Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Heliyon ; 9(4): e14979, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37064439

RESUMO

Neurodegenerative disorders are known to be associated with neuroinflammation caused by microglia. Therefore, regulation of microglia activation and polarization to inhibit neuroinflammatory reactions seems to hold promise as a therapeutic approach in neurodegenerative disorders. Spatholobus suberectus Dunn (SSD) has been utilized as a traditional Chinese medicine remedy for brain diseases for thousands of years. SSD possesses various pharmacological activities, such as circulation invigoration, neuroprotection, and anti-inflammatory. The objective of this research was to examine the anti-neuroinflammatory effects and molecular mechanisms of an active fraction from SSD (ASSD) in vitro culture BV2 cells, a type of mouse microglia cell line. The inflammatory responses in BV2 cells were induced by stimulating them with 1 µg/mL lipopolysaccharide (LPS) and the effects of ASSD on LPS-stimulated inflammation were monitored. Besides, by using the methods of Western blot, immunofluorescence, and RT-PCR, the mechanisms of ASSD on microglia activation, M1/M2 polarization, and the TLR4/MyD88/NF-κB pathway were investigated. Our findings demonstrate that the treatment doses of ASSD neither induce cytotoxicity nor promote the production of inflammatory cytokines. In addition, immunofluorescence analysis show that ASSD inhibited the expression of ionized calcium-binding adapter molecule 1(Iba1) and inducible nitricoxide synthase (iNOS), and induced arginase 1 (Arg1) expression. Moreover, Western blot analysis indicated that ASSD significantly down-regulated TLR4, MyD88, p-IκB, NF-κB p65, and NF-κB p-p65 protein expression levels. Furthermore, RT-qPCR assay show that ASSD significantly down-regulated iNOS, TLR4, MyD88, and NF-κB mRNA expression levels, and up-regulated Arg1 mRNA expression level. According to the findings, ASSD can suppress microglia-mediated inflammatory responses by modulating microglia activation, inducing a shift from M1 to M2 polarization, and inhibiting the TLR4/MyD88/NF-κB signaling pathway.

2.
Food Funct ; 12(12): 5668, 2021 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-34019057

RESUMO

Correction for 'Yulangsan polysaccharide improves redox homeostasis and immune impairment in d-galactose-induced mimetic aging' by Van Minh Doan et al., Food Funct., 2015, 6, 1712-1718, DOI: 10.1039/C5FO00238A.

3.
Mol Med Rep ; 22(5): 4254-4264, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32901878

RESUMO

Our previous study demonstrated that hyperbaric oxygen (HBO) improves heart function predominantly through reducing oxygen stress, modulating energy metabolism and inhibiting cell apoptosis. The present study aimed to investigate the protective effects of HBO on mitochondrial function and autophagy using rats with a ligated left anterior descending artery. The cardioprotective effects of HBO were mainly evaluated using ELISA, fluorescent probes, transmission electron microscopy and reverse transcription­quantitative PCR (RT­qPCR). HBO pretreatment for 14 days (once a day) using a 0.25 MPa chamber improved mitochondrial morphology and decreased the number of autophagic vesicles, as observed using a transmission electron microscope. HBO pretreatment significantly increased the levels of ATP, ADP, energy charge and the opening of the mitochondrial permeability transition pore, but decreased the levels of AMP, cytochrome c and reactive oxygen species. Moreover, HBO pretreatment significantly increased the gene or protein expression levels of eIF4E­binding protein 1, mammalian target of rapamycin (mTOR), mitochondrial DNA, NADH dehydrogenase subunit 1, mitofusin 1 and mitofusin 2, whereas it decreased the gene or protein expression levels of autophagy­related 5 (Atg5), cytochrome c, dynamin­related protein 1 and p53, as determined using RT­qPCR or immunohistochemistry. In conclusion, HBO treatment was observed to protect cardiomyocytes during myocardial ischemia­reperfusion injury (MIRI) by preventing mitochondrial dysfunction and inhibiting autophagy. Thus, these results provide novel evidence to support the use of HBO as a potential agent for the mitigation of MIRI.


Assuntos
Oxigenoterapia Hiperbárica/métodos , Mitocôndrias/metabolismo , Traumatismo por Reperfusão Miocárdica/terapia , Animais , Autofagia , Modelos Animais de Doenças , Metabolismo Energético , Regulação da Expressão Gênica , Masculino , Traumatismo por Reperfusão Miocárdica/metabolismo , Ratos , Resultado do Tratamento
4.
Life Sci ; 241: 117170, 2020 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-31838137

RESUMO

AIMS: In this study, we investigate the effect and underlying mechanism of hyperbaric oxygen (HBO) treatment on a model of repeated cerebral ischemia-reperfusion injury (IR). MAIN METHODS: Eighty rats were randomly separated into sham, vehicle, hyperbaric air (HBA; 0.25 MPa, 60 min), and HBO (0.25 MPa, 60 min) groups. Repeated cerebral IR was induced by ligating the right and left bilateral common carotid arteries for 10 min and then allowing reperfusion for 10 min. This pattern was repeated three times. The neuroprotective effects of HBO were assessed by animal behavior, neuron morphology, inflammatory markers, intracellular calcium ion content, and autophagy-related protein and gene expression. KEY FINDINGS: Our result showed that HBO improved learning and memory in the navigation trail and probe trail of the Morris water maze, and these findings were supported by the observation data from 2,3,5-Triphenyltet-razolium chloride staining, Nissl staining, and electron microscopic. Importantly, we found that HBO reduced excessive autophagy in the prefrontal cortex, which was evidenced by activating of the mammalian target of the rapamycin (mTOR) and 4E-BP1, as well as suppression of LC3II and ATG5. Moreover, HBO significantly inhibited the cerebral IR-induced inflammatory reaction. Furthermore, HBO treatment modulated autophagy pathway-related factors, including producing a decrease in the intracellular calcium ion concentration and p53 level; meanwhile, the levels of BDNF and p-Akt were increased. SIGNIFICANCE: Our results indicated that HBO protected against IR-induced neuron injury by attenuating autophagy, inflammation, and calcium overload. These results provide a new mechanism and laboratory evidence for clinical treatment of VD.


Assuntos
Autofagia , Isquemia Encefálica/complicações , Disfunção Cognitiva/prevenção & controle , Modelos Animais de Doenças , Oxigenoterapia Hiperbárica/métodos , Fármacos Neuroprotetores , Traumatismo por Reperfusão/complicações , Animais , Comportamento Animal , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/patologia , Feminino , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley
5.
Med Chem ; 15(5): 459-471, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30569869

RESUMO

Hyperbaric Oxygenation Therapy (HBOT) is used as an adjunctive method for multiple diseases. The method meets the routine treating and is non-invasive, as well as provides 100% pure oxygen (O2), which is at above-normal atmospheric pressure in a specialized chamber. It is well known that in the condition of O2 deficiency, it will induce a series of adverse events. In order to prevent the injury induced by anoxia, the capability of offering pressurized O2 by HBOT seems involuntary and significant. In recent years, HBOT displays particular therapeutic efficacy in some degree, and it is thought to be beneficial to the conditions of angiogenesis, tissue ischemia and hypoxia, nerve system disease, diabetic complications, malignancies, Carbon monoxide (CO) poisoning and chronic radiation-induced injury. Single and combination HBOT are both applied in previous studies, and the manuscript is to review the current applications and possible mechanisms of HBOT. The applicability and validity of HBOT for clinical treatment remain controversial, even though it is regarded as an adjunct to conventional medical treatment with many other clinical benefits. There also exists a negative side effect of accepting pressurized O2, such as oxidative stress injury, DNA damage, cellular metabolic, activating of coagulation, endothelial dysfunction, acute neurotoxicity and pulmonary toxicity. Then it is imperative to comprehensively consider the advantages and disadvantages of HBOT in order to obtain a satisfying therapeutic outcome.


Assuntos
Oxigenoterapia Hiperbárica , Animais , Doenças Cardiovasculares/terapia , Humanos , Hipóxia/terapia , Isquemia/terapia , Neovascularização Fisiológica/fisiologia , Doenças do Sistema Nervoso/terapia
6.
Kidney Blood Press Res ; 43(1): 220-233, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29490297

RESUMO

BACKGROUND/AIMS: The objective of this study is to evaluate the hypouricemic and nephroprotective effects of an active fraction from Polyrhachis vicina Roger (AFPR) in potassium oxonate-induced hyperuricemic rats. METHODS: Hyperuricemia was induced by potassium oxonate in male rats. AFPR was orally administered to hyperuricemic rats for 12 consecutive weeks. Serum, liver and kidney samples were collected for effects and mechanism analysis. The levels of serum uric acid (SUA) were measured by the phosphotungstic acid method, xanthine oxidase (XOD) activity in the hepatic and serum samples were measured by ultraviolet spectrophotometry, serum levels of interleukin-1 (IL-1ß), interleukin-1 (IL-6) and tumor necrosis factor-α (TNF-α) were measured by ELISA, the levels of serum creatinine (SCr), blood urea nitrogen (BUN), super oxide dismutase (SOD) and malondialdehyde (MDA) in serum were determined by colorimetric method. Protein expression of renal URAT1, GLUT9, and OAT1 were analyzed by Western blot. RESULTS: AFPR significantly decreased the levels of SUA, serum and hepatic XOD, SCr, BUN, and MDA as well as increased SOD. In addition, AFPR treatment significantly reduced the levels of proinflammatory cytokines in serum, including IL-1ß, IL-6 and TNF-α. Moreover, we found the significant decrease in protein expression of URAT1 and GLUT9, and the significant increase in protein expression of OAT1 in the kidney in AFPR treated groups compared to the model groups of hyperuricemia. CONCLUSION: These findings suggest that AFPR has anti-hyperuricemic activity attributed to the inhibition of uric acid generation in the liver and probably to the enhancement of urate excretion in the kidney, and possess nephroprotective effect in hyperuricemic rats due to its anti-inflammatory and antioxidant activities.


Assuntos
Formigas/química , Hiperuricemia/tratamento farmacológico , Ácido Oxônico/efeitos adversos , Substâncias Protetoras/farmacologia , Animais , Anti-Inflamatórios , Antioxidantes , Hiperuricemia/induzido quimicamente , Rim/metabolismo , Fígado/metabolismo , Masculino , Substâncias Protetoras/isolamento & purificação , Ratos , Ácido Úrico/antagonistas & inibidores
7.
Neurochem Res ; 43(4): 918-929, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29455417

RESUMO

We previously reported that Yulangsan polysaccharide (YLSP), which was isolated from the root of Millettia pulchra Kurz, attenuates withdrawal symptoms of morphine dependence by regulating the nitric oxide pathway and modulating monoaminergic neurotransmitters. In this study, we investigated the effects and mechanism of YLSP on the reinstatement of morphine-induced conditioned place preference (CPP) in rats. A CPP procedure was employed to assess the behavior of rats, and indicators of serum and four brain regions (nucleus accumbens, ventral tegmental area, hippocampus and prefrontal cortex) were determined to explore its underlying mechanism. YLSP inhibited priming morphine-induced reinstatement of CPP in a dose-dependent manner. YLSP markedly reduced nitric oxide and nitric oxide synthase levels in the brain. Moreover, YLSP significantly decreased the dopamine and norepinephrine levels in the serum and brain. Furthermore, YLSP significantly decreased cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) concentrations, inhibited the expression of dopamine D1 receptors and cAMP response element binding protein mRNA, and improved the expression of dopamine D2 receptor mRNA in the four brain regions. Our findings indicated that YLSP could inhibit the reinstatement of morphine-induced CPP possibly by modulating the NO-cGMP and D1R-cAMP signaling pathways.


Assuntos
Condicionamento Clássico/efeitos dos fármacos , Millettia , Dependência de Morfina/tratamento farmacológico , Morfina/administração & dosagem , Extratos Vegetais/farmacologia , Polissacarídeos/farmacologia , Animais , Condicionamento Clássico/fisiologia , Relação Dose-Resposta a Droga , Masculino , Dependência de Morfina/metabolismo , Dependência de Morfina/psicologia , Extratos Vegetais/isolamento & purificação , Extratos Vegetais/uso terapêutico , Polissacarídeos/isolamento & purificação , Polissacarídeos/uso terapêutico , Ratos , Ratos Sprague-Dawley
8.
Neurochem Res ; 43(3): 531-539, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29313212

RESUMO

In this study, we examined whether hyperbaric oxygen (HBO2) plays a detoxification role in withdrawal symptoms in a morphine-dependent rat model. The model was established through injections of morphine at increasing doses for 7 days. Withdrawal symptoms were induced by naloxone injection on the 8th day. The detoxification effect of HBO2 was evaluated using the withdrawal symptom scores, biochemical indices and neurotransmitters. Compared with the model group, HBO2 therapy significantly attenuated the withdrawal symptom scores, body weight loss and the level of norepinephrine level, whereas it increased the dopamine level and tyrosine hydroxylase expression in the nucleus accumbens. Moreover, HBO2 therapy substantially alleviated the NO, NOS, cAMP, and cGMP levels. Our findings indicate that HBO2 can effectively alleviate withdrawal symptoms induced by morphine dependence, and these effects may be attributed to the modulation of monoaminergic neurotransmitters and the suppression of the NO-cGMP signaling pathway.


Assuntos
Oxigenoterapia Hiperbárica , Morfina/farmacologia , Neurotransmissores/metabolismo , Óxido Nítrico/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Animais , Masculino , Dependência de Morfina/metabolismo , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Núcleo Accumbens/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/tratamento farmacológico
9.
Sci Rep ; 7(1): 6704, 2017 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-28751740

RESUMO

Metastasis causes approximately 90% of breast cancer-related deaths in women. Previously, we have demonstrated that 2-dodecyl-6-methoxycyclohexa-2,5-diene- 1,4-dione (DMDD) remarkably inhibited the growth of human breast cancer cells with little toxicity. In this study, we investigated the toxicity and efficacy of DMDD to treat metastatic breast tumors using an in vivo mouse model of the 4T1 mammary carcinoma. DMDD caused no observable toxicity and significantly extended the survival of 4T1 tumor-bearing mice. DMDD effectively inhibited the growth of 4T1 cells in vitro, and suppressed the growth and metastasis of mammary tumor in vivo. The levels of inflammatory cytokines in the serum (TNF-α, IL-6, IL-12, TGF-ß, and VEGF) were down regulated by DMDD. Immunohistochemical analysis demonstrated that the inhibition of tumor growth and metastasis was associated with activation of Bax, cleaved caspases-3 and -9, and down-regulation of Bcl-2, MMP-2 and -9, NF-κB and IκBα. We speculate that DMDD inhibits cytokine production in the tumor cells in mice, which leads to deactivation of NF-κB pathway, and consequently inhibits the expression of many anti-apoptosis and metastasis-promoting genes, such as Bcl-2 and MMPs. Collectively, our results demonstrate the potential of DMDD as a safe and effective antitumor agent in the treatment of late-stage breast cancer.


Assuntos
Cicloexenos/farmacologia , Neoplasias Mamárias Animais/patologia , Animais , Apoptose/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cicloexenos/efeitos adversos , Citocinas/sangue , Citocinas/metabolismo , Feminino , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Animais/sangue , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Fosforilação/efeitos dos fármacos , Análise de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Curr Top Med Chem ; 17(21): 2440-2455, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28474551

RESUMO

Addiction is a chronic, relapsing disease of the brain that includes drug-induced compulsive seeking behavior and consumption of drugs. Dopamine (DA) is considered to be critical in drug addiction due to reward mechanisms in the midbrain. In this article, we review the major animal models in addictive drug experiments in vivo and in vitro. We discuss the relevance of the structure and pharmacological function of DA receptors. To improve the understanding of the role of DA receptors in reward pathways, specific brain regions, including the Ventral tegmental area, Nucleus accumbens, Prefrontal cortex, and Habenula, are highlighted. These factors contribute to the development of novel therapeutic targets that act at DA receptors. In addiction, the development of neuroimaging method will increase our understanding of the mechanisms underlying drug addiction.


Assuntos
Comportamento Aditivo , Encéfalo/metabolismo , Dopamina/metabolismo , Receptores Dopaminérgicos/metabolismo , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Animais , Agonistas de Dopamina/química , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/química , Antagonistas de Dopamina/farmacologia , Recompensa
11.
Neurochem Res ; 42(4): 1240-1253, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28078611

RESUMO

Our previous study demonstrated that hyperbaric oxygen (HBO) improved cognitive impairments mainly by regulating oxidative stress, inflammatory responses and aging-related gene expression. However, a method for preventing cognitive dysfunction has yet to be developed. In the present study, we explored the protective effects of HBO on the cholinergic system and apoptosis in D-galactose (D-gal)-treated mice. A model of aging was established via systemic intraperitoneal injection of D-gal daily for 8 weeks. HBO was administered during the last 2 weeks of D-gal injection. Our results showed that HBO in D-gal-treated mice significantly improved behavioral performance on the open field test and passive avoidance task. Studies on the potential mechanisms of this effect showed that HBO significantly reduced oxidative stress and blocked the nuclear factor-κB pathway. Moreover, HBO significantly increased the levels of choline acetyltransferase and acetylcholine and decreased the activity of acetylcholinesterase in the hippocampus. Furthermore, HBO markedly increased expression of the anti-apoptosis protein Bcl-2 and glial fibrillary acidic protein meanwhile decreased expression of the pro-apoptosis proteins Bax and caspase-3. Importantly, there was a significant reduction in expression of Aß-related genes, such as amyloid precursor protein, ß-site amyloid cleaving enzyme-1 and cathepsin B mRNA. These decreases were accompanied by significant increases in expression of neprilysin and insulin-degrading enzyme mRNA. Moreover, compared with the Vitamin E group, HBO combined with Vitamin E exhibited significant difference in part of the above mention parameters. These findings suggest that HBO may act as a neuroprotective agent in preventing cognitive impairments.


Assuntos
Apoptose/fisiologia , Neurônios Colinérgicos/metabolismo , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/prevenção & controle , Galactose/toxicidade , Oxigenoterapia Hiperbárica/métodos , Animais , Apoptose/efeitos dos fármacos , Neurônios Colinérgicos/efeitos dos fármacos , Disfunção Cognitiva/induzido quimicamente , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Camundongos , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Distribuição Aleatória , Vitamina A/farmacologia , Vitamina A/uso terapêutico
12.
Oncotarget ; 8(67): 111522-111534, 2017 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-29340072

RESUMO

Our previous study demonstrated that hyperbaric oxygen (HBO) preconditioning protected against myocardial ischemia reperfusion injury (MIRI) and improved myocardial infarction. However, HBO's effect on MIRI-induced inflammation and autophagy remains unclear. In this study, we investigate the potential impact and underlying mechanism of HBO preconditioning on an MIRI-induced inflammatory response and autophagy using a ligation of the left anterior descending (LAD) coronary artery rat model. Our results showed that HBO restored myocardial enzyme levels and decreased the apoptosis of cardiomyocytes, which were induced by MIRI. Moreover, HBO significantly suppressed MIRI-induced inflammatory cytokines. This effect was associated with the inhibition of the TLR4-nuclear factor kappa-B (NF-κB) pathway. Interestingly, lower expression levels of microtubule-associated protein 1 light chain 3B (LC3B) and Beclin-1 were observed in the HBO-treatment group. Furthermore, we observed that HBO reduced excessive autophagy by activating the mammalian target of the rapamycin (mTOR) pathway, as evidenced by higher expression levels of threonine protein kinase (Akt) and phosphorylated-mTOR. In conclusion, HBO protected cardiomocytes during MIRI by attenuating inflammation and autophagy. Our results provide a new mechanistic insight into the cardioprotective role of HBO against MIRI.

13.
Neurochem Res ; 41(11): 3032-3041, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27485714

RESUMO

Memory decline is characteristic of aging and age-related neurodegenerative disorders. This study was designed to investigate the protective effect of hyperbaric oxygen (HBO) against cognitive impairment induced by D-galactose (D-gal) in mice. D-gal was intraperitoneally injected into mice daily for 8 weeks to establish the aging model. HBO was simultaneously administered once daily. The results indicate that HBO significantly reversed D-gal-induced learning and memory impairments. Studies on the potential mechanisms of this action showed that HBO significantly reduced oxidative stress by increasing superoxide dismutase, glutathione peroxidase, and catalase levels, as well as the total anti-oxidation capability, while decreasing the content of malondialdehyde, nitric oxide, and nitric oxide synthase in the hippocampal CA1 region. HBO also inhibited advanced glycation end-product formation and decreased levels of tumor necrosis factor-α and interleukin-6. Moreover, HBO significantly attenuated D-gal-induced pathological injury in the hippocampus, as well as ß-amyloid protein1-42 expression and retained BDNF expression. Furthermore, HBO decreased p16, p21 and p53 gene and protein expression in the hippocampus of D-gal-treated mice. In conclusion, the protective effect of HBO against D-gal-induced cognitive impairment was mainly due to its ability to reduce oxidative damage, suppress inflammatory responses, and regulate aging-related gene expression.


Assuntos
Transtornos Cognitivos/tratamento farmacológico , Galactose/farmacologia , Hipocampo/efeitos dos fármacos , Memória/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Envelhecimento/metabolismo , Animais , Transtornos Cognitivos/induzido quimicamente , Modelos Animais de Doenças , Hipocampo/metabolismo , Oxigenoterapia Hiperbárica/métodos , Masculino , Camundongos , Estresse Oxidativo/efeitos dos fármacos
14.
Cell Physiol Biochem ; 38(5): 2015-29, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27160091

RESUMO

BACKGROUND/AIMS: In this study, we examined whether the combination of hyperbaric oxygen (HBO) and diltiazem therapy provided a cardioprotective effect on myocardial ischemia-reperfusion injury (MIRI) rat model. METHODS: Sixty healthy Sprague-Dawley rats were randomly divided into sham, IR, diltiazem (5 mg/kg), HBO (0.25 MPa, 60 min) and combination therapy (HBO plus diltiazem) groups. MIRI model was established by ligating the left anterior descending for 30 min, followed by 60 min of reperfusion. RESULTS: The results show that HBO and diltiazem preconditioning significantly improves cardiac function and myocardial infarction area, increases nitric oxide, endothelial nitric oxide synthase and ATPase (Na+-K+-ATPase and Ca2+-Mg2+-ATPase) activity and decreases levels of oxygen stress, myocardial enzymes and endothelin-1. Notably, HBO and diltiazem preconditioning significantly increased Bcl-2 protein expression and decreased Bax protein and caspase-3 mRNA expression. CONCLUSIONS: These data indicate that combination therapy protected against heart MIRI by reducing oxygen stress damage, correcting energy metabolism, improving endothelial function and inhibiting cell apoptosis.


Assuntos
Diltiazem/uso terapêutico , Oxigenoterapia Hiperbárica , Traumatismo por Reperfusão Miocárdica/terapia , Substâncias Protetoras/uso terapêutico , Animais , Caspase 3/genética , Caspase 3/metabolismo , Terapia Combinada , Diltiazem/farmacologia , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Endotelina-1/genética , Endotelina-1/metabolismo , Feminino , Coração/diagnóstico por imagem , Precondicionamento Isquêmico , Masculino , Infarto do Miocárdio , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Substâncias Protetoras/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Sprague-Dawley , ATPase Trocadora de Sódio-Potássio/metabolismo , Regulação para Cima/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo
15.
Environ Toxicol Pharmacol ; 41: 1-7, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26645132

RESUMO

Although numerous studies have proven the medicinal values of Yulangsan polysaccharide (YLSP), the toxicity of this active ingredient is unknown. In the acute toxicity study, a single oral administration of 24 g/kg YLSP caused neither toxicological symptoms nor mortality, and the LD50 was estimated >24 g/kg. In the chronic toxicity study, we administered doses of 0, 0.6, 1.2 and 2.4 g/kg YLSP in rats by oral gavage for 26 weeks followed by a 3-week recovery period. There was no mortality or remarkable clinical signs observed during this 26-week study. Additionally, there were no toxic differences in the following parameters: body weight, food consumption, hematology, clinical biochemistry, organ weight, and macroscopic findings. There were no adverse effects on histopathology observed in males or female rats treated with YLSP. Based on the results, the no-observed-adverse-effect-level of YLSP in rats is greater than 2.4 g/kg when administered orally for 26 consecutive weeks.


Assuntos
Extratos Vegetais/química , Raízes de Plantas/química , Polissacarídeos/administração & dosagem , Polissacarídeos/toxicidade , Administração Oral , Animais , Peso Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Masculino , Nível de Efeito Adverso não Observado , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Wistar
16.
Food Funct ; 6(5): 1712-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25920068

RESUMO

Yulangsan polysaccharide (YLSP) is a traditional Chinese medicine used in long-term treatment as a modulator of brain dysfunction and immunity. In this study, we evaluated the protective effect of YLSP against D-galactose-induced impairment of oxidative stress and the immune system and evaluated its possible mechanism of action. D-galactose was subcutaneously injected into the dorsal neck of mice daily for 8 weeks to establish the aging model. YLSP was simultaneously administered once daily. The results indicate that YLSP significantly improves the general appearance of the aging mice. YLSP significantly increased the levels of antioxidant enzymes, such as super oxide dismutase, glutathione peroxidase, catalase and total anti-oxidation capability, while decreasing the content of malondialdehyde in different tissues, including the liver, brain, and serum. YLSP also increased the interleukin-2 level while decreasing the interleukin-6 level. Moreover, YLSP significantly inhibited advanced glycation end product formation. Furthermore, YLSP decreased p21 and p53 gene expressions in the liver and brain of D-galactose-treated mice. These results suggest that YLSP may have a protective effect suppressing the aging process by enhancing antioxidant activity and immunity, as well as modulating aging-related gene expression.


Assuntos
Envelhecimento/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Galactose/efeitos adversos , Sistema Imunitário/efeitos dos fármacos , Millettia/química , Polissacarídeos/farmacologia , Envelhecimento/imunologia , Envelhecimento/metabolismo , Animais , Catalase/metabolismo , Glutationa Peroxidase/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Homeostase/efeitos dos fármacos , Humanos , Sistema Imunitário/metabolismo , Interleucina-2/metabolismo , Interleucina-6/metabolismo , Masculino , Malondialdeído/metabolismo , Camundongos , Oxirredução/efeitos dos fármacos , Superóxido Dismutase/metabolismo
17.
Neurosci Lett ; 570: 63-8, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24747687

RESUMO

Yulangsan polysaccharide (YLSP) has been utilized as a phytomedicine to managing nervous dysfunction in China. Thus, this study aimed to evaluate the potential YLSP-mediated detoxification role against morphine dependence in rats. The results indicated that the morphine dependence model significantly increased withdrawal symptoms, levels of NO and NOS (P<0.05). Furthermore, monoaminergic neurotransmitters, including DA and NE, were detected at elevated levels in the ventral tegmental area (VTA), hippocampus (HIP) and prefrontal cortex (PFC), respectively, while the level of DA was decreased and NE was increased in the nucleus accumbens (NAc). Conversely, YLSP administration significantly reversed naloxone-induced withdrawal symptoms, expression of brain NO and NOS, and monoaminergic neurotransmitters (P<0.05). Interestingly, YLSP shows an even more effective trend in attenuating withdrawal symptoms than does clonidine, although without a significant difference. These findings indicate that YLSP attenuation of the naloxone-induced withdrawal symptoms of morphine dependence may be mediated by regulation of the NO pathway and modulation of monoaminergic neurotransmitters.


Assuntos
Dependência de Morfina/tratamento farmacológico , Morfina/efeitos adversos , Óxido Nítrico/metabolismo , Polissacarídeos/uso terapêutico , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Dopamina/metabolismo , Medicamentos de Ervas Chinesas/uso terapêutico , Masculino , Dependência de Morfina/metabolismo , Dependência de Morfina/fisiopatologia , Naloxona , Óxido Nítrico Sintase/metabolismo , Norepinefrina/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais , Síndrome de Abstinência a Substâncias/metabolismo , Síndrome de Abstinência a Substâncias/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...