Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 215
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biomed Sci ; 30(1): 66, 2023 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-37568205

RESUMEN

BACKGROUND: Parkinson's disease (PD) is the second most frequent age-related neurodegenerative disorder and is characterized by the loss of dopaminergic neurons. Both environmental and genetic aspects are involved in the pathogenesis of PD. Osmotin is a structural and functional homolog of adiponectin, which regulates the phosphorylation of 5' adenosine monophosphate-activated protein kinase (AMPK) via adiponectin receptor 1 (AdipoR1), thus attenuating PD-associated pathology. Therefore, the current study investigated the neuroprotective effects of osmotin using in vitro and in vivo models of PD. METHODS: The study used 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced and neuron-specific enolase promoter human alpha-synuclein (NSE-hαSyn) transgenic mouse models and 1-methyl-4-phenylpyridinium (MPP+)- or alpha-synuclein A53T-treated cell models. MPTP was injected at a dose of 30 mg/kg/day for five days, and osmotin was injected twice a week at a dose of 15 mg/kg for five weeks. We performed behavioral tests and analyzed the biochemical and molecular changes in the substantia nigra pars compacta (SNpc) and the striatum. RESULTS: Based on our study, osmotin mitigated MPTP- and α-synuclein-induced motor dysfunction by upregulating the nuclear receptor-related 1 protein (Nurr1) transcription factor and its downstream markers tyrosine hydroxylase (TH), dopamine transporter (DAT), and vesicular monoamine transporter 2 (VMAT2). From a pathological perspective, osmotin ameliorated neuronal cell death and neuroinflammation by regulating the mitogen-activated protein kinase (MAPK) signaling pathway. Additionally, osmotin alleviated the accumulation of α-synuclein by promoting the AMPK/mammalian target of rapamycin (mTOR) autophagy signaling pathway. Finally, in nonmotor symptoms of PD, such as cognitive deficits, osmotin restored synaptic deficits, thereby improving cognitive impairment in MPTP- and α-synuclein-induced mice. CONCLUSIONS: Therefore, our findings indicated that osmotin significantly rescued MPTP/α-synuclein-mediated PD neuropathology. Altogether, these results suggest that osmotin has potential neuroprotective effects in PD neuropathology and may provide opportunities to develop novel therapeutic interventions for the treatment of PD.


Asunto(s)
Fármacos Neuroprotectores , Enfermedad de Parkinson , Humanos , Ratones , Animales , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , alfa-Sinucleína/farmacología , Fármacos Neuroprotectores/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Sustancia Negra/metabolismo , Transducción de Señal , Neuronas Dopaminérgicas/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Serina-Treonina Quinasas TOR/farmacología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Mamíferos
2.
Int J Mol Sci ; 24(6)2023 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-36982361

RESUMEN

The blood-brain barrier (BBB) is a functional interface that provides selective permeability, protection from toxic substances, transport of nutrients, and clearance of brain metabolites. Additionally, BBB disruption has been shown to play a role in many neurodegenerative conditions and diseases. Therefore, the aim of this study was to establish a functional, convenient, and efficient in vitro co-cultured BBB model that can be used for several physiological conditions related to BBB disruption. Mouse brain-derived endothelial (bEnd.3) and astrocyte (C8-D1A) cells were co-cultured on transwell membranes to establish an intact and functional in vitro model. The co-cultured model and its effects on different neurological diseases and stress conditions, including Alzheimer's disease (AD), neuroinflammation, and obesity, have been examined by transendothelial electrical resistance (TEER), fluorescein isothiocyanate (FITC) dextran, and tight junction protein analyses. Scanning electron microscope images showed evidence of astrocyte end-feet processes passing through the membrane of the transwell. Moreover, the co-cultured model showed effective barrier properties in the TEER, FITC, and solvent persistence and leakage tests when compared to the mono-cultured model. Additionally, the immunoblot results showed that the expression of tight junction proteins such as zonula occludens-1 (ZO-1), claudin-5, and occludin-1 was enhanced in the co-culture. Lastly, under disease conditions, the BBB structural and functional integrity was decreased. The present study demonstrated that the co-cultured in vitro model mimicked the BBB's structural and functional integrity and, under disease conditions, the co-cultured model showed similar BBB damages. Therefore, the present in vitro BBB model can be used as a convenient and efficient experimental tool to investigate a wide range of BBB-related pathological and physiological studies.


Asunto(s)
Barrera Hematoencefálica , Encéfalo , Ratones , Animales , Barrera Hematoencefálica/metabolismo , Técnicas de Cocultivo , Fluoresceína-5-Isotiocianato/metabolismo , Encéfalo/metabolismo , Astrocitos/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Uniones Estrechas/metabolismo , Células Cultivadas
3.
Int J Mol Sci ; 24(12)2023 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-37373089

RESUMEN

Trolox is a potent antioxidant and a water-soluble analog of vitamin E. It has been used in scientific studies to examine oxidative stress and its impact on biological systems. Trolox has been shown to have a neuroprotective effect against ischemia and IL-1ß-mediated neurodegeneration. In this study, we investigated the potential protective mechanisms of Trolox against a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinson's disease mouse model. Western blotting, immunofluorescence staining, and ROS/LPO assays were performed to investigate the role of trolox against neuroinflammation, the oxidative stress mediated by MPTP in the Parkinson's disease (PD) mouse model (wild-type mice (C57BL/6N), eight weeks old, average body weight 25-30 g). Our study showed that MPTP increased the expression of α-synuclein, decreased tyrosine hydroxylase (TH) and dopamine transporter (DAT) levels in the striatum and substantia nigra pars compacta (SNpc), and impaired motor function. However, Trolox treatment significantly reversed these PD-like pathologies. Furthermore, Trolox treatment reduced oxidative stress by increasing the expression of nuclear factor erythroid-2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). Lastly, Trolox treatment inhibited the activated astrocytes (GFAP) and microglia (Iba-1), also reducing phosphorylated nuclear factor-κB, (p-NF-κB) and tumor necrosis factor-alpha (TNF-α) in the PD mouse brain. Overall, our study demonstrated that Trolox may exert neuroprotection on dopaminergic neurons against MPTP-induced oxidative stress, neuroinflammation, motor dysfunction, and neurodegeneration.


Asunto(s)
Trastornos Motores , Fármacos Neuroprotectores , Enfermedad de Parkinson , Animales , Ratones , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/efectos adversos , Enfermedades Neuroinflamatorias , Vitamina E/farmacología , Trastornos Motores/metabolismo , Sustancia Negra/metabolismo , Ratones Endogámicos C57BL , Tirosina 3-Monooxigenasa/metabolismo , Neuronas Dopaminérgicas/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Fármacos Neuroprotectores/metabolismo , Estrés Oxidativo , Modelos Animales de Enfermedad
4.
Int J Mol Sci ; 23(3)2022 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-35163692

RESUMEN

Recently, the world has been witnessing a global pandemic with no effective therapeutics yet, while cancer continues to be a major disease claiming many lives. The natural compound curcumin is bestowed with multiple medicinal applications in addition to demonstrating antiviral and anticancer activities. In order to elucidate the impact of curcumin on COVID-19 and cancer, the current investigation has adapted several computational techniques to unfold its possible inhibitory activity. Accordingly, curcumin and similar compounds and analogues were retrieved and assessed for their binding affinities at the binding pocket of SARS-CoV-2 main protease and DDX3. The best binding pose was escalated to molecular dynamics simulation (MDS) studies to assess the time dependent stability. Our findings have rendered one compound that has demonstrated good molecular dock score complemented by key residue interactions and have shown stable MDS results inferred by root mean square deviation (RMSD), radius of gyration (Rg), binding mode, hydrogen bond interactions, and interaction energy. Essential dynamics results have shown that the systemadapts minimum energy conformation to attain a stable state. The discovered compound (curA) could act as plausible inhibitor against SARS-CoV-2 and DDX3. Furthermore, curA could serve as a chemical scaffold for designing and developing new compounds.


Asunto(s)
Curcumina/análogos & derivados , Curcumina/farmacología , SARS-CoV-2/efectos de los fármacos , Antivirales/farmacología , Biología Computacional/métodos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Simulación del Acoplamiento Molecular/métodos , Simulación de Dinámica Molecular , Neoplasias/tratamiento farmacológico , Inhibidores de Proteasas/farmacología , Unión Proteica/efectos de los fármacos , SARS-CoV-2/patogenicidad , Tratamiento Farmacológico de COVID-19
5.
Int J Mol Sci ; 23(11)2022 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-35682768

RESUMEN

Traumatic brain injury (TBI) signifies a major cause of death and disability. TBI causes central nervous system (CNS) damage under a variety of mechanisms, including protein aggregation, mitochondrial dysfunction, oxidative stress, and neuroinflammation. Astrocytes and microglia, cells of the CNS, are considered the key players in initiating an inflammatory response after injury. Several evidence suggests that activation of astrocytes/microglia and ROS/LPO have the potential to cause more harmful effects in the pathological processes following traumatic brain injury (TBI). Previous studies have established that lupeol provides neuroprotection through modulation of inflammation, oxidative stress, and apoptosis in Aß and LPS model and neurodegenerative disease. However, the effects of lupeol on apoptosis caused by inflammation and oxidative stress in TBI have not yet been investigated. Therefore, we explored the role of Lupeol on antiapoptosis, anti-inflammatory, and antioxidative stress and its potential mechanism following TBI. In these experiments, adult male mice were randomly divided into four groups: control, TBI, TBI+ Lupeol, and Sham group. Western blotting, immunofluorescence staining, and ROS/LPO assays were performed to investigate the role of lupeol against neuroinflammation, oxidative stress, and apoptosis. Lupeol treatment reversed TBI-induced behavioral and memory disturbances. Lupeol attenuated TBI-induced generation of reactive oxygen species/lipid per oxidation (ROS/LPO) and improved the antioxidant protein level, such as nuclear factor erythroid 2-related factor 2 (Nrf2) and heme-oxygenase 1 (HO-1) in the mouse brain. Similarly, our results indicated that lupeol treatment inhibited glial cell activation, p-NF-κB, and downstream signaling molecules, such as TNF-α, COX-2, and IL-1ß, in the mouse cortex and hippocampus. Moreover, lupeol treatment also inhibited mitochondrial apoptotic signaling molecules, such as caspase-3, Bax, cytochrome-C, and reversed deregulated Bcl2 in TBI-treated mice. Overall, our study demonstrated that lupeol inhibits the activation of astrocytes/microglia and ROS/LPO that lead to oxidative stress, neuroinflammation, and apoptosis followed by TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Enfermedades Neurodegenerativas , Animales , Lesiones Traumáticas del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/metabolismo , Neuroglía/metabolismo , Estrés Oxidativo , Triterpenos Pentacíclicos , Especies Reactivas de Oxígeno/farmacología
6.
Int J Mol Sci ; 24(1)2022 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-36613586

RESUMEN

This review article is built on the beneficial effects of Lactobacillus against different diseases, and a special focus has been made on its effects against neurological disorders, such as depression, multiple sclerosis, Alzheimer's, and Parkinson's disease. Probiotics are live microbes, which are found in fermented foods, beverages, and cultured milk and, when administered in an adequate dose, confer health benefits to the host. They are known as "health-friendly bacteria", normally residing in the human gut and involved in maintaining homeostatic conditions. Imbalance in gut microbiota results in the pathophysiology of several diseases entailing the GIT tract, skin, immune system, inflammation, and gut-brain axis. Recently, the use of probiotics has gained tremendous interest, because of their profound effects on the management of these disease conditions. Recent findings suggest that probiotics enrichment in different human and mouse disease models showed promising beneficial effects and results in the amelioration of disease symptoms. Thus, this review focuses on the current probiotics-based products, different disease models, variable markers measured during trials, and evidence obtained from past studies on the use of probiotics in the prevention and treatment of different diseases, covering the skin to the central nervous system diseases.


Asunto(s)
Enfermedades del Sistema Nervioso Central , Microbioma Gastrointestinal , Probióticos , Animales , Humanos , Ratones , Lactobacillus , Probióticos/farmacología , Probióticos/uso terapéutico
7.
Int J Mol Sci ; 22(17)2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34502488

RESUMEN

The current study was undertaken to unveil the protective effects of Luteolin, a natural flavonoid, against amyloid-beta (Aß1-42)-induced neuroinflammation, amyloidogenesis, and synaptic dysfunction in mice. For the development of an AD mouse model, amyloid-beta (Aß1-42, 5 µL/5 min/mouse) oligomers were injected intracerebroventricularly (i.c.v.) into mice's brain by using a stereotaxic frame. After that, the mice were treated with Luteolin for two weeks at a dose of 80 mg/kg/day. To monitor the biochemical changes, we conducted western blotting and immunofluorescence analysis. According to our findings, the infusion of amyloid-beta activated c-Jun N-terminal kinases (p-JNK), p38 mitogen-activated protein kinases, glial fibrillary acidic protein (GFAP), and ionized calcium adaptor molecule 1 (Iba-1) in the cortex and hippocampus of the experimental mice; these changes were significantly inhibited in Aß1-42 + Luteolin-treated mice. Likewise, we also checked the expression of inflammatory markers, such as p-nuclear factor-kB p65 (p-NF-kB p65 (Ser536), tissue necrosis factor (TNF-α), and Interleukin1-ß (IL-1ß), in Aß1-42-injected mice brain, which was attenuated in Aß1-42 + Luteolin-treated mice brains. Further, we investigated the expression of pro- and anti-apoptotic cell death markers such as Bax, Bcl-2, Caspase-3, and Cox-2, which was significantly reduced in Aß1-42 + Lut-treated mice brains compared to the brains of the Aß-injected group. The results also indicated that with the administration of Aß1-42, the expression levels of ß-site amyloid precursor protein cleaving enzyme (BACE-1) and amyloid-beta (Aß1-42) were significantly enhanced, while they were reduced in Aß1-42 + Luteolin-treated mice. We also checked the expression of synaptic markers such as PSD-95 and SNAP-25, which was significantly enhanced in Aß1-42 + Lut-treated mice. To unveil the underlying factors responsible for the protective effects of Luteolin against AD, we used a specific JNK inhibitor, which suggested that Luteolin reduced Aß-associated neuroinflammation and neurodegeneration via inhibition of JNK. Collectively, our results indicate that Luteolin could serve as a novel therapeutic agent against AD-like pathological changes in mice.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides/toxicidad , Encéfalo/metabolismo , Luteolina/farmacología , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/toxicidad , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Modelos Animales de Enfermedad , Masculino , Ratones
8.
J Neuroinflammation ; 17(1): 303, 2020 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-33059700

RESUMEN

BACKGROUND: Glycine is the smallest nonessential amino acid and has previously unrecognized neurotherapeutic effects. In this study, we examined the mechanism underlying the neuroprotective effect of glycine (Gly) against neuroapoptosis, neuroinflammation, synaptic dysfunction, and memory impairment resulting from D-galactose-induced elevation of reactive oxygen species (ROS) during the onset of neurodegeneration in the brains of C57BL/6N mice. METHODS: After in vivo administration of D-galactose (D-gal; 100 mg/kg/day; intraperitoneally (i/p); for 60 days) alone or in combination with glycine (1 g/kg/day in saline solution; subcutaneously; for 60 days), all of the mice were sacrificed for further biochemical (ROS/lipid peroxidation (LPO) assay, Western blotting, and immunohistochemistry) after behavioral analyses. An in vitro study, in which mouse hippocampal neuronal HT22 cells were treated with or without a JNK-specific inhibitor (SP600125), and molecular docking analysis were used to confirm the underlying molecular mechanism and explore the related signaling pathway prior to molecular and histological analyses. RESULTS: Our findings indicated that glycine (an amino acid) inhibited D-gal-induced oxidative stress and significantly upregulated the expression and immunoreactivity of antioxidant proteins (Nrf2 and HO-1) that had been suppressed in the mouse brain. Both the in vitro and in vivo results indicated that D-gal induced oxidative stress-mediated neurodegeneration primarily by upregulating phospho-c-Jun N-terminal kinase (p-JNK) levels. However, D-gal + Gly cotreatment reversed the neurotoxic effects of D-gal by downregulating p-JNK levels, which had been elevated by D-gal. We also found that Gly reversed D-gal-induced neuroapoptosis by significantly reducing the protein expression levels of proapoptotic markers (Bax, cytochrome c, cleaved caspase-3, and cleaved PARP-1) and increasing the protein expression level of the antiapoptotic protein Bcl-2. Both the molecular docking approach and the in vitro study (in which the neuronal HT22 cells were treated with or without a p-JNK-specific inhibitor (SP600125)) further verified our in vivo findings that Gly bound to the p-JNK protein and inhibited its function and the JNK-mediated apoptotic pathway in the mouse brain and HT22 cells. Moreover, the addition of Gly alleviated D-gal-mediated neuroinflammation by inhibiting gliosis via attenuation of astrocytosis (GFAP) and microgliosis (Iba-1) in addition to reducing the protein expression levels of various inflammatory cytokines (IL-1ßeta and TNFα). Finally, the addition of Gly reversed D-gal-induced synaptic dysfunction by upregulating the expression of memory-related presynaptic protein markers (synaptophysin (SYP), syntaxin (Syn), and a postsynaptic density protein (PSD95)) and markedly improved behavioral measures of cognitive deficits in D-gal-treated mice. CONCLUSION: Our findings demonstrate that Gly-mediated deactivation of the JNK signaling pathway underlies the neuroprotective effect of Gly, which reverses D-gal-induced oxidative stress, apoptotic neurodegeneration, neuroinflammation, synaptic dysfunction, and memory impairment. Therefore, we suggest that Gly (an amino acid) is a safe and promising neurotherapeutic candidate that might be used for age-related neurodegenerative diseases.


Asunto(s)
Galactosa/toxicidad , Glicina/uso terapéutico , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Trastornos de la Memoria/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuroprotección/efectos de los fármacos , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Relación Dosis-Respuesta a Droga , Glicina/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/prevención & control , Ratones , Ratones Endogámicos C57BL , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/prevención & control , Neuroprotección/fisiología
9.
Int J Mol Sci ; 22(1)2020 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-33396372

RESUMEN

The receptor for advanced glycation end products (RAGE), a pattern recognition receptor signaling event, has been associated with several human illnesses, including neurodegenerative diseases, particularly in Alzheimer's disease (AD). Vanillic acid (V.A), a flavoring agent, is a benzoic acid derivative having a broad range of biological activities, including antioxidant, anti-inflammatory, and neuroprotective effects. However, the underlying molecular mechanisms of V.A in exerting neuroprotection are not well investigated. The present study aims to explore the neuroprotective effects of V.A against lipopolysaccharides (LPS)-induced neuroinflammation, amyloidogenesis, synaptic/memory dysfunction, and neurodegeneration in mice brain. Behavioral tests and biochemical and immunofluorescence assays were applied. Our results indicated increased expression of RAGE and its downstream phospho-c-Jun n-terminal kinase (p-JNK) in the LPS-alone treated group, which was significantly reduced in the V.A + LPS co-treated group. We also found that systemic administration of LPS-injection induced glial cells (microglia and astrocytes) activation and significantly increased expression level of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-KB) and secretion of proinflammatory cytokines including tumor necrosis factor alpha (TNF-α), interleukin-1 ß (IL1-ß), and cyclooxygenase (COX-2). However, V.A + LPS co-treatment significantly inhibited the LPS-induced activation of glial cells and neuroinflammatory mediators. Moreover, we also noted that V.A treatment significantly attenuated LPS-induced increases in the expression of AD markers, such as ß-site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1) and amyloid-ß (Aß). Furthermore, V.A treatment significantly reversed LPS-induced synaptic loss via enhancing the expression level of pre- and post-synaptic markers (PSD-95 and SYP), and improved memory performance in LPS-alone treated group. Taken together; we suggest that neuroprotective effects of V.A against LPS-induced neurotoxicity might be via inhibition of LPS/RAGE mediated JNK signaling pathway; and encourage future studies that V.A would be a potential neuroprotective and neurotherapeutic candidate in various neurological disorders.


Asunto(s)
Encéfalo/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Gliosis/tratamiento farmacológico , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos/toxicidad , Fármacos Neuroprotectores/farmacología , Ácido Vanílico/farmacología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Gliosis/inducido químicamente , Gliosis/metabolismo , Gliosis/patología , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Masculino , Ratones , Ratones Endogámicos C57BL
10.
Int J Obes (Lond) ; 43(12): 2422-2433, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31164725

RESUMEN

OBJECTIVES: Obesity is characterized by excessive fat accumulation due to an imbalance between energy intake and expenditure. Osmotin, a plant derived natural protein, is a known homolog of adiponectin. To analyze the role of Osmotin in controlling energy metabolism by suppressing abdominal fat accumulation. METHODS: We investigated the effects of osmotin in C57BL/6 mice on high-fat diet and in 3T3-L1 adipocytes by Biochemical tests, Immunofluorescence confocal Microscopy, RT-PCR, and Flow cytometry. RESULTS: In this study, we investigated the anti-obesity effects of osmotin on adipocyte differentiation and regulation of the related factors lipolysis and glucose uptake in 3T3-L1 cells in vitro. Moreover, we analyzed the role of osmotin in prevention of insulin resistance, excess fat accumulation and metabolic syndrome in high-fat diet mouse model via AMPK and MAPK pathways in vivo. In addition, osmotin caused cell cycle arrest in G0/G1 phase by regulating expression of p21, p27 and CDK2 and improved glucose control, as concluded from glucose and insulin tolerance tests. CONCLUSION: These results reveal the role of osmotin in AMPK downstream signaling. These results provide the first indication that osmotin exerts therapeutic effects on obesity, which could promote development of therapeutic aspects for obesity and related diseases.


Asunto(s)
Grasa Abdominal/efectos de los fármacos , Adiponectina/análogos & derivados , Fármacos Antiobesidad/farmacología , Proteínas de Plantas/farmacología , Células 3T3-L1 , Grasa Abdominal/metabolismo , Adipocitos/efectos de los fármacos , Animales , Fármacos Antiobesidad/química , Peso Corporal/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Dieta Alta en Grasa , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Plantas/química
11.
Cereb Cortex ; 28(8): 2854-2872, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29088310

RESUMEN

Traumatic brain injury (TBI) is a global risk factor that leads to long-term cognitive impairments. To date, the disease remains without effective therapeutics because of the multifactorial nature of the disease. Here, we demonstrated that activation of the c-Jun N-terminal kinase (JNK) is involved in multiple pathological features of TBI. Therefore, we investigated the disease-modifying therapeutic potential of JNK-specific inhibitor (SP600125) in TBI mice. Treating 2 different models of TBI mice with SP600125 for 7 days dramatically inhibited activated JNK, resulting in marked reductions of amyloid precursor protein (APP) expression level and in amyloid beta production and hyperphosphorylated tau and regulation of the abnormal expression of secretases. Furthermore, SP600125 strongly inhibited inflammatory responses, blood-brain barrier breakdown, apoptotic neurodegeneration, and synaptic protein loss, regulated prosurvival processes and improved motor function and behavioral outcomes in TBI mice. More interestingly, we found that SP600125 treatment ameliorated amyloidogenic APP processing and promoted the nonamyloidogenic pathway in TBI mouse brains. Our findings strongly suggest that active JNK is critically involved in disease development after TBI and that inhibition of JNK with SP600125 is highly efficient for slowing disease progression by reducing multiple pathological features in TBI mouse brains and regulating cognitive dysfunction.


Asunto(s)
Antracenos/uso terapéutico , Lesiones Traumáticas del Encéfalo/complicaciones , Encéfalo/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Discapacidades para el Aprendizaje/tratamiento farmacológico , Discapacidades para el Aprendizaje/etiología , Péptidos beta-Amiloides/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiopatología , Encéfalo/metabolismo , Edema Encefálico/etiología , Edema Encefálico/prevención & control , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fluoresceínas/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/metabolismo , Desempeño Psicomotor/efectos de los fármacos
13.
Int J Mol Sci ; 20(5)2019 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-30862043

RESUMEN

Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder characterized by progressive memory dysfunction and a decline in cognition. One of the biggest challenges to study the pathological process at a molecular level is that there is no simple, cost-effective, and comprehensive gene-expression analysis tool. The present study provides the most detailed (Reverse transcription polymerase chain reaction) RT-PCR-based gene-expression assay, encompassing important genes, based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) disease pathway. This study analyzed age-dependent disease progression by focusing on pathological events such as the processing of the amyloid precursor protein, tau pathology, mitochondrial dysfunction, endoplasmic reticulum stress, disrupted calcium signaling, inflammation, and apoptosis. Messenger RNA was extracted from the cortex and hippocampal region of APP/PS1 transgenic mice. Samples were divided into three age groups, six-, nine-, and 12-month-old transgenic mice, and they were compared with normal C57BL/6J mice of respective age groups. Findings of this study provide the opportunity to design a simple, effective, and accurate clinical analysis tool that can not only provide deeper insight into the disease, but also act as a clinical diagnostic tool for its better diagnosis.


Asunto(s)
Envejecimiento/genética , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/patología , Perfilación de la Expresión Génica , Transcriptoma , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Apoptosis/genética , Señalización del Calcio , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Estrés del Retículo Endoplásmico , Redes Reguladoras de Genes , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Agregado de Proteínas , Agregación Patológica de Proteínas/metabolismo , Transducción de Señal , Proteínas tau/metabolismo
14.
Int J Mol Sci ; 20(10)2019 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-31117242

RESUMEN

Oxidative stress has been considered as the main mediator in neurodegenerative diseases. A high-fat diet (HFD) and metabolic diseases result in oxidative stress generation, leading to various neurodegenerative diseases via molecular mechanisms that remain largely unknown. Protein kinases play an important role in the homeostasis between cell survival and cell apoptosis. The mammalian sterile 20-like kinase-1 (MST1) protein kinase plays an important role in cellular apoptosis in different organ systems, including the central nervous system. In this study, we evaluated the MST1/c-Jun N-terminal kinase (JNK) dependent oxidative damage mediated cognitive dysfunction in HFD-fed mice and stress-induced hippocampal HT22 (mice hippocampal) cells. Our Western blot and immunofluorescence results indicate that HFD and stress-induced hippocampal HT22 cells activate MST1/JNK/Caspase-3 (Casp-3) signaling, which regulates neuronal cell apoptosis and beta-amyloid-cleaving enzyme (BACE1) expression and leads to impaired cognition. Moreover, MST1 expression inhibition by shRNA significantly reduced JNK/Casp-3 signaling. Our in vivo and in vitro experiments mimicking metabolic stress, such as a high-fat diet, hyperglycemia, and an inflammatory response, determined that MST1 plays a key regulatory role in neuronal cell death and cognition, suggesting that MST1 could be a potential therapeutic target for numerous neurodegenerative diseases.


Asunto(s)
Apoptosis , Encéfalo/metabolismo , Dieta Alta en Grasa , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Animales , Encéfalo/fisiología , Caspasa 3/metabolismo , Línea Celular , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
15.
J Proteome Res ; 17(8): 2715-2726, 2018 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-29893128

RESUMEN

Cell-penetrating peptides (CPPs) can enter cells as a variety of biologically active conjugates and have various biomedical applications. To offset the cost and effort of designing novel CPPs in laboratories, computational methods are necessitated to identify candidate CPPs before in vitro experimental studies. We developed a two-layer prediction framework called machine-learning-based prediction of cell-penetrating peptides (MLCPPs). The first-layer predicts whether a given peptide is a CPP or non-CPP, whereas the second-layer predicts the uptake efficiency of the predicted CPPs. To construct a two-layer prediction framework, we employed four different machine-learning methods and five different compositions including amino acid composition (AAC), dipeptide composition, amino acid index, composition-transition-distribution, and physicochemical properties (PCPs). In the first layer, hybrid features (combination of AAC and PCP) and extremely randomized tree outperformed state-of-the-art predictors in CPP prediction with an accuracy of 0.896 when tested on independent data sets, whereas in the second layer, hybrid features obtained through feature selection protocol and random forest produced an accuracy of 0.725 that is better than state-of-the-art predictors. We anticipate that our method MLCPP will become a valuable tool for predicting CPPs and their uptake efficiency and might facilitate hypothesis-driven experimental design. The MLCPP server interface along with the benchmarking and independent data sets are freely accessible at www.thegleelab.org/MLCPP .


Asunto(s)
Péptidos de Penetración Celular/farmacocinética , Biología Computacional , Máquina de Vectores de Soporte , Aminoácidos/análisis , Animales , Péptidos de Penetración Celular/química , Diseño de Fármacos , Humanos , Aprendizaje Automático , Modelos Teóricos
16.
J Neuroinflammation ; 15(1): 119, 2018 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-29679979

RESUMEN

BACKGROUND: Melatonin is a well-known potent endogenous antioxidant pharmacological agent with significant neuroprotective actions. Here in the current study, we explored the nuclear factor erythroid 2-related factor 2 (Nrf2) gene-dependent antioxidant mechanism underlying the neuroprotective effects of the acute melatonin against acute ethanol-induced elevated reactive oxygen species (ROS)-mediated neuroinflammation and neurodegeneration in the developing rodent brain. METHODS: In vivo rat pups were co-treated with a single dose of acute ethanol (5 g/kg, subcutaneous (S.C.)) and a single dose of acute melatonin (20 mg/kg, intraperitoneal (I.P.)). Four hours after a single S.C. and I.P. injections, all of the rat pups were sacrificed for further biochemical (Western blotting, ROS- assay, LPO-assay, and immunohistochemical) analyses. In order to corroborate the in vivo results, we used the in vitro murine-hippocampal HT22 and microglial BV2 cells, which were subjected to knockdown with small interfering RNA (siRNA) of Nrf2 genes and exposed with melatonin (100 µM) and ethanol (100 mM) and proceed for further biochemical analyses. RESULTS: Our biochemical, immunohistochemical, and immunofluorescence results demonstrate that acute melatonin significantly upregulated the master endogenous antioxidant Nrf2 and heme oxygenase-1, consequently reversing the acute ethanol-induced elevated ROS and oxidative stress in the developing rodent brain, and in the murine-hippocampal HT22 and microglial BV2 cells. In addition, acute melatonin subsequently reduced the activated MAPK-p-P38-JNK pathways and attenuated neuroinflammation by decreasing the expression of activated gliosis and downregulated the p-NF-K-B/p-IKKß pathway and decreased the expression levels of other inflammatory markers in the developing rodent brain and BV2 cells. Of note, melatonin acted through the Nrf2-dependent mechanism to attenuate neuronal apoptosis in the postnatal rodent brain and HT22 cells. Immunohistofluorescence results also showed that melatonin prevented ethanol-induced neurodegeneration in the developing rodent brain. The in vitro results indicated that melatonin induced neuroprotection via Nrf2-dependent manner and reduced ethanol-induced neurotoxicity. CONCLUSIONS: The pleiotropic and potent neuroprotective antioxidant characteristics of melatonin, together with our in vivo and in vitro findings, suppose that acute melatonin could be beneficial to prevent and combat the acute ethanol-induced neurotoxic effects, such as elevated ROS, neuroinflammation, and neurodegeneration in the developing rodent brain.


Asunto(s)
Antioxidantes , Melatonina , Factor 2 Relacionado con NF-E2 , Síndromes de Neurotoxicidad , Animales , Femenino , Masculino , Animales Recién Nacidos , Antioxidantes/uso terapéutico , Proteínas de Unión al Calcio/metabolismo , Línea Celular Transformada , Depresores del Sistema Nervioso Central/toxicidad , Discapacidades del Desarrollo/tratamiento farmacológico , Discapacidades del Desarrollo/etiología , Modelos Animales de Enfermedad , Etanol/toxicidad , Hemo-Oxigenasa 1/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Melatonina/uso terapéutico , Proteínas de Microfilamentos/metabolismo , Síndromes de Neurotoxicidad/complicaciones , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/prevención & control , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos
17.
J Nanobiotechnology ; 15(1): 12, 2017 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-28173812

RESUMEN

BACKGROUND: In order to increase the bioavailability of hydrophilic unstable drugs like anthocyanins, we employed a polymer-based nanoparticles approach due to its unique properties such as high stability, improved bioavailability and high water-soluble drug loading efficiency. Anthocyanins constitute a subfamily of flavonoids that possess anti-oxidative, anti-inflammatory and neuroprotective properties. However, anthocyanins are unstable because their phenolic hydroxyl groups are easily oxidized into quinones, causing a reduced biological activity. To overcome this drawback and improve the free radical scavenging capabilities of anthocyanins, in the current study we for the first time encapsulated the anthocyanins in biodegradable nanoparticle formulation based on poly (lactide-co-glycolide) (PLGA) and a stabilizer polyethylene glycol (PEG)-2000. The biological activity and neuroprotective effect of anthocyanin loaded nanoparticles (An-NPs) were investigated in SH-SY5Y cell lines. RESULTS: Morphological examination under transmission electron microscopy (TEM) showed the formation of smooth spherically shaped nanoparticles. The average particle size and zeta potential of An-NPs were in the range of 120-165 nm and -12 mV respectively, with a low polydispersity index (0.4) and displayed a biphasic release profile in vitro. Anthocyanins encapsulation in PLGA@PEG nanoparticles (NPs) did not destroy its inherent properties and exhibit more potent neuroprotective properties. An-NPs were nontoxic to SH-SY5Y cells and increased their cell viability against Aß1-42 by its free radical scavenging characteristics and abrogated ROS generation via the p38-MAPK/JNK pathways accompanied by induction of endogenous nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1). Comparative to native bulk anthocyanins, An-NPs effectively attenuated Alzheimer's markers like APP (amyloid precursor protein), BACE-1 (beta-site amyloid precursor protein cleaving enzyme 1), neuroinflammatory markers such as p-NF-kB (phospho-nuclear factor kappa B), TNF-α (tumor necrosis factor) and iNOS (inducible nitric oxide synthase) and neuroapoptotic markers including Bax, Bcl2, and Caspase-3 protein expressions accompanied by neurodegeneration against Aß1-42 in SH-SY5Y cell lines. CONCLUSIONS: Overall, this data not only confirmed the therapeutic potential of anthocyanins in reducing AD pathology but also offer an effective way to improve the efficiency of anthocyanins through the use of nanodrug delivery systems.


Asunto(s)
Antocianinas/farmacología , Depuradores de Radicales Libres/farmacología , Ácido Láctico/química , Sistema de Señalización de MAP Quinasas , Nanopartículas/química , Ácido Poliglicólico/química , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/farmacología , Disponibilidad Biológica , Técnicas de Cultivo de Célula , Línea Celular , Supervivencia Celular , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Humanos , Microscopía Electrónica de Transmisión , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo , Tamaño de la Partícula , Fragmentos de Péptidos/farmacología , Polietilenglicoles/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
18.
Nanomedicine ; 13(8): 2533-2544, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28736294

RESUMEN

Amyloid-beta (Aß1-42) plaques and neurofibrillary tangles (NFTs) are the main hallmarks considered to be associated with neuroinflammation in Alzheimer's disease (AD). Recently, nanoparticle-based targeted drug delivery approaches have been found to be a useful tool in the neurotherapeutics field. Therefore, we examined and compared the neuroprotective effect of anthocyanins alone and anthocyanin-loaded poly (ethylene glycol)-gold nanoparticles (PEG-AuNPs) in Aß1-42-injected mouse and in vitro models of AD. We determined that anthocyanins alone or conjugated with PEG-AuNPs (AnPEG-AuNPs) reduced Aß1-42-induced neuroinflammatory and neuroapoptotic markers via inhibiting the p-JNK/NF-κB/p-GSK3ß pathway in both in vivo and in vitro AD models. However, anthocyanins loaded with PEG-AuNPs were more effective compared to anthocyanins alone. Taken together, these results demonstrate that PEG-coated gold anthocyanins nanoparticles could be a new therapeutic agent in the field of nanomedicine to prevent neurodegenerative diseases such as AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/antagonistas & inhibidores , Antocianinas/uso terapéutico , Oro/química , Inflamación/tratamiento farmacológico , Nanopartículas del Metal/química , Fármacos Neuroprotectores/uso terapéutico , Fragmentos de Péptidos/antagonistas & inhibidores , Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Animales , Antocianinas/administración & dosificación , Sistemas de Liberación de Medicamentos , Glucógeno Sintasa Quinasa 3 beta/inmunología , Inflamación/inmunología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , FN-kappa B/inmunología , Fármacos Neuroprotectores/administración & dosificación , Fragmentos de Péptidos/inmunología , Polietilenglicoles/química , Transducción de Señal/efectos de los fármacos
19.
Pak J Med Sci ; 33(3): 581-585, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28811775

RESUMEN

OBJECTIVE: Seizures are one of the neurodegenerative disorders of human being. Metformin has antioxidant properties and commonly used as an oral antidiabetic drug. The current study was aimed to observe the neuroprotective effect of metformin against PTZ-induced apoptotic neurodegeneration in human cortical neuronal cell culture. METHODS: To observe that exposure of pentylenetetrazol (PTZ) at the dose of (30mM) for 30 minutes induced neuronal cell death by activation of caspase-3 in human cortical neuronal 2 (HCN-2) cell line. While the metformin at the dose of (20mM) along with PTZ for 30 minutes showed neuroprotection against PTZ-induced neuronal cell loss by MTT assay and Western blot analysis. RESULTS: The results of this study showed that PTZ-induced neuronal cell death by activation of pro apoptotic proteins caspase-3 and 9 whereas the exposure of metformin showed its protective effect against neuronal loss in HCN-2 cell line. Finally, our results showed that exposure of metformin can prevent the harmful effect induced by PTZ in neuronal cells cultures. CONCLUSIONS: Our finding suggest that metformin exposure attenuates PTZ-induced neuronal cell death may act as a safe therapeutics and neuroprotective agent for the treatment of neuronal loss as result of seizure.

20.
Int J Cancer ; 138(5): 1232-45, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26413934

RESUMEN

Elevated expression and aberrant activation of Ras have been implicated in breast cancer aggressiveness. H-Ras, but not N-Ras, induces breast cell invasion. A crucial link between lipid rafts and H-Ras function has been suggested. This study sought to identify the lipid raft protein(s) responsible for H-Ras-induced tumorigenicity and invasiveness of breast cancer. We conducted a comparative proteomic analysis of lipid raft proteins from invasive MCF10A human breast epithelial cells engineered to express active H-Ras and non-invasive cells expressing active N-Ras. Here, we identified a lipid raft protein flotillin-1 as an important regulator of H-Ras activation and breast cell invasion. Flotillin-1 was required for epidermal growth factor-induced activation of H-Ras, but not that of N-Ras, in MDA-MB-231 triple-negative breast cancer (TNBC) cells. Flotillin-1 knockdown inhibited the invasiveness of MDA-MB-231 and Hs578T TNBC cells in vitro and in vivo. In xenograft mouse tumor models of these TNBC cell lines, we showed that flotillin-1 played a critical role in tumor growth. Using human breast cancer samples, we provided clinical evidence for the metastatic potential of flotillin-1. Membrane staining of flotillin-1 was positively correlated with metastatic spread (p = 0.013) and inversely correlated with patient disease-free survival rates (p = 0.005). Expression of flotillin-1 was associated with H-Ras in breast cancer, especially in TNBC (p < 0.001). Our findings provide insight into the molecular basis of Ras isoform-specific interplay with flotillin-1, leading to tumorigenicity and aggressiveness of breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Genes ras , Proteínas de la Membrana/fisiología , Adulto , Anciano , Animales , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Movimiento Celular , Receptores ErbB/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación , Proteómica , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA