrópico/efectos de los fármacos , Vitamina D/farmacología , Animales , Conducta Animal/efectos de los fármacos , Disfunción Cognitiva/etiología , Modelos Animales de Enfermedad , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/etiología , Síndrome Metabólico/complicaciones , Fármacos Neuroprotectores/administración & dosificación , Ratas , Ratas Wistar , Vitamina D/administración & dosificación
5.
Ann Neurol ; 91(3): 389-403, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34979595

RESUMEN

OBJECTIVE: Cardiac arrest (CA) is a major health burden with brain damage being a significant contributor to mortality. We found lysophosphatidylcholine (LPC), including a species containing docosahexaenoic acid (LPC-DHA), was significantly decreased in plasma post-CA, supplementation of which significantly improved neurological outcomes. The aim of this study is to understand the protective role of LPC-DHA supplementation on the brain post-CA. METHODS: We first evaluated associations between the plasma level of LPC-DHA and neurological injury and outcomes of human patients with CA. We then utilized a rat CA model and cell cultures to investigate therapeutic and mechanistic aspects of plasma LPC-DHA supplementation. RESULTS: We found that decreased plasma LPC-DHA was strongly associated with neurological outcomes and disappearance of the difference between gray and white matter in the brain after CA in human patients. In rats, the decreased plasma LPC-DHA was associated with decreased levels of brain LPC-DHA after CA, and supplementing plasma LPC-DHA normalized brain levels of LPC-DHA and alleviated neuronal cell death, activation of astrocytes, and expression of various inflammatory and mitochondrial dynamics genes. We also observed deceased severity of metabolic alterations with LPC-DHA supplementation using untargeted metabolomics analysis. Furthermore, LPC treatment showed a similar protective effect for neurons and astrocytes in mixed primary brain cell cultures. INTERPRETATION: The observed neuroprotection accompanied with normalized brain LPC-DHA level by plasma supplementation implicate the importance of preventing the decrease of brain LPC-DHA post-CA for attenuating brain injury. Furthermore, the data supports the causative role of decreased plasma LPC-DHA for brain damage after CA. ANN NEUROL 2022;91:389-403.


Asunto(s)
Astrocitos/efectos de los fármacos , Lesiones Encefálicas/tratamiento farmacológico , Muerte Celular/efectos de los fármacos , Paro Cardíaco/complicaciones , Lisofosfatidilcolinas/administración & dosificación , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Animales , Encéfalo/efectos de los fármacos , Lesiones Encefálicas/sangre , Lesiones Encefálicas/etiología , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/administración & dosificación , Ácidos Docosahexaenoicos/sangre , Ácidos Docosahexaenoicos/uso terapéutico , Humanos , Lisofosfatidilcolinas/sangre , Lisofosfatidilcolinas/uso terapéutico , Masculino , Fármacos Neuroprotectores/uso terapéutico , Ratas , Ratas Sprague-Dawley
6.
Biomed Pharmacother ; 145: 112445, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34844103

RESUMEN

BACKGROUND AND AIM: Buyang Huanwu Decoction (BHD) is a traditional Chinese herbal medicine that is effective for treating cerebral ischemia (CI). However, the molecular mechanisms of BHD in CI have not been fully elucidated. In this study, we integrated the circular RNA (circRNA)-microRNA (miRNA)-messenger RNA (mRNA) network of middle cerebral artery occlusion (MACO) rats treated with BHD. METHODS: SD rats were randomly divided into a control group, model group, model+BHD group (2.5, 5, 10 g/kg) and model+butylphthalide (NBP) group (54 mg/kg). The neurological functions of the rats were evaluated by a modified neurological severity scoring (mNSS) system. Pathological lesions were assessed by Nissl staining, and the effects of BHD on neurovascular unit (NVU) associated protein microtubule-associated protein 2 (MAP2), glial fibrillary acidic protein (GFAP) and von Willebrand factor (VWF) were assessed by immunohistochemistry. CeRNA and miRNA microarrays were used to establish the circRNA, miRNA, and mRNA profiles. Finally, a circRNA-miRNA-mRNA ternary transcription network was constructed. RESULTS: BHD improved the neurobehavioral test scores (P < 0.01) and the histopathological changes in ischemic brain tissue in MCAO rats. The expression of MAP2 and VWF decreased and the expression of GFAP increased in the ischemic side brain tissue of MCAO rats (P < 0.01), and treatment with BHD reversed the above changes (P < 0.01 or 0.05). We identified seven, three, and 86 significantly dysregulated circRNAs, miRNAs, and mRNAs, respectively, that were associated with the neuroprotective effects of BHD. Furthermore, bioinformatics analysis showed that these targets may exert therapeutic effects through multiple pathways, such as the VEGF and Hippo signaling pathways. Finally, we constructed a circRNA-miRNA-mRNA network. CONCLUSIONS: In brief, our study provides novel insights into ceRNA-mediated gene regulation in the progression of NVU after CI and the mechanism of action for BHD.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Isquemia Encefálica/genética , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/administración & dosificación , Regulación de la Expresión Génica/genética , Infarto de la Arteria Cerebral Media , Masculino , MicroARNs/genética , Fármacos Neuroprotectores/administración & dosificación , ARN Circular/genética , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
7.
CNS Neurosci Ther ; 28(2): 247-258, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34837343

RESUMEN

BACKGROUND: Fisetin, the effective ingredient of the traditional Chinese medicine named Cotinus coggygria, is recommended to be active therapeutic in many disorders. However, its role in sepsis-associated encephalopathy (SAE) remains unclarified. METHODS: Cecal ligation and puncture (CLP) operation was performed to establish a rat model of SAE. Rats were grouped according to the surgery operation and fisetin administration. Cognitive impairment was assessed by Morris water maze test. Disruption of blood-brain barrier (BBB) integrity was detected by Evan's blue staining. The mitophagy, reactive oxygen species (ROS) generation, NLRP3 inflammasome activation, and pro-inflammatory cytokines levels were measured through western blot and double immunofluorescence labeling. A transmission electron microscope was applied for the observation of mitochondrial autophagosomes. RESULTS: Rats in the CLP group presented increased expression of IL-1R1, pNF-κB, TNF-α, and iNOS in microglial cells, indicating severe inflammation in the central nervous system (CNS). Nevertheless, there was no increase in BBB permeability. Meanwhile, NLRP3 inflammasome was activated in cerebral microvascular endothelial cells (CMECs), presented with an elevation of caspase-1 expression and IL-1ß secretion into CNS. In addition, we found fisetin significantly improved cognitive dysfunction in rats with SAE. Neuroprotective effects of fisetin might be associated with inhibition of neuroinflammation, represented with decreased expression of IL-1R1, pNF-κB, TNF-α, and iNOS in microglia. Furthermore, fisetin induced mitophagy, scavenged ROS, blocked NLRP3 inflammasome activation of CMECs, as evidenced by decreased expression of caspase-1 and reduced release of IL-1ß into CNS. CONCLUSION: Collectively, fisetin-blocked NLRP3 inflammasome activation via promoting mitophagy in CMECs may suppress the secretion of IL-1ß into CNS, reduce neuroinflammation, and contribute to the amelioration of cognitive impairment.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Flavonoles/farmacología , Mitofagia/efectos de los fármacos , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Encefalopatía Asociada a la Sepsis/complicaciones , Animales , Conducta Animal/efectos de los fármacos , Disfunción Cognitiva/etiología , Modelos Animales de Enfermedad , Flavonoles/administración & dosificación , Inflamasomas/efectos de los fármacos , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Ratas
8.
Braz. J. Pharm. Sci. (Online) ; 58: e20023, 2022. graf
Artículo en Inglés | LILACS | ID: biblio-1403706

RESUMEN

Abstract Caffeic acid is a phenolic compound widely distributed in plants and beverages such as coffee. Although its mechanism of action is poorly understood, caffeic acid reportedly induces antidepressant-like and neuroprotective effects. This study aimed to investigate the involvement of cellular signaling pathways in acute antidepressant-like effect induced by caffeic acid in mice. All procedures were approved by the Institutional Animal Ethics Committee of the UNIVALI n. 021/2013. Female Swiss mice were administered with vehicle, caffeic acid (5 mg/ kg, p.o.), inhibitor (H-89, U0126, chelerythrine, or PD9859, i.c.v.) or caffeic acid plus inhibitor. The behavioral effects were evaluated 1h after the administration of compounds to mice using tail suspension test (TST) and open field test (OFT). The results showed that the antidepressant- like effect of caffeic acid in mice was possibly mediated by the activation of PKA, MEK 1/2, PKC and MAPK (as assessed using TST), without compromising their locomotor activity (as assessed using OFT). Our results demonstrated, at least in part, the pathways involved in the neuroprotective and behavioral effects of caffeic acid.


Asunto(s)
Animales , Femenino , Ratones , Ácidos Cafeicos/análisis , Café/efectos adversos , Fármacos Neuroprotectores/administración & dosificación , Antidepresivos/efectos adversos , Plantas , Transducción de Señal , Quinasas de Proteína Quinasa Activadas por Mitógenos , Comités de Atención Animal/clasificación , Prueba de Campo Abierto
9.
Acta cir. bras ; 37(6): e370603, 2022. tab, graf, ilus
Artículo en Inglés | LILACS, VETINDEX | ID: biblio-1402964

RESUMEN

Purpose: To explore the neuroprotective effects of Lutongkeli (LTKL) in traumatic brain injury (TBI) and detect the related mechanism. Methods: TBI model was established with LTKL administration (2 and 4 g/kg/d, p.o.). Motor function of rats was examined by Rotarod test. Nissl staining was used to show neuron morphology. Furthermore, the disease-medicine common targets were obtained with the network pharmacology and analyzed with Kyoto Encyclopedia of Genes and Genomes. Lastly, the predicted targets were validated by real-time polymerase chain reaction. Results: After LTKL administration, neural behavior was significantly improved, and the number of spared neurons in brain was largely increased. Moreover, 68 bioactive compounds were identified, corresponding to 148 LTKL targets; 2,855 genes were closely associated with TBI, of which 87 overlapped with the LTKL targets and were considered to be therapeutically relevant. Functional enrichment analysis suggested LTKL exerted its pharmacological effects in TBI by modulating multiple pathways including apoptosis, inflammation, etc. Lastly, we found LTKL administration could increase the mRNA level of Bcl-2 and decrease the expression of Bax and caspase-3. Conclusions: This study reported the neuroprotective effect of LTKL against TBI is accompanied with anti-apoptosis mechanism, which provides a scientific explanation for the clinical application of LTKL in the treatment of TBI.


Asunto(s)
Animales , Masculino , Ratas , Apoptosis/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Lesiones Traumáticas del Encéfalo/terapia , Ratas Sprague-Dawley , Medicina Tradicional China
10.
Int J Mol Sci ; 22(23)2021 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-34884863

RESUMEN

The ability of NQO2 to increase the production of free radicals under enhanced generation of quinone derivatives of catecholamines is considered to be a component of neurodegenerative disease pathogenesis. The present study aimed to investigate the neuroprotective mechanisms of original NQO2 inhibitor M-11 (2-[2-(3-oxomorpholin-4-il)-ethylthio]-5-ethoxybenzimidazole hydrochloride) in a cellular damage model using NQO2 endogenous substrate adrenochrome (125 µM) and co-substrate BNAH (100 µM). The effects of M-11 (10-100 µM) on the reactive oxygen species (ROS) generation, apoptosis and lesion of nuclear DNA were evaluated using flow cytometry and single-cell gel electrophoresis assay (comet assay). Results were compared with S29434, the reference inhibitor of NQO2. It was found that treatment of HT-22 cells with M-11 results in a decline of ROS production triggered by incubation of cells with NQO2 substrate and co-substrate. Pre-incubation of HT-22 cells with compounds M-11 or S29434 results in a decrease of DNA damage and late apoptotic cell percentage reduction. The obtained results provide a rationale for further development of the M-11 compound as a potential neuroprotective agent.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Quinona Reductasas/antagonistas & inhibidores , Adrenocromo/metabolismo , Animales , Apoptosis/efectos de los fármacos , Bencimidazoles/química , Línea Celular , Daño del ADN , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/química , Hipocampo/citología , Masculino , Ratones Endogámicos ICR , Neuronas/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/química , Piridinas/farmacología , Alcaloides de Pirrolicidina/farmacología , Quinona Reductasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
11.
Int J Mol Sci ; 22(23)2021 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-34884629

RESUMEN

In recent years, much attention has been paid to the study of the therapeutic effect of the microelement selenium, its compounds, especially selenium nanoparticles, with a large number of works devoted to their anticancer effects. Studies proving the neuroprotective properties of selenium nanoparticles in various neurodegenerative diseases began to appear only in the last 5 years. Nevertheless, the mechanisms of the neuroprotective action of selenium nanoparticles under conditions of ischemia and reoxygenation remain unexplored, especially for intracellular Ca2+ signaling and neuroglial interactions. This work is devoted to the study of the cytoprotective mechanisms of selenium nanoparticles in the neuroglial networks of the cerebral cortex under conditions of ischemia/reoxygenation. It was shown for the first time that selenium nanoparticles dose-dependently induce the generation of Ca2+ signals selectively in astrocytes obtained from different parts of the brain. The generation of these Ca2+ signals by astrocytes occurs through the release of Ca2+ ions from the endoplasmic reticulum through the IP3 receptor upon activation of the phosphoinositide signaling pathway. An increase in the concentration of cytosolic Ca2+ in astrocytes leads to the opening of connexin Cx43 hemichannels and the release of ATP and lactate into the extracellular medium, which trigger paracrine activation of the astrocytic network through purinergic receptors. Incubation of cerebral cortex cells with selenium nanoparticles suppresses ischemia-induced increase in cytosolic Ca2+ and necrotic cell death. Activation of A2 reactive astrocytes exclusively after ischemia/reoxygenation, a decrease in the expression level of a number of proapoptotic and proinflammatory genes, an increase in lactate release by astrocytes, and suppression of the hyperexcitation of neuronal networks formed the basis of the cytoprotective effect of selenium nanoparticles in our studies.


Asunto(s)
Astrocitos/citología , Calcio/metabolismo , Gliosis/tratamiento farmacológico , Nanopartículas/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Daño por Reperfusión/prevención & control , Selenio/administración & dosificación , Animales , Antioxidantes/administración & dosificación , Antioxidantes/química , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Astrocitos/metabolismo , Señalización del Calcio , Gliosis/inmunología , Gliosis/metabolismo , Gliosis/patología , Nanopartículas/química , Neuronas/efectos de los fármacos , Neuronas/inmunología , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/química , Ratas , Daño por Reperfusión/etiología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Selenio/química
12.
Cell Mol Biol (Noisy-le-grand) ; 67(1): 101-105, 2021 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-34817361

RESUMEN

Parkinson's disease (PD) is one of the most prevalent neurodegenerative disorders worldwide. It is caused by the degeneration of dopaminergic neurons from the substantia nigra pars compacta. This neuronal loss causes the dopamine deficiency that leads to a series of functional changes within the basal ganglia, producing motor control abnormalities. L-DOPA is considered the gold standard for PD treatment, and it may alleviate its clinical manifestations for some time. However, its prolonged administration produces tolerance and several severe side effects, including dyskinesias and gastrointestinal disorders. Thus, there is an urgent need to find effective medications, and current trends have proposed some natural products as emerging options for this purpose. Concerning this, curcumin represents a promising bioactive compound with high therapeutic potential. Diverse studies in cellular and animal models have suggested that curcumin could be employed for the treatment of PD. Therefore, the objective of this narrative mini-review is to present an overview of the possible therapeutic effects of curcumin and the subjacent molecular mechanisms. Moreover, we describe several possible nanocarrier-based approaches to improve the bioavailability of curcumin and enhance its biological activity.


Asunto(s)
Encéfalo/efectos de los fármacos , Curcumina/administración & dosificación , Nanopartículas/administración & dosificación , Enfermedad de Parkinson/tratamiento farmacológico , Animales , Disponibilidad Biológica , Encéfalo/metabolismo , Curcumina/química , Curcumina/farmacocinética , Liberación de Fármacos , Glutatión Peroxidasa/metabolismo , Humanos , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Nanopartículas/química , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacocinética , Enfermedad de Parkinson/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Resultado del Tratamiento , Regulación hacia Arriba/efectos de los fármacos
13.
Nutrients ; 13(11)2021 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-34836132

RESUMEN

Injuries to the developing brain due to hypoxia-ischemia (HI) are common causes of neurological disabilities in preterm babies. HI, with oxygen deprivation to the brain or reduced cerebral blood perfusion due to birth asphyxia, often leads to severe brain damage and sequelae. Injury mechanisms include glutamate excitotoxicity, oxidative stress, blood-brain barrier dysfunction, and exacerbated inflammation. Nutritional intervention is emerging as a therapeutic alternative to prevent and rescue brain from HI injury. Lactoferrin (Lf) is an iron-binding protein present in saliva, tears, and breast milk, which has been shown to have antioxidant, anti-inflammatory and anti-apoptotic properties when administered to mothers as a dietary supplement during pregnancy and/or lactation in preclinical studies of developmental brain injuries. However, despite Lf's promising neuroprotective effects, there is no established dose. Here, we tested three different doses of dietary maternal Lf supplementation using the postnatal day 3 HI model and evaluated the acute neurochemical damage profile using 1H Magnetic Resonance Spectroscopy (MRS) and long-term microstructure alterations using advanced diffusion imaging (DTI/NODDI) allied to protein expression and histological analysis. Pregnant Wistar rats were fed either control diet or bovine Lf supplemented chow at 0.1, 1, or 10 g/kg/body weight concentration from the last day of pregnancy (embryonic day 21-E21) to weaning. At postnatal day 3 (P3), pups from both sexes had their right common carotid artery permanently occluded and were exposed to 6% oxygen for 30 min. Sham rats had the incision but neither surgery nor hypoxia episode. At P4, MRS was performed on a 9.4 T scanner to obtain the neurochemical profile in the cortex. At P4 and P25, histological analysis and protein expression were assessed in the cortex and hippocampus. Brain volumes and ex vivo microstructural analysis using DTI/NODDI parameters were performed at P25. Acute metabolic disturbance induced in cortical tissue by HIP3 was reversed with all three doses of Lf. However, data obtained from MRS show that Lf neuroprotective effects were modulated by the dose. Through western blotting analysis, we observed that HI pups supplemented with Lf at 0.1 and 1 g/kg were able to counteract glutamatergic excitotoxicity and prevent metabolic failure. When 10 g/kg was administered, we observed reduced brain volumes, increased astrogliosis, and hypomyelination, pointing to detrimental effects of high Lf dose. In conclusion, Lf supplementation attenuates, in a dose-dependent manner, the acute and long-term cerebral injury caused by HI. Lf reached its optimal effects at a dose of 1 g/kg, which pinpoints the need to better understand effects of Lf, the pathways involved and possible harmful effects. These new data reinforce our knowledge regarding neuroprotection in developmental brain injury using Lf through lactation and provide new insights into lactoferrin's neuroprotection capacities and limitation for immature brains.


Asunto(s)
Lesiones Encefálicas/prevención & control , Suplementos Dietéticos , Hipoxia-Isquemia Encefálica/terapia , Lactoferrina/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Animales , Animales Recién Nacidos , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Lesiones Encefálicas/etiología , Relación Dosis-Respuesta a Droga , Femenino , Hipoxia-Isquemia Encefálica/complicaciones , Lactancia , Masculino , Neuroprotección/efectos de los fármacos , Embarazo , Ratas , Ratas Wistar
14.
ACS Appl Mater Interfaces ; 13(39): 46406-46420, 2021 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-34569225

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disease associated with amyloid-ß (Aß) deposition, leading to neurotoxicity (oxidative stress and neuroinflammation) and gut microbiota imbalance. Resveratrol (Res) has neuroprotective properties, but its bioavailability in vivo is very low. Herein, we developed a small Res-selenium-peptide nanocomposite to enable the application of Res for eliminating Aß aggregate-induced neurotoxicity and mitigating gut microbiota disorder in aluminum chloride (AlCl3) and d-galactose(d-gal)-induced AD model mice. Res functional selenium nanoparticles (Res@SeNPs) (8 ± 0.34 nm) were prepared first, after which the surface of Res@SeNPs was decorated with a blood-brain barrier transport peptide (TGN peptide) to generate Res-selenium-peptide nanocomposites (TGN-Res@SeNPs) (14 ± 0.12 nm). Oral administration of TGN-Res@SeNPs improves cognitive disorder through (1) interacting with Aß and decreasing Aß aggregation, effectively inhibiting Aß deposition in the hippocampus; (2) decreasing Aß-induced reactive oxygen species (ROS) and increasing activity of antioxidation enzymes in PC12 cells and in vivo; (3) down-regulating Aß-induced neuroinflammation via the nuclear factor kappa B/mitogen-activated protein kinase/Akt signal pathway in BV-2 cells and in vivo; and (4) alleviating gut microbiota disorder, particularly with respect to oxidative stress and inflammatory-related bacteria such as Alistipes, Helicobacter, Rikenella, Desulfovibrio, and Faecalibaculum. Thus, we anticipate that Res-selenium-peptide nanocomposites will offer a new potential strategy for the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Portadores de Fármacos/química , Nanocompuestos/química , Fármacos Neuroprotectores/uso terapéutico , Resveratrol/uso terapéutico , Administración Oral , Cloruro de Aluminio , Enfermedad de Alzheimer/inducido químicamente , Péptidos beta-Amiloides/metabolismo , Animales , Bacterias/efectos de los fármacos , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/toxicidad , Galactosa , Microbioma Gastrointestinal/efectos de los fármacos , Proteínas Inmovilizadas/administración & dosificación , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/toxicidad , Masculino , Memoria/efectos de los fármacos , Ratones Endogámicos ICR , Nanopartículas Multifuncionales/administración & dosificación , Nanopartículas Multifuncionales/química , Nanopartículas Multifuncionales/toxicidad , Nanocompuestos/administración & dosificación , Nanocompuestos/toxicidad , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/química , Estrés Oxidativo/efectos de los fármacos , Células PC12 , Fragmentos de Péptidos/metabolismo , Péptidos/administración & dosificación , Péptidos/química , Péptidos/toxicidad , Multimerización de Proteína/efectos de los fármacos , Ratas , Resveratrol/administración & dosificación , Resveratrol/química , Selenio/administración & dosificación , Selenio/química , Selenio/toxicidad
15.
Molecules ; 26(17)2021 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-34500818

RESUMEN

Prevention of neurodegeneration during aging, and support of optimal brain function throughout the lifespan, requires protection of membrane structure and function. We review the synergistic action of different classes of dietary micronutrients, as well as further synergistic contributions from exercise and stress reduction, in supporting membrane structure and function. We address membrane-associated inflammation involving reactive oxygen species (ROS) that produce immune regulators from polyunsaturated fatty acids (PUFAs) of membrane phospholipids. The potential of dietary micronutrients to maintain membrane fluidity and prevent chronic inflammation is examined with a focus on synergistically acting membrane-soluble components (zeaxanthin, lutein, vitamin E, and omega-3 PUFAs) and water-soluble components (vitamin C and various phenolics). These different classes of micronutrients apparently operate in a series of intertwined oxidation-reduction cycles to protect membrane function and prevent chronic inflammation. At this time, it appears that combinations of a balanced diet with regular moderate exercise and stress-reduction practices are particularly beneficial. Effective whole-food-based diets include the Mediterranean and the MIND diet (Mediterranean-DASH Intervention for Neurodegenerative Delay diet, where DASH stands for Dietary Approaches to Stop Hypertension).


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Enfermedades Neurodegenerativas/prevención & control , Fármacos Neuroprotectores/farmacología , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/uso terapéutico , Antioxidantes/administración & dosificación , Antioxidantes/uso terapéutico , Suplementos Dietéticos , Sinergismo Farmacológico , Humanos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/uso terapéutico
16.
Sci Rep ; 11(1): 19392, 2021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34588483

RESUMEN

The synthetic copper-containing compound, CuATSM, has emerged as one of the most promising drug candidates developed for the treatment of amyotrophic lateral sclerosis (ALS). Multiple studies have reported CuATSM treatment provides therapeutic efficacy in various mouse models of ALS without any observable adverse effects. Moreover, recent results from an open label clinical study suggested that daily oral dosing with CuATSM slows disease progression in patients with both sporadic and familial ALS, providing encouraging support for CuATSM in the treatment of ALS. Here, we assessed CuATSM in high copy SOD1G93A mice on the congenic C57BL/6 background, treating at 100 mg/kg/day by gavage, starting at 70 days of age. This dose in this specific model has not been assessed previously. Unexpectedly, we report a subset of mice initially administered CuATSM exhibited signs of clinical toxicity, that necessitated euthanasia in extremis after 3-51 days of treatment. Following a 1-week washout period, the remaining mice resumed treatment at the reduced dose of 60 mg/kg/day. At this revised dose, treatment with CuATSM slowed disease progression and increased survival relative to vehicle-treated littermates. This work provides the first evidence that CuATSM produces positive disease-modifying outcomes in high copy SOD1G93A mice on a congenic C57BL/6 background. Furthermore, results from the 100 mg/kg/day phase of the study support dose escalation determination of tolerability as a prudent step when assessing treatments in previously unassessed models or genetic backgrounds.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Fármacos Neuroprotectores/administración & dosificación , Compuestos de Organocobre , Superóxido Dismutasa-1/metabolismo , Animales , Progresión de la Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Compuestos de Organocobre/administración & dosificación , Compuestos de Organocobre/efectos adversos , Compuestos de Organocobre/farmacología
17.
Expert Opin Investig Drugs ; 30(10): 1025-1035, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34555978

RESUMEN

INTRODUCTION: Recent years have witnessed unprecedented progress in stroke care, but unmet needs persist regarding the efficacy of acute treatment and secondary prevention. Novel approaches are being tested to enhance the efficacy of thrombolysis or provide neuroprotection in non-thrombolized patients. AREAS COVERED: The current review highlights pharmaceutical agents under evaluation in clinical trials concerning the acute, subacute, and chronic phase post-stroke. We examine the evidence in favor of tenecteplase as an alternative thrombolytic drug to alteplase, nerinetide as a promising neuroprotective agent, and glibenclamide for reducing edema in malignant hemispheric infarction. We discuss the use of ticagrelor and the promising novel category of factor XI inhibitors in the subacute phase after stroke. We offer our insights on combined rivaroxaban and antiplatelet therapy, PCSK-9 inhibitors, and other non-statin hypolipidemic agents, as well as novel antidiabetic agents that have been shown to reduce cardiovascular events in the long-term. EXPERT OPINION: Current approaches in stroke treatment and stroke prevention have already transformed stroke care from a linear one-for-all treatment paradigm to a more individualized approach that targets specific patient subgroups with novel pharmaceutical agents. This tendency enriches the therapeutic armamentarium with novel agents developed for specific stroke subgroups. ABBREVIATIONS: IVT: intravenous thrombolysis; RCTs: randomized-controlled clinical trials; TNK: Tenecteplase; COVID-19: Coronavirus 2019 Disease; EXTEND-IA TNK: The Tenecteplase versus Alteplase Before Endovascular Therapy for Ischemic Stroke trial; AIS: acute ischemic stroke; NNT: number needed to treat; MT: mechanical thrombectomy; sICH: symptomatic intracranial hemorrhage; mRS: modified Rankin Scale; AHA/ASA: American Heart Association/American Stroke Association; ESO: European Stroke Organization; NA-1: Nerinetide; ENACT: Evaluating Neuroprotection in Aneurysm Coiling Therapy; CTA: CT angiography; TIA: transient ischemic attack; CHANCE: Clopidogrel in High-risk patients with Acute Non-disabling Cerebrovascular Events; LOF: loss-of-function; PRINCE: Platelet Reactivity in Acute Nondisabling Cerebrovascular Events; THALES: Acute Stroke or Transient Ischemic Attack Treated with Ticagrelor and ASA [acetylsalicylic acid] for Prevention of Stroke and Death; CHANCE-2: Clopidogrel With Aspirin in High-risk Patients With Acute Non-disabling Cerebrovascular Events II; FXI: Factor XI; PACIFIC-STROKE: Program of Anticoagulation via Inhibition of FXIa by the Oral Compound BAY 2433334-NonCardioembolic Stroke study; COMPASS: Cardiovascular Outcomes for People Using Anticoagulation Strategies; CANTOS-ICAD: Combination Antithrombotic Treatment for Prevention of Recurrent Ischemic Stroke in Intracranial Atherosclerotic Disease; SAMMPRIS: Stenting and Aggressive Medical Therapy for Preventing Recurrent Stroke in Intracranial Stenosis; WASID: Warfarin-Aspirin Symptomatic Intracranial Disease; SPARCL: Stroke Prevention by Aggressive Reduction in Cholesterol Levels; LDL-C: low-density lipoprotein cholesterol; TST: Treat Stroke to Target; IMPROVE-IT: Improved Reduction of Outcomes: Vytorin Efficacy International Trial; PCSK9: proprotein convertase subtilisin-kexin type 9; FOURIER: Further Cardiovascular Outcomes Research with PCSK9 Inhibition in Subjects with Elevated Risk; CLEAR: Cholesterol Lowering via Bempedoic acid, an ACL-inhibiting Regimen; REDUCE-IT: Reduction of Cardiovascular Events With EPA Intervention Trial; STRENGTH: Outcomes Study to Assess STatin Residual Risk Reduction With EpaNova in HiGh CV Risk PatienTs With Hypertriglyceridemia; ACCORD: Action to Control Cardiovascular Risk in Diabetes; ADVANCE: Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation; VADT: Veterans Affairs Diabetes Trial; GLP-1R: Glucagon-like peptide-1 receptor; SGLT2: sodium-glucose cotransporter 2; CONVINCE: COlchicine for preventioN of Vascular Inflammation in Non-CardioEmbolic stroke; PROBE: Prospective Randomized Open-label Blinded Endpoint assessment.


Asunto(s)
Anticolesterolemiantes/administración & dosificación , Isquemia Encefálica/tratamiento farmacológico , Ensayos Clínicos como Asunto/métodos , Fibrinolíticos/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Accidente Cerebrovascular/tratamiento farmacológico , Isquemia Encefálica/sangre , Isquemia Encefálica/epidemiología , Factor XI/antagonistas & inhibidores , Factor XI/metabolismo , Humanos , Inhibidores de PCSK9 , Proproteína Convertasa 9/sangre , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/epidemiología , Resultado del Tratamiento
18.
Mar Drugs ; 19(8)2021 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-34436273

RESUMEN

The anti-amnesic effect of a mixture (4:6 = phlorotannin:fucoidan from Ecklonia cava, P4F6) was evaluated on amyloid-beta peptide (Aß)-induced cognitive deficit mice. The cognitive function was examined by Y-maze, passive avoidance, and Morris water maze tests, and the intake of the mixture (P4F6) showed an ameliorating effect on Aß-induced learning and memory impairment. After the behavioral tests, superoxide dismutase (SOD) activity and thiobarbituric acid-reactive substances (TBARS) contents were confirmed in brain tissue, and in the results, the mixture (P4F6) attenuated Aß-induced oxidative stress. In addition, mitochondrial activity was evaluated by mitochondrial reactive oxygen species (ROS) content, mitochondrial membrane potential (MMP), adenosine triphosphate (ATP) content, and mitochondria-mediated apoptotic signaling pathway, and the mixture (P4F6) enhanced mitochondrial function. Furthermore, the mixture (P4F6) effectively regulated tau hyperphosphorylation by regulating the protein kinase B (Akt) pathway, and promoted brain-derived neurotrophic factor (BDNF) in brain tissue. Moreover, in the cholinergic system, the mixture (P4F6) ameliorated acetylcholine (ACh) content by regulating acetylcholinesterase (AChE) activity and choline acetyltransferase (ChAT) expression in brain tissue. Based on these results, we suggest that this mixture of phlorotannin and fucoidan (P4F6) might be a substance for improving cognitive function by effectively regulating cognition-related molecules.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Kelp , Fármacos Neuroprotectores/administración & dosificación , Polisacáridos/administración & dosificación , Taninos/administración & dosificación , Acetilcolina/metabolismo , Animales , Organismos Acuáticos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Colinérgicos/metabolismo , Modelos Animales de Enfermedad , Quimioterapia Combinada , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Mitocondrias/metabolismo , Fármacos Neuroprotectores/farmacología , Fitoterapia , Polisacáridos/farmacología , Taninos/farmacología
19.
Biomed Pharmacother ; 142: 112048, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34435588

RESUMEN

Mannitol, a representative of hyperosmolar therapy, is indispensable for the treatment of malignant cerebral infarction, but its therapeutic effect is limited by its exacerbation of blood-brain barrier (BBB) disruption. This study was to explore whether Danhong injection (DHI), a standardized product extracted from Salvia miltiorrhiza Bunge and Carthamus tinctorius L., inhibits the destructive effect of mannitol on BBB and thus enhancing the treatment of hemispheric ischemic stroke. SD rats were subjected to pMCAO followed by intravenous bolus injections of mannitol with/without DHI intervention. Neurological deficit score, brain edema, infarct volume at 24 h after MCAO and histopathology, microvascular ultrastructure, immunohistochemistry and immunofluorescence staining of endothelial cell junctions, energy metabolism in the ischemic penumbra were assessed. Intravenous mannitol after MCAO resulted in a decrease in 24 h mortality and cerebral edema, whereas no significant benefit on neurological deficits, infarct volume and microvascular ultrastructure. Moreover, mannitol led to the loss of endothelial integrity, manifested by the decreased expression of occludin, junctional adhesion molecule-1 (JAM-1) and zonula occluden-1 (ZO-1) and the discontinuity of occludin staining around the periphery of endothelial cells. Meanwhile, after mannitol treatment, energy-dependent vimentin and F-actin, ATP content, and ATP5D expression were down-regulated, while MMP2 and MMP9 expression increased in the ischemic penumbra. All the insults after mannitol treatment were attenuated by addition of intravenous DHI. The results suggest DHI as a potential remedy to attenuate mannitol-related BBB disruption, and the potential of DHI to upregulate energy metabolism and inhibit the activity of MMPs is likely attributable to its effects observed.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Manitol/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Edema Encefálico/tratamiento farmacológico , Isquemia Encefálica/patología , Citoesqueleto/efectos de los fármacos , Modelos Animales de Enfermedad , Quimioterapia Combinada/métodos , Medicamentos Herbarios Chinos/administración & dosificación , Células Endoteliales/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Inyecciones , Uniones Intercelulares/efectos de los fármacos , Accidente Cerebrovascular Isquémico/patología , Manitol/uso terapéutico , Metaloproteinasas de la Matriz/efectos de los fármacos , Microvasos/efectos de los fármacos , Microvasos/ultraestructura , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Fármacos Neuroprotectores/administración & dosificación , Ratas Sprague-Dawley , Tasa de Supervivencia
20.
Food Funct ; 12(17): 7816-7824, 2021 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-34232246

RESUMEN

Gastrodin is the main bioactive ingredient of a famous Chinese herb Rhizoma Gastrodiae. Many studies have reported that gastrodin has antioxidative and neuroprotective effects, although its effect on longevity and the mechanism of neuroprotection have not been well studied. Here, we use Drosophila melanogaster as a model to investigate the longevity and neuroprotective effects of gastrodin. Gastrodin significantly extended the lifespan, increased the climbing ability, enhanced the resistance to oxidative stress, increased the enzyme activities of superoxide dismutase (SOD) and catalase (CAT), and promoted the expression of anti-oxidative genes in old flies. The food intake, reproduction and starvation resistance were not affected in flies treated with gastrodin. Moreover, gastrodin delayed the onset of Parkinson-like phenotypes in Pink1B9 mutant flies, including the prolongation of the lifespan, rescue of the climbing ability, rescue of the progressive loss of a cluster of dopaminergic neurons in the protocerebral posterial lateral 1 region, and increase of the dopamine content in the brain. Gastrodin did not ameliorate the tau-induced neurobehavioral deficits in the fly AD model of taupathy. Together, these results indicate that gastrodin could prolong the lifespan by regulating the antioxidant ability, and protect against neurodegeneration in the Pink1B9 model of PD. This suggests that gastrodin can be considered as an ideal therapeutic candidate for drug development towards anti-aging.


Asunto(s)
Alcoholes Bencílicos/administración & dosificación , Drosophila melanogaster/efectos de los fármacos , Medicamentos Herbarios Chinos/administración & dosificación , Gastrodia/química , Glucósidos/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Enfermedad de Parkinson/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Femenino , Humanos , Longevidad/efectos de los fármacos , Masculino , Neuroprotección/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/fisiopatología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA