Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 357
Filtrar
1.
Biomed Pharmacother ; 153: 113456, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36076569

RESUMEN

Dexamethasone acts as an immunosuppressive drug and has been used recently in the management of specific coronavirus disease 2019 (COVID-19) cases; however, various adverse effects could limit its use. In this work, we studied the mitigation effects of black pepper oil (BP oil) on glycemic parameters, dyslipidemia, oxidative and nitrosative stress and pancreatic fibrosis in dexamethasone-treated rats. Animals were divided into five groups that were treated with vehicle, dexamethasone (10 mg/kg, SC) or black pepper oil (BP oil, 0.5 mL, or 1 mL/kg) or metformin (50 mg/kg) plus dexamethasone for 4 consecutive days. Serum insulin, blood glucose, total cholesterol, triglycerides, and Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) were higher in the dexamethasone group vs the control group and decreased in BP oil and metformin groups relative to the dexamethasone group. Pancreatic nitric oxide, inducible nitric oxide synthase and malondialdehyde levels were increased in the dexamethasone group vs the control group and decreased in BP oil and metformin groups relative to the dexamethasone group. Pancreatic endothelial nitric oxide synthase and reduced glutathione were declined in the dexamethasone group vs the control group. They were increased in BP oil and metformin groups relative to the dexamethasone group. Moreover, the pancreatic islets diameter and collagen deposition were assessed and found to be higher in the dexamethasone group vs the control group. BP oil and metformin groups showed to regress this effect. In conclusion, BP oil may alleviate hyperglycemia, hyperinsulinemia, insulin resistance, dyslipidemia and pancreatic structural derangements and fibrosis by suppressing oxidative stress, increasing endogenous antioxidant levels, modulating nitric oxide signaling, preventing pancreatic stellate cells transition and collagen deposition.


Asunto(s)
Dexametasona , Metformina , Páncreas , Piper nigrum , Aceites de Plantas , Animales , Glucemia , Dexametasona/efectos adversos , Dexametasona/farmacología , Dislipidemias/tratamiento farmacológico , Fibrosis , Resistencia a la Insulina , Metformina/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estrés Oxidativo/efectos de los fármacos , Páncreas/efectos de los fármacos , Páncreas/patología , Piper nigrum/química , Aceites de Plantas/farmacología , Aceites de Plantas/uso terapéutico , Ratas , Ratas Wistar , Tratamiento Farmacológico de COVID-19
2.
J Ethnopharmacol ; 290: 115086, 2022 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-35157952

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Chestnut flowers were one of the by-products during chestnut industrial processing. Chestnut (Castanea mollissima Blume) flower is rich in flavonoids and has been used as a traditional medicine to treat a variety of diseases including respiratory disorders for a long history. AIM OF THE STUDY: The present study aims to investigate the potential anti-inflammatory effect of flavonoids from chestnut flower (FCF) in lipopolysaccharide (LPS)-treated RAW 264.7 cells and stimulated acute lung injury (ALI) in mice. MATERIALS AND METHODS: HPLC-ESI-MS/MS was applied to identify flavonoids from Chestnut flower. The ROS content in cells and lung tissue was measured by flow cytometry. The malondialdehyde (MDA) content, superoxide dismutase (SOD) activity and glutathione (GSH) content in cells and bronchoalveolar lavage fluid (BALF) was analyzed by photometry. Furthermore, the level of pro-inflammatory factors was analyzed by ELISA, and the expression of inflammatory gene mRNA by fluorescence quantitative PCR. H&E staining was used to evaluate the degree of lung tissue injury in mice. MPO activity was used to measure the degree of neutrophil infiltration. Total protein content was detected by BCA method. RESULTS: A total of forty-nine flavonoids compounds were tentatively identified in FCF by mass spectrometry analysis. The results of cell experiment suggested that FCF could alleviate oxidative injury via increasing SOD activity and GSH content, as well as inhibiting the production of intracellular ROS and MDA. FCF exerted its protective effect by suppressing the expression of both inducible nitric oxide synthase (iNOS) and cycooxygenase 2 (COX-2) to inhibit the synthesis of pro-inflammatory factors and cytokines, including NO, PGE2, TNF-α, IL-6 and IL-1ß. Besides, FCF treatment could alleviate the thickening of alveolar wall and pulmonary congestion in LPS-treated ALI mice, and significantly inhibit the activity of myeloperoxidas (MPO) and the expression of cytokines in BALF. CONCLUSIONS: FCF could ameliorate inflammation and oxidative stress in LPS-treated inflammation, resulting in an overall improvement in both macroscopic and histological parameters.


Asunto(s)
Lesión Pulmonar Aguda/patología , Antiinflamatorios/farmacología , Flavonoides/farmacología , Extractos Vegetales/farmacología , Animales , Líquido del Lavado Bronquioalveolar/citología , Supervivencia Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Citocinas/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Flores , Glutatión/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Macrófagos/efectos de los fármacos , Masculino , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Células RAW 264.7 , Distribución Aleatoria , Superóxido Dismutasa/efectos de los fármacos , Espectrometría de Masas en Tándem
3.
Hum Exp Toxicol ; 41: 9603271211066065, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35130744

RESUMEN

Cardiovascular disorders are the leading cause of death globally. Rosuvastatin is a member of statins (inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase) with many pleiotropic properties. This study investigated cardioprotective effects of rosuvastatin in isoprenaline-induced myocardial injury. Male rats were given rosuvastatin (1, 5, or 10 mg/kg, oral) daily for 1 week and on seventh and eighth day isoprenaline (150 mg/kg, subcutaneous) was given to induce cardiac injury. On ninth day, rats were euthanized and different samples were harvested for analysis. Isoprenaline administration resulted in increased cardiac mass, increased cardiac injury marker levels (cTnI, CK-MB, ALT, and AST), increased lipid/protein oxidation, and increased cardiac nitrite levels. It also decreased superoxide dismutase, CAT, GST, and glutathione reductase activities, and total antioxidant activity. Isoprenaline also increased TNF-α and IL-6 levels. Decreased mRNA expression of Nrf2 and Bcl-2 along with increased mRNA expression of Bax, eNOS and iNOS genes was observed in isoprenaline treated animals. Histopathological evaluations of rosuvastatin pre-treated groups showed reduction of myocardial necrosis. Pretreatment with rosuvastatin (5 and 10 mg/kg) reduced many of these pathological changes. The current study showed that rosuvastatin significantly reduces myocardial injury induced by isoprenaline.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Isoproterenol/efectos adversos , Infarto del Miocardio/prevención & control , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Rosuvastatina Cálcica/administración & dosificación , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antioxidantes , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Isoproterenol/uso terapéutico , Masculino , Infarto del Miocardio/inducido químicamente , Infarto del Miocardio/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/genética , Óxido Nítrico Sintasa de Tipo II/genética , Sustancias Protectoras/administración & dosificación , Proteínas Proto-Oncogénicas c-bcl-2/genética , Ratas , Ratas Wistar
4.
Acta Pharmacol Sin ; 43(3): 602-612, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34011968

RESUMEN

Cardiac fibrosis (CF) is an irreversible pathological process that occurs in almost all kinds of cardiovascular diseases. Phosphorylation-dependent activation of c-Jun N-terminal kinase (JNK) induces cardiac fibrosis. However, whether S-nitrosylation of JNK mediates cardiac fibrosis remains an open question. A biotin-switch assay confirmed that S-nitrosylation of JNK (SNO-JNK) increased significantly in the heart tissues of hypertrophic patients, transverse aortic constriction (TAC) mice, spontaneously hypertensive rats (SHRs), and neonatal rat cardiac fibroblasts (NRCFs) stimulated with angiotensin II (Ang II). Site to site substitution of alanine for cysteine in JNK was applied to determine the S-nitrosylated site. S-Nitrosylation occurred at both Cys116 and Cys163 and substitution of alanine for cysteine 116 and cysteine 163 (C116/163A) inhibited Ang II-induced myofibroblast transformation. We further confirmed that the source of S-nitrosylation was inducible nitric oxide synthase (iNOS). 1400 W, an inhibitor of iNOS, abrogated the profibrotic effects of Ang II in NRCFs. Mechanistically, SNO-JNK facilitated the nuclear translocation of JNK, increased the phosphorylation of c-Jun, and induced the transcriptional activity of AP-1 as determined by chromatin immunoprecipitation and EMSA. Finally, WT and iNOS-/- mice were subjected to TAC and iNOS knockout reduced SNO-JNK and alleviated cardiac fibrosis. Our findings demonstrate an alternative mechanism by which iNOS-induced SNO-JNK increases JNK pathway activity and accelerates cardiac fibrosis. Targeting SNO-JNK might be a novel therapeutic strategy against cardiac fibrosis.


Asunto(s)
Fibrosis/patología , Cardiopatías/patología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Angiotensina II/farmacología , Animales , Aorta/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Humanos , Iminas/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Ratas , Ratas Endogámicas SHR , Transducción de Señal/efectos de los fármacos
5.
Neurochem Int ; 152: 105221, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34780806

RESUMEN

The study aims to investigate whether kaemperfol (KAE) inhibits microglia pyroptosis and subsequent neuroinflammatory response to exert neuroprotective effects, along with the underlying mechanisms. The results showed KAE could ameliorate the behavioral deficits of Parkinson's disease (PD) rats, inhibit the activation of microglia and astrocytes, reduce the loss of TH-positive neurons, down-regulate levels of pyroptosis-related NOD-like receptor family pyrin domain containing 3 (NLRP3), GasderminD-N Term (GSDMD-NT), caspase1, apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC), interleukin (IL)-1ß, and IL-18, and decrease the levels of inflammatory molecules (inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2)) and p38 mitogen-activated protein kinase/nuclear factor-kappaB (p38MAPK/NF-κB) signaling pathway molecules (p38MAPK, p-p38MAPK, NF-κB, and p-NF-κB) in the substantia nigra of PD rats. Further in vitro study indicated that KAE reversed the activation of BV2 cells and down-regulated the expressions of pyrolytic proteins, inflammatory mediators and key molecules in p38MAPK/NF-κB signaling pathway. Collectively, KAE inhibits the microglia pyroptosis and subsequent neuroinflammatory response to exert neuroprotective effects on 6-hydroxydopamine (6-OHDA)-induced PD rats and lipopolysaccharide (LPS)-induced BV2 inflammatory cells through inhibiting p38MAPK/NF-κB signaling pathway.


Asunto(s)
Microglía/efectos de los fármacos , Oxidopamina/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Piroptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Microglía/metabolismo , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Enfermedad de Parkinson/metabolismo , Ratas Sprague-Dawley
6.
Pak J Pharm Sci ; 34(2): 493-498, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34275821

RESUMEN

P. petiolosa as a typical Chinese herbal medicine has been generally utilized as Chinese native medicine formulation for treatment of chronic bronchitis, bronchial asthma and pneumoconiosis. The objective of this study was to evaluate the anti-inflammatory and antibacterial activities of P. petiolosa ethyl acetate extract (PPEAE) against S. aureusin mice. In our study, mice were infected pneumonia by S. aureus, colonization of S. aureus in lung tissue was calculated and the number of white blood cells (WBC) in blood was measured. Meanwhile, the hematoxylin-eosin staining (H&E) was observed and the Real-time PCR was employed to determine the relative mRNA expression. The results showed that, after treated with PPEAE the wet/dry (W/D) weight ratio and the number of WBC decreased dramatically, the number of S. aureus was significantly reduced. Furthermore, H&E staining showed that PPEAE obviously relieved the inflammation of infected mice and real-time PCR results indicated that PPEAE significantly down regulated the inflammatory iNOS, TNF-α and up regulated the anti-inflammatory HO-1 mRNA. In summary, our study revealed that application of crude product PPEAE had prominent antibacterial activity against S. aureus. PPEAE significantly reduced the biomass of S. aureus and effectively relieved the inflammation of S. aureus-induced pneumonia.


Asunto(s)
Antibacterianos/farmacología , Antiinflamatorios/farmacología , Medicamentos Herbarios Chinos/farmacología , Pulmón/efectos de los fármacos , Extractos Vegetales/farmacología , Neumonía Estafilocócica/genética , Polypodiaceae , Staphylococcus aureus/efectos de los fármacos , Animales , Hemo-Oxigenasa 1/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Inflamación/genética , Inflamación/metabolismo , Pulmón/metabolismo , Pulmón/microbiología , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/genética , Ratones , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/genética , Neumonía Estafilocócica/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética
7.
Neurochem Int ; 148: 105100, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34139299

RESUMEN

Nitric oxide generation is related to the activity of certain proteins located at synaptic sites. Previous findings show that NOS activity, nNOS protein expression, respiratory parameters and mitochondrial complex activities are altered in rat cerebral cortex by administration of levocabastine, an antagonist of histamine H1 and neurotensin NTS2 receptors. ATP provision by mitochondria may play an important role in the functional interaction between synaptic proteins NMDA receptor and PSD-95 with NO synthesis. In this context, our purpose was to evaluate the effect of levocabastine administration on protein expression of PSD-95, GluN2B and iNOS, as well as on mitochondrial ATP production. Male Wistar rats received a single (i.p.) dose of levocabastine (50 µg/kg) or saline solution (controls) and were decapitated 18 h later. Mitochondrial and synaptosomal membrane fractions were isolated from cerebral cortex by differential and sucrose gradient centrifugation. Expression of synaptic proteins was evaluated by Western blot assays in synaptosomal membrane fractions. Oxygen consumption, mitochondrial membrane potential and ATP production rate were determined in fresh crude mitochondrial fractions. After levocabastine treatment, protein expression of PSD-95, GluN2B and ß-actin decreased 97, 45 and 55%, respectively, whereas that of iNOS enhanced 3.5-fold versus controls. In crude mitochondrial fractions levocabastine administration reduced roughly 15% respiratory control rate as assayed with malate-glutamate or succinate as substrates, decreased mitochondrial membrane potential (21%), and ATP production rates (57%). Results suggested that levocabastine administration induces alterations in synaptic proteins of the protein complex PSD-95/NMDA receptor/nNOS and in neuron cytoskeleton. Mitochondrial bioenergetics impairment may play a role in the functional link between synaptic proteins and NO synthesis.


Asunto(s)
Homólogo 4 de la Proteína Discs Large/metabolismo , Antagonistas de los Receptores Histamínicos H1/farmacología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Piperidinas/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Adenosina Trifosfato/biosíntesis , Animales , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinaptosomas/efectos de los fármacos
8.
Aging (Albany NY) ; 13(4): 5369-5382, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33536347

RESUMEN

Osteoarthritis (OA) is one of the most common degenerative diseases, ultimately leading to long-term joint pain and severe articular malformation. Controlling local chronic inflammation is a crucial strategy for delaying OA development. Linarin is a natural flavonoid glycoside that is widely available in Compositae, Chrysanthemum indicum and Dendrocalamus and processes protective effects in several animal models. The purpose of our work was to study the protective effect of Linarin for OA. Cellular experiments data showed that Linarin suppressed lipopolysaccharide (LPS)-caused the overproduction of nitric oxide (NO), prostaglandin E2 (PGE2), interleukin-6 (IL-6) and tumour necrosis factor-alpha (TNF-α) in chondrocyte. In addition, LPS-stimulated expression of cyclooxygenase-2 (COX-2) and inducible nitric oxide nitrate (iNOS) was decreased by Linarin pre-treatment. Together, Linarin prevented the catabiosis of extracellular matrix caused by LPS. For mechanism, Linarin inhibited the formation of Toll-like receptor 4 (TLR4) / myeloid differentiation protein-2 (MD-2) dipolymer complex and subsequently intervened NF-κB activation. Our mouse DMM model further clarified the protection of Linarin in vivo. In summary, our results suggested that Linarin may be a potential effective agent for OA.


Asunto(s)
Condrocitos/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Glicósidos/farmacología , Antígeno 96 de los Linfocitos/efectos de los fármacos , Osteoartritis/metabolismo , Receptor Toll-Like 4/efectos de los fármacos , Animales , Condrocitos/metabolismo , Ciclooxigenasa 2/efectos de los fármacos , Ciclooxigenasa 2/genética , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Humanos , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Antígeno 96 de los Linfocitos/metabolismo , Meniscos Tibiales/cirugía , Ratones , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/genética , Osteoartritis/patología , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Neurobiol Dis ; 150: 105244, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33385516

RESUMEN

There is a growing body of evidence demonstrating the significant involvement of the sigma-1 chaperone protein in the modulation of seizures. Several sigma-1 receptor (Sig1R) ligands have been demonstrated to regulate the seizure threshold in acute and chronic seizure models. However, the mechanism by which Sig1R modulates the excitatory and inhibitory pathways in the brain has not been elucidated. The aim of this study was to compare the susceptibility to seizures of wild type (WT) and Sig1R knockout (Sig1R-/-) mice in intravenous pentylenetetrazol (PTZ) and (+)-bicuculline (BIC) infusion-induced acute seizure and Sig1R antagonist NE-100-induced seizure models. To determine possible molecular mechanisms, we used quantitative PCR, Western blotting and immunohistochemistry to assess the possible involvement of several seizure-related genes and proteins. Peripheral tissue contractile response of WT and Sig1R-/- mice was studied in an isolated vasa deferentia model. The most important finding was the significantly decreased expression of the R2 subunit of the GABA-B receptor in the hippocampus and habenula of Sig1R-/- mice. Our results demonstrated that Sig1R-/- mice have decreased thresholds for PTZ- and BIC-induced tonic seizures. In the NE-100-induced seizure model, Sig1R-/- animals demonstrated lower seizure scores, shorter durations and increased latency times of seizures compared to WT mice. Sig1R-independent activities of NE-100 included downregulation of the gene expression of iNOS and GABA-A γ2 and inhibition of KCl-induced depolarization in both WT and Sig1R-/- animals. In conclusion, the results of this study indicate that the lack of Sig1R resulted in decreased expression of the R2 subunit of the GABA-B receptor and increased susceptibility to seizures. Our results confirm that Sig1R is a significant molecular target for seizure modulation and warrants further investigation for the development of novel anti-seizure drugs.


Asunto(s)
Convulsivantes/toxicidad , Habénula/metabolismo , Hipocampo/metabolismo , Receptores de GABA-B/genética , Receptores sigma/genética , Convulsiones/genética , Animales , Anisoles/toxicidad , Bicuculina/toxicidad , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Predisposición Genética a la Enfermedad , Habénula/efectos de los fármacos , Hipocampo/efectos de los fármacos , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/genética , Pentilenotetrazol/toxicidad , Propilaminas/toxicidad , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/genética , Receptores de GABA-B/metabolismo , Convulsiones/inducido químicamente , Receptor Sigma-1
10.
Neuroreport ; 32(4): 296-305, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33470764

RESUMEN

AIM: To evaluate neuroprotective efficacy of fisetin against the experimental model of spinal cord injury (SCI). MATERIALS AND METHODS: SCI was induced in male Sprague-Dawley rats by placing an aneurysm clip extradurally. Rats were treated either with vehicle or fisetin for 28 days after SCI. RESULTS: Treatment with fisetin significantly attenuated SCI-induced alternations in mechano-tactile and thermal allodynia, hyperalgesia and nerve conduction velocities. SCI-induced upregulated tumor necrosis factor-alpha, interleukins, inducible nitric oxide synthase, cyclooxygenase-II, Bcl-2-associated X protein and caspase-3 mRNA expressions in the spinal cord and these were markedly reduced by fisetin. Spinal nuclear factor kappa B and nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor-alpha protein levels were also significantly downregulated by fisetin. Hematoxylin and eosin staining of spinal cord suggested that fisetin significantly ameliorated histological aberrations such as neuronal degeneration, necrosis and inflammatory infiltration induced in it. CONCLUSION: Fisetin exerts neuroprotection via modulation of nuclear factor kappa B/nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor-alpha pathway by inhibiting release of inflammatory mediators (inducible nitric oxide synthase and cyclooxygenase-II), proinflammatory cytokines (tumor necrosis factor-alpha and interleukins), apoptotic mediators (Bcl-2-associated X protein and caspase-3).


Asunto(s)
Flavonoles/farmacología , Inhibidor NF-kappaB alfa/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Traumatismos de la Médula Espinal/metabolismo , Médula Espinal/efectos de los fármacos , Animales , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Ciclooxigenasa 2/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Hiperalgesia/fisiopatología , Locomoción/efectos de los fármacos , Masculino , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/metabolismo , Conducción Nerviosa/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas , Médula Espinal/metabolismo , Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/fisiopatología , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Proteína X Asociada a bcl-2/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
11.
Eur J Pharmacol ; 894: 173874, 2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33460615

RESUMEN

Hyperglycemic milieu in diabetes mellitus stimulates macrophages for exaggerated pro-inflammatory cytokine response, particularly IL-1ß, IL-6, and TNF-α. Although hyperglycemia causes macrophages to produce pro-inflammatory cytokines, AGEs (advanced glycation end products) active inflammation, produced as a result of chronic hyperglycemia, inducers cause polarization of macrophages into pro-inflammatory M1 phenotype. AGEs in diabetes accelerate atherosclerotic plaque initiation and progression via promoting macrophages polarization towards pro-inflammatory state. Gliclazide (Glz) is a well known antidiabetic drug with excellent safety profile. Its repurposing in the management of diabetes-associated late complications has tremendous merit. The present study demonstrated that Glz retards diabetic atherosclerotic progression, and cytokines storm in a concentration dependent manner over a concentration range of 1-100 µM than those of AGEs (200 µg/ml)-treated cells through a mechanism that alters macrophage M1 polarization state. Glz exerted these beneficial effects, independent of its antidiabetic effect. Glz pretreatment significantly (P < 0.05) inhibited the AGEs-induced pro-inflammatory mediators (NO•, reactive oxygen species, i-NOS), and production of pro-inflammatory cytokines, including IL-1ß, IL-6, and TNF-α. It also significantly (P < 0.05) promoted the production of anti-inflammatory cytokines (IL-10 and TGF-ß) in RAW 264.7 mouse macrophages. Glz pretreatment also effectively abated the AGEs-induced RAGE (~2-fold decrease), and CD86 surface marker expressions (P < 0.001 at 100 µM) on macrophages by inhibiting the NF-kß activation in a concentration dependent manner (1-100 µM) (P < 0.001). In conclusion, our data demonstrates that Glz alleviates the diabetic atherosclerosis progression by ameliorating the AGEs-mediated M1 pro-inflammatory phenotype via blocking AGE-RAGE/TLR4-reactive oxygen species -activated NF-kß nexus in macrophages.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Complicaciones de la Diabetes/tratamiento farmacológico , Gliclazida/farmacología , Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Animales , Antígenos de Superficie/efectos de los fármacos , Aterosclerosis/etiología , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Diabetes Mellitus/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Ratones , Subunidad p50 de NF-kappa B/antagonistas & inhibidores , Subunidad p50 de NF-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Células RAW 264.7 , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/antagonistas & inhibidores
12.
J Alzheimers Dis ; 79(1): 211-224, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33252072

RESUMEN

BACKGROUND: Diabetes is one of the strongest disease-related risk factors for Alzheimer's disease (AD). In diabetics, hyperglycemia-induced microvascular complications are the major cause of end-organ injury, contributing to morbidity and mortality. Microvascular pathology is also an important and early feature of AD. The cerebral microvasculature may be a point of convergence of both diseases. Several lines of evidence also implicate thrombin in AD as well as in diabetes. OBJECTIVE: Our objective was to investigate the role of thrombin in glucose-induced brain microvascular endothelial injury. METHODS: Cultured Human brain microvascular endothelial cells (HBMVECs) were treated with 30 mM glucose±100 nM thrombin and±250 nM Dabigatran or inhibitors of PAR1, p38MAPK, MMP2, or MMP9. Cytotoxicity and thrombin activity assays on supernatants and western blotting for protein expression in lysates were performed. RESULTS: reatment of HBMVECs with 30 mM glucose increased thrombin activity and expression of inflammatory proteins TNFα, IL-6, and MMPs 2 and 9; this elevation was reduced by the thrombin inhibitor dabigatran. Direct treatment of brain endothelial cells with thrombin upregulated p38MAPK and CREB, and induced TNFα, IL6, MMP2, and MMP9 as well as oxidative stress proteins NOX4 and iNOS. Inhibition of thrombin, thrombin receptor PAR1 or p38MAPK decrease expression of inflammatory and oxidative stress proteins, implying that thrombin may play a central role in glucose-induced endothelial injury. CONCLUSION: Since preventing brain endothelial injury would preserve blood-brain barrier integrity, prevent neuroinflammation, and retain intact functioning of the neurovascular unit, inhibiting thrombin, or its downstream signaling effectors, could be a therapeutic strategy for mitigating diabetes-induced dementia.


Asunto(s)
Antitrombinas/farmacología , Encéfalo/irrigación sanguínea , Dabigatrán/farmacología , Células Endoteliales/metabolismo , Endotelio Vascular/fisiopatología , Glucosa/toxicidad , Trombina/metabolismo , Enfermedad de Alzheimer/epidemiología , Enfermedad de Alzheimer/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/metabolismo , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Humanos , Inflamación , Interleucina-6/metabolismo , Metaloproteinasa 2 de la Matriz/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , Microvasos/citología , NADPH Oxidasa 4/efectos de los fármacos , NADPH Oxidasa 4/metabolismo , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estrés Oxidativo/efectos de los fármacos , Trombina/efectos de los fármacos , Trombina/farmacología , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Inflamm Res ; 69(12): 1215-1234, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33044562

RESUMEN

OBJECTIVE AND DESIGN: Macrophages exhibit strong phenotypic plasticity and can mediate renal inflammation by polarizing into an M1 phenotype. They play a pivotal role in diabetic nephropathy (DN). Here, we have investigated the regulatory role of transforming growth factor ß-activated kinase 1-binding protein 1 (TAB1) in glycolysis and activation of macrophages during DN. METHODS: TAB1 was inhibited using siRNA in high glucose (HG)-stimulated bone marrow-derived macrophages (BMMs) and lentiviral vector-mediated TAB1 knockdown was used in streptozotocin (STZ)-induced diabetic mice. Western blotting, flow cytometry, qRT-PCR, ELISA, PAS staining and immunohistochemical staining were used for assessment of TAB1/nuclear factor-κB (NF-κB)/hypoxia-inducible factor-1α (HIF-1α), iNOS, glycolysis, inflammation and the clinical and pathological manifestations of diabetic nephropathy. RESULTS: We found that TAB1/NF-κB/HIF-1α, iNOS and glycolysis were up-regulated in BMMs under HG conditions, leading to release of further inflammatory factors, Downregulation of TAB1 could inhibit glycolysis/polarization of macrophages and inflammation in vivo and in vitro. Furthermore, albuminuria, the tubulointerstitial damage index and glomerular mesangial expansion index of STZ-induced diabetic nephropathy mice were decreased by TAB1 knockdown. CONCLUSIONS: Our results suggest that the TAB1/NF-κB/HIF-1α signaling pathway regulates glycolysis and activation of macrophages in DN.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neuropatías Diabéticas/genética , Neuropatías Diabéticas/metabolismo , Glucólisis/genética , Activación de Macrófagos/genética , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Albuminuria/tratamiento farmacológico , Albuminuria/patología , Animales , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Técnicas de Silenciamiento del Gen , Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Células Mesangiales/patología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/efectos de los fármacos , Nefritis Intersticial/patología , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , ARN Interferente Pequeño/farmacología
14.
Pharmacology ; 105(9-10): 505-513, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32784309

RESUMEN

OBJECTIVE: This research was aimed to explore protective effects of allicin on rat model of myocardial infarction via JNK signaling pathway. METHODS: Rat myocardial ischemia model was established with subcutaneous injection of isoproterenol (ISO). Seventy-five rats were randomly divided into 5 groups (n = 15): sham group, ISO group, low-dose group (1.2 mg/kg/days for 7 days), medium-dose group (1.8 mg/kg/days for 7 days), and high-dose group (3.6 mg/kg/days for 7 days). Routine HE staining and Masson staining were performed to observe myocardial histopathology. The expression of oxidative stress-related indicators, heart tissue apoptosis-related proteins, and JNK and p-JNK proteins were measured for different groups. RESULTS: Compared with the sham group, the T wave value of the ISO group was significantly increased (p < 0.01). When allicin was administered, the T wave values at different time points in all groups were all decreased. Compared with the sham group, the ratio of eNOS, Bcl-2/Bax was significantly decreased, and p-eNOS, iNOS, caspase-3, caspase-9, and Cyt-c were significantly elevated in the ISO group (p < 0.05). After allicin was administered, significant changes in these proteins were observed in the medium- and high-dose groups. There was no significant change in the expression of JNK protein in the ISO group compared with the sham group; however, the expression of eNOS and p-JNK protein were significantly upregulated (p < 0.01) and the expression of p-eNOS and iNOS were significantly downregulated (p < 0.01). When allicin was administered, expression of p-JNK protein was significantly downregulated. CONCLUSION: Allicin can reduce oxidative stress damage and cardiomyocyte apoptosis in rat model of myocardial infarction and can significantly regulate JNK signaling pathway.


Asunto(s)
Antioxidantes/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Ácidos Sulfínicos/farmacología , Animales , Antioxidantes/uso terapéutico , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Disulfuros , Isoproterenol/toxicidad , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Infarto del Miocardio/inducido químicamente , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Ácidos Sulfínicos/uso terapéutico
15.
J Nat Prod ; 83(6): 1740-1750, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32496797

RESUMEN

Bitter taste receptors (hTAS2R) are expressed ectopically in various tissues, raising the possibility of a pharmacological exploitation. This seems of particular relevance in airways, since hTAS2Rs are involved in the protection of the aerial tissues from infections and in bronchodilation. The bis-guaianolide absinthin (1), one of the most bitter compounds known, targets the hTAS2R46 bitter receptor. Absinthin (1), an unstable compound, readily turns into anabsinthin (2) with substantial retention of the bitter properties, and this compound was used as a starting material to explore the chemical space around the bis-guaianolide bitter pharmacophore. Capitalizing on the chemoselective opening of the allylic lactone ring, the esters 3 and 4, and the nor-azide 6 were prepared and assayed on human bronchoepithelial (BEAS-2B) cells expressing hTAS2R46. Anti-inflammatory activity was evaluated by measuring the expression of MUC5AC, iNOS, and cytokines, as well as the production of superoxide anion, qualifying the methyl ester 3 as the best candidate for additional studies.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Bronquios/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Sesquiterpenos de Guayano/farmacología , Artemisia/química , Bronquios/citología , Calcio/metabolismo , Línea Celular , Citocinas/antagonistas & inhibidores , Ésteres/química , Humanos , Estructura Molecular , Mucina 5B/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Superóxidos/metabolismo , Papilas Gustativas
16.
Biochim Biophys Acta Mol Basis Dis ; 1866(10): 165850, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32497615

RESUMEN

Abnormal wound healing with excessive scarring is a major health problem with socioeconomic and psychological impacts. In human, chronic wounds and scarring are associated with upregulation of the inducible nitric oxide synthase (iNOS). Recently, we have shown physiological regulation of iNOS in wound healing. Here, we sought to investigate the possible mechanistic role of iNOS in wound healing using biochemical and immunohistochemical assays. We found: (a) iNOS is the main source of wound nitric oxide (NO), (b) NOS inhibition in the wound, downregulated iNOS protein, mRNA and enzymatic activity, and reduced wound NO, and (c) iNOS inhibition resulted in delayed healing at early time points, and excessive scarring at late time points. Furthermore, molecular and cellular analysis of the wound showed that iNOS inhibition significantly (P < 0.05) increased TGF-ß1 mRNA and protein levels, fibroblasts and collagen deposition. These latter findings suggest that iNOS might be exerting its action in the wound by signaling through TGF-ß1 that activates wound fibroblasts to produce excessive collagen. Our current findings provide further support that iNOS is crucial for physiological wound healing, and suggest that dysregulation of iNOS during the inflammatory phase impairs healing, and results in disfiguring post-healing scarring. Thus, the mutual feedback regulation between iNOS and TGF-ß1 at the gene, protein and functional levels might be the mechanism through which iNOS regulates the healing. Monitoring and maintenance of wound NO levels might be important for healing and avoiding long-term complications in susceptible people including patients with diabetic wounds, venous ulcers or keloid prone.


Asunto(s)
Óxido Nítrico Sintasa de Tipo II/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Cicatrización de Heridas/fisiología , Animales , Arginasa/metabolismo , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Guanidinas/farmacología , Humanos , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Piel/metabolismo , Piel/patología
17.
Int Immunopharmacol ; 86: 106576, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32575007

RESUMEN

Osmanthus fragrans var. aurantiacus has been used for the treatment of menopausal pain, foul breath, and intestinal bleeding. Four phenylpropyl triterpenoids, 3-O-trans-p-coumaroyltormentic acid (1), 3ß-trans-p-coumaroyloxy-2α-hydroxyl-urs-12-en-28-oic acid (2), 3ß-cis-p-coumaroyloxy-2α-hydroxyl-urs-12-en-28-oic acid (3), 3-O-cis-coumaroylmaslinic acid (4), were isolated from the leaves of O. fragrans var. aurantiacus and the inhibitory effect on nitric oxide (NO) production in lipopolysaccharide (LPS)-induced macrophages were evaluated. Among them, compounds 2-4 concentration dependently showed NO production inhibitory effects. To determine the signaling factors involved in the inhibition of NO production by compounds 2-4, we assessed anti-inflammatory activity. Western blot analysis revealed compounds 2-4 significantly decreased the expression of LPS-stimulated protein, inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, nuclear factor-kappa B (NF-κB) and phosphorylated extracellular regulated kinase (pERK)1/2. Also, compounds 2-4 downregulated tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6 and IL-8 levels in LPS-induced macrophages and colonic epithelial cells. This study demonstrated that phenylpropyl triterpenoids 2-4 isolated from O. fragrans var. aurantiacus leaves can be used as potential candidates for the prevention and treatment of inflammatory bowel disease.


Asunto(s)
Antiinflamatorios/farmacología , Oleaceae/química , Triterpenos/farmacología , Animales , Antiinflamatorios/química , Antiinflamatorios/aislamiento & purificación , Supervivencia Celular/efectos de los fármacos , Ciclooxigenasa 2/efectos de los fármacos , Citocinas/efectos de los fármacos , Células HT29 , Humanos , Lipopolisacáridos/toxicidad , Ratones , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Subunidad p50 de NF-kappa B/efectos de los fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Hojas de la Planta/química , Células RAW 264.7 , Triterpenos/química , Triterpenos/aislamiento & purificación
18.
Neurosci Lett ; 728: 134952, 2020 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-32283112

RESUMEN

Nitric oxide has been implicated in symptoms of ethanol withdrawal in animal models. Zebrafish have been used as models to study neurobehavioral effects of ethanol (EtOH) withdrawal, but the mechanisms associated with these effects are not yet clear. Adult zebrafish were treated with 1% EtOH for 20 min per day for 8 days, injected with the nitric oxide synthase 2 (NOS-2) inhibitor aminoguanidine (50 mg/kg), and allowed to experience withdrawal (WD) in their hometanks for 7 days. EtOH WD increased anxiety-like behavior in the novel tank test, an effect that was blocked by aminoguanidine. EtOH WD also increased brain levels of nitrite, an effect that was partially blocked by aminoguanidine. These results underline a novel mechanism by which NOS-2 controls anxiety-like responses to ethanol withdrawal, with implications for the mechanistic study of symptoms associated with chronic ethanol abuse. Preprint: https://dx.doi.org/10.20944/preprints201912.0219.v1 Data and scripts: https://github.com/lanec-unifesspa/etoh-withdrawal/tree/master/NOS2.


Asunto(s)
Alcoholismo/metabolismo , Etanol/farmacología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Animales , Ansiedad/tratamiento farmacológico , Conducta Animal , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Pez Cebra/metabolismo
19.
Int J Neurosci ; 130(2): 176-185, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31524564

RESUMEN

Aim: The objective of the current investigation was to explore the analgesic effect of naturally occurring furanocoumarin, imperatorin and the involvement of inducible cyclooxygenase (COX-2), inducible nitric oxide synthase (iNOS), NFκB and cytokines in the observed effect.Materials and methods: Anti-nociceptive effect was explored by inducing chemical hyperalgesia using acetic acid and formalin in mice. ED50 of imperatorin was calculated in acetic acid model. Modulation of cyclooxygenase and nitric oxide pathway by imperatorin was examined by stimulator/precursor challenge with substance P and L-arginine, respectively and quantification of COX-2, iNOS and NFκB expression by immunohistochemical analysis in spinal tissues. Involvement of inflammatory cytokines TNF-α and IL-1ß was investigated using LPS challenge and subsequent ELISA analysis of these inflammatory mediators in serum. Carrageenan inflicted paw edema was employed to explore the anti-inflammatory activity of imperatorin.Results: A significant reduction in the nociceptive behaviour was observed with imperatorin treatment in acetic acid and formalin test. ED50 of imperatorin was found to be 4.53 mg/kg. Pre-treatment with substance P and L-arginine significantly attenuated the anti-nociceptive activity of imperatorin in formalin test. Immunohistochemical findings revealed marked decrease in spinal COX-2, iNOS and NFκB expression. Imperatorin administration significantly reduced LPS induced rise in level of TNF-α and IL-1ß dose dependently. In carrageenan-induced paw edema test, maximum possible anti-inflammatory effect of imperatorin was evident after 240 min of carrageenan administration.Conclusion: Current investigation revealed that anti-nociceptive and anti-inflammatory potential of imperatorin is probably mediated through the attenuation of COX-2, iNOS, NFκB activity and reduction in circulatory cytokines.


Asunto(s)
Analgésicos/farmacología , Antiinflamatorios/farmacología , Ciclooxigenasa 2/efectos de los fármacos , Furocumarinas/farmacología , Hiperalgesia/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Interleucina-18 , FN-kappa B/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Ratones
20.
Mol Neurobiol ; 57(2): 1085-1098, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31677009

RESUMEN

Growing evidence suggests that docosahexaenoic acid (DHA) exerts neuroprotective effects, although the mechanism(s) underlying these beneficial effects are not fully understood. Here we demonstrate that DHA, but not arachidonic acid (ARA), suppressed oligomeric amyloid-ß peptide (oAß)-induced reactive oxygen species (ROS) production in primary mouse microglia and immortalized mouse microglia (BV2). Similarly, DHA but not ARA suppressed oAß-induced increases in phosphorylated cytosolic phospholipase A2 (p-cPLA2), inducible nitric oxide synthase (iNOS), and tumor necrosis factor-α (TNF-α) in BV2 cells. LC-MS/MS assay indicated the ability for DHA to cause an increase in 4-hydroxyhexenal (4-HHE) and suppress oAß-induced increase in 4-hydroxynonenal (4-HNE). Although oAß did not alter the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, exogenous DHA, ARA as well as low concentrations of 4-HHE and 4-HNE upregulated this pathway and increased production of heme oxygenase-1 (HO-1) in microglial cells. These results suggest that DHA modulates ARA metabolism in oAß-stimulated microglia through suppressing oxidative and inflammatory pathways and upregulating the antioxidative stress pathway involving Nrf2/HO-1. Understanding the mechanism(s) underlying the beneficial effects of DHA on microglia should shed light into nutraceutical therapy for the prevention and treatment of Alzheimer's disease (AD).


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Microglía/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Ácido Araquidónico/metabolismo , Ácido Araquidónico/farmacología , Ratones Endogámicos C57BL , Microglía/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Espectrometría de Masas en Tándem/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA