Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Int J Biol Macromol ; 273(Pt 1): 133067, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38866287

RESUMO

Adjuvants, as the essential component of vaccines, are crucial in enhancing the magnitude, breadth and durability of immune responses. Unfortunately, commonly used Alum adjuvants predominantly provoke humoral immune response, but fail to evoke cellular immune response, which is crucial for the prevention of various chronic infectious diseases and cancers. Thus, it is necessary to develop effective adjuvants to simultaneously induce humoral and cellular immune response. In this work, we obtained a water soluble polysaccharide isolated and purified from Poria cocos, named as PCP, and explored the possibility of PCP as a vaccine adjuvant. The PCP, with Mw of 20.112 kDa, primarily consisted of →6)-α-D-Galp-(1→, with a small amount of →3)-ß-D-Glcp-(1 â†’ and →4)-ß-D-Glcp-(1→. Our results demonstrated that the PCP promoted the activation of dendritic cells (DCs) and macrophages in vitro. As the adjuvant to ovalbumin, the PCP facilitated the activation of DCs in lymph nodes, and evoked strong antibody response with a combination of Th1 and Th2 immune responses. Moreover, compared to Alum adjuvant, the PCP markedly induced a potent cellular response, especially the cytotoxic T lymphocytes response. Therefore, we confirmed that the PCP has great potential to be an available adjuvant for simultaneously inducing humoral and cellular immune responses.


Assuntos
Adjuvantes Imunológicos , Células Dendríticas , Polissacarídeos , Solubilidade , Água , Animais , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/química , Polissacarídeos/farmacologia , Polissacarídeos/química , Polissacarídeos/isolamento & purificação , Camundongos , Água/química , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Feminino , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Wolfiporia/química , Ovalbumina/imunologia , Imunidade Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Poria/química
2.
Poult Sci ; 103(7): 103768, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38703758

RESUMO

Baicalein (BAI) is a natural flavonoid with antioxidant, antitumor and antibacterial properties. However, the bioavailability of BAI was limited due to low solubility. This study aims to improve the solubility of BAI through the amorphous solid dispersion (ASD) and evaluate changes in its pharmacokinetics and pharmacodynamics in Taihang chickens. Polyethylene caprolactam-polyvinyl acetate-polyethylene glycol grafted copolymer (Soluplus) was chosen as the carrier, and ASD was prepared by rotary evaporation and was characterized by powder X-ray diffractions (PXRD), differential scanning calorimetry (DSC) and fourier transform infrared spectroscopy (FT-IR). In vitro dissolution assays were used to screen the optimal ratio of drug to carrier, in vivo pharmacokinetic assays were conducted to investigate the promoting effect on the absorption. In addition, the effects of ASD on the growth performance, meat quality, antioxidant capacity and intestinal flora were investigated. ASD (1:9 and 2:8) did not exhibit crystal diffraction peaks of BAI in PXRD or endothermic peaks in DSC, indicating the successful preparation of ASD. The results of in vitro dissolution assay showed that the cumulative dissolution rate of ASD (2:8) within 600 min was 52.67%, which was 7.84-fold higher than BAI. The pharmacokinetic results showed that the peak concentration (Cmax) and the area under the drug-time curve (AUC0∼24) of ASD (2:8) was (5.20 ± 0.82) µg/mL and (17.03 ± 0.67) µg·h/mL, which was 1.91 and 2.64-fold higher than BAI, respectively. Dietary supplementation of BAI and ASD could increase average daily gain (ADG), while decrease feed conversion ratio (FCR), but there was no significant difference (P > 0.05). The drip loss of BAIASD group was lower than BAI group (P < 0.05). In addition, the antioxidant capacity of Taihang chickens were enhanced, the diversity and the abundance of beneficial bacteria was improved. Results of BAI upon the dietary supplementation tested in Taihang chickens, after preparation of ASD, indicating a superior enhancement effect in growth performance, meat quality, antioxidant capacity and intestinal flora due to an improved solubility and optimized bioavailability.


Assuntos
Ração Animal , Antioxidantes , Disponibilidade Biológica , Galinhas , Dieta , Flavanonas , Microbioma Gastrointestinal , Carne , Solubilidade , Animais , Galinhas/crescimento & desenvolvimento , Antioxidantes/metabolismo , Flavanonas/administração & dosagem , Flavanonas/química , Flavanonas/farmacologia , Carne/análise , Ração Animal/análise , Microbioma Gastrointestinal/efeitos dos fármacos , Dieta/veterinária , Polivinil/química , Polivinil/administração & dosagem , Masculino , Polietilenoglicóis/química , Polietilenoglicóis/administração & dosagem , Suplementos Nutricionais/análise
3.
Pharmaceutics ; 16(3)2024 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-38543200

RESUMO

Genistein (GEN) is an active pharmaceutical ingredient that presents the challenges of poor water solubility and low oral bioavailability. To tackle these challenges, a GEN solid dispersion was prepared by solvent rotary evaporation using polyvinylpyrrolidone K30 (PVP K30) as a carrier. The optimal formulation was determined by drug loading efficiency and in vitro release. The physical state of the solid dispersion was characterized by DSC, XRD, SEM and FT-IR. And the results of the in vitro release study indicate that the drug release of SD (1:7) increased 482-fold that of pure GEN at 60 min. Following oral administration to rats, the Cmax and AUC0-24 of SD (1:7) was increased 6.86- and 2.06-fold to that of pure GEN. The adipose fat index and body weight of the SD (1:7) group were significantly lower than those of the GEN group (p < 0.05). Meanwhile, the levels of TC and TG in the serum were significantly decreased in the SD (1:7) group compared with the GEN group (p < 0.05). All experiments revealed that solid dispersion could be a promising formulation approach to improve the dissolution rate, oral bioavailability, and effect on the reduction of lipid accumulation in high-fat diet-induced obesity mice.

4.
ACS Omega ; 6(42): 28410-28420, 2021 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-34723038

RESUMO

Oleanolic acid (OA) is a triterpenoid commonly found in plants and has shown extensive pharmaceutical activities. This study aimed to investigate the underlying mechanism of antiosteoporosis (OP) action of OA by utilizing the network pharmacology approach and molecular docking methods. First, the targets of OA were identified using the GeneCards, Stitch, and Swisstarget databases, and the targets related to OP were mined using the NCBI, Genecards, and DisGeNet databases. The overlapped targets of OA and OP were regarded as candidate targets, and the String database was used to obtain the protein-protein interactions among the targets. Then, Gene Ontology (GO) functional enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) metabolic pathway enrichment pathways of the candidate targets were performed using the DAVID database. In addition, the top 16 targets in the protein interaction network were used for molecular docking. Finally, an animal model constructed using d-galactose-induced oxidative stress and a low-calcium diet with accelerated bone loss was used to verify the in vivo effects of OA on osteoporotic mice. A total of 42 candidate targets for OA to treat OP were obtained. According to the protein-protein interaction network, MAPK1 showed the highest connectivity with other proteins. Additionally, GO analysis identified the top 20 biological processes, 9 cellular components, and top 20 molecular functions. Moreover, the candidate targets were mainly involved in 13 signaling pathways such as TNF signaling pathway, insulin resistance, MAPK signaling pathway, apoptosis, and PI3K-Akt signaling pathways. Furthermore, molecular docking revealed that OA has a high degree of connections with 16 key proteins. In addition, the anti-OP effects of OA are further validated through the in vivo model. Altogether, our study elucidated the candidate targets for OA to alleviate OP, explored the protein-protein interactions and related signaling pathways of the targets, and validated the anti-OP effects of OA. It could provide a better understanding of the action mechanism in OA to treat OP.

5.
J Agric Food Chem ; 68(1): 168-175, 2020 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-31850758

RESUMO

Naringin is a polymethoxylated flavonoid commonly found in citrus species and has therapeutic potential in intestinal disorders. However, the effect and mechanism of naringin on gut-vascular barrier disruption has not yet been reported. This study aimed to investigate the distinguishing and selectively protective effects of naringin on tumor necrosis factor (TNF)-α-induced gut-vascular barrier disruption and elucidate the potential mechanism. In the present study, an in vitro gut-vascular barrier model composed of rat intestinal microvascular endothelial cells (RIMVECs) was studied. Evans blue-albumin efflux assay showed that naringin (50 µM) evidently protected the integrity of RIMVEC monolayer barriers against TNF-α-induced disruption. Naringin maintained the expression and distribution of tight junction proteins including zona occludin-1, occludin, claudin-1, and claudin-2. Additionally, naringin protected RIMVECs from TNF-α-induced apoptosis and cell migration suppression (41.1 ± 2.2 vs 51.1 ± 3.5%; 61.0 ± 5.1 vs 72.2 ± 6.2%). Our results indicate that naringin effectively ameliorates gut-vascular barrier disruption.


Assuntos
Células Endoteliais/efeitos dos fármacos , Flavanonas/farmacologia , Mucosa Intestinal/irrigação sanguínea , Mucosa Intestinal/efeitos dos fármacos , Extratos Vegetais/farmacologia , Substâncias Protetoras/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose/efeitos dos fármacos , Citrus/química , Claudina-1/genética , Claudina-1/metabolismo , Células Endoteliais/metabolismo , Técnicas In Vitro , Mucosa Intestinal/metabolismo , Microvasos/citologia , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Ocludina/genética , Ocludina/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/genética
6.
Mol Med Rep ; 19(3): 1958-1964, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30569099

RESUMO

Inflammatory bowel disease (IBD) is a chronic, idiopathic inflammatory disease of the small and/or large intestine. Endothelial expression of inflammatory mediators, including cytokines and adhesion molecules, serves a critical role in the initiation and progression of IBD. The dietary flavonoid, kaempferol, has been reported to inhibit expression of inflammatory mediators; however, the underlying mechanisms require further investigation. In the present study, a novel molecular mechanism of kaempferol against IBD was identified. The potential anti­inflammatory effect of kaempferol in a cellular model of intestinal inflammation was assessed using lipopolysaccharide (LPS)­induced rat intestinal microvascular endothelial cells (RIMVECs), and an underlying key molecular mechanism was identified. RIMVECs were pretreated with kaempferol of various concentrations (12.5, 25 and 50 µM) followed by LPS (10 µg/ml) stimulation. ELISA was used to examine the protein levels of tumor necrosis factor­α (TNF­α), interleukin­1ß (IL­1ß), IL­6, intercellular adhesion molecule-1 (ICAM­1) and vascular cell adhesion molecule-1 (VCAM­1) in the supernatant. Protein expression levels of Toll­like receptor 4 (TLR4), nuclear factor­κB (NF­κB) p65, inhibitor of NF­κB, mitogen­activated protein kinase p38 and signal transducer and activator of transcription (STAT) in cells were measured by western blotting. Kaempferol significantly reduced the overproduction of TNF­α, IL­1ß, interleukin­6, ICAM­1 and VCAM­1 induced by LPS, indicating the negative regulation of kaempferol in TLR4, NF­κB and STAT signaling underlying intestinal inflammation. The present results provide support for the potential use of kaempferol as an effective therapeutic agent for IBD treatment.


Assuntos
Mediadores da Inflamação/administração & dosagem , Inflamação/tratamento farmacológico , Doenças Inflamatórias Intestinais/tratamento farmacológico , Quempferóis/administração & dosagem , Animais , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/patologia , Doenças Inflamatórias Intestinais/induzido quimicamente , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Molécula 1 de Adesão Intercelular/genética , Interleucina-1beta/genética , Interleucina-6/genética , Intestinos/efeitos dos fármacos , Intestinos/patologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Molécula 1 de Adesão de Célula Vascular/genética
7.
Life Sci ; 216: 168-175, 2019 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-30471284

RESUMO

AIMS: In this study, we explored the underlying mechanisms of protective effects of rhein against intestinal barrier injury in a rat model, induced by intraperitoneal injection of lipopolysaccharide (LPS). MAIN METHODS: Twenty-four male rats were assigned equally to three groups. Rats were given an oral administration of rhein (66.7 mg/kg/day) or not for three continuous days. LPS or saline were injected intraperitoneally in an hour after the last oral administration. The rats were sacrificed at 7 h after LPS or saline administration. Both blood samples and intestinal samples were collected. KEY FINDINGS: Rhein pretreatment markedly inhibited the levels of serum diamine oxidase (DAO), D-lactate (D-lac) and intestinal histological damage, significantly recovered the levels of intestinal DAO, ZO-1 and occludin. Additionally, rhein suppressed LPS-induced intestinal inflammation and oxidative stress, by decreased serum and intestinal, tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6 and nitric oxide levels, up-regulated intestinal catalase, glutathione peroxidase (GSH-Px) activities and HO-1 expression, and down-regulated malondialdehyde (MDA) level in the small intestine. Finally, rhein inhibited JNK, p38 MAPK phosphorylation and activated Nrf2 pathway. SIGNIFICANCE: Rhein could exert the anti-inflammatory and anti-oxidative effects against LPS-induced intestinal barrier injury by suppressing p38 MAPK and JNK and activating Nrf2 pathway.


Assuntos
Antraquinonas/farmacologia , Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Inflamação/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Inflamação/patologia , Interleucina-1beta/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/patologia , Lipopolissacarídeos/toxicidade , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Malondialdeído/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Óxido Nítrico/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Molecules ; 23(9)2018 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-30201908

RESUMO

Tetrahydroxystilbene glucoside (TSG) is a unique component of the bone-reinforcing herb Radix Polygoni Multiflori Preparata (RPMP). It has the ability to promote bone formation and protect osteoblasts. However, the underlying mechanism remains unclear. To better understand its biological function, we determined TSG's effect on murine pre-osteoblastic MC3T3-E1 cells by the MTT assay, flow cytometry, FQ-PCR, Western blot, and ELISA. The results showed that TSG caused an elevation of the MC3T3-E1 cell number, the number of cells in the S phase, and the mRNA levels of the runt-related transcription factor-2 (Runx2), osterix (Osx), and collagen type I α1 (Col1a1). In addition, the osteoprotegerin (OPG) mRNA level was up-regulated, while the nuclear factor-κB ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) mRNA levels were down-regulated by TSG. Furthermore, TSG activated the phosphoinosmde-3-kinase/protein kinase B (also known as PI3K/Akt) pathway, and blocking this pathway by the inhibitor LY-294002 could impair TSG's functions in relation to the MC3T3-E1 cells. In conclusion, TSG could activate the PI3K/Akt pathway and thus promote MC3T3-E1 cell proliferation and differentiation, and influence OPG/RANKL/M-CSF expression. TSG merits further investigation as a potential therapeutic agent for osteoporosis treatment.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Glucosídeos/farmacologia , Fator Estimulador de Colônias de Macrófagos/genética , Osteoprotegerina/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ligante RANK/genética , Estilbenos/farmacologia , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cromonas/farmacologia , DNA/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Glucosídeos/química , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos , Morfolinas/farmacologia , Osteoprotegerina/metabolismo , Ligante RANK/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estilbenos/química , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
9.
J Ethnopharmacol ; 170: 88-95, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-25978950

RESUMO

UNLABELLED: Fructus Ligustri Lucidi, fruits of Ligustrum lucidum Ait. (Oleaceae), has the effects of tonifying the liver and the kidney and strengthening the bones and muscles. In ancient times, Fructus Ligustri Lucidi can be prepared in ethanol or in water. Some active compounds have been found in Fructus Ligustri Lucidi, like Oleanolic acid and Ursolic acid, and Ursolic acid were proved to have osteogenic effects. METHODS AND RESULTS: To prove that Fructus Ligustri Lucidi water extract have osteogenic effects on MC3T3-E1 cells and how these effects work, we used CCK-8 (cell counting kit-8), ELISA (enzyme-linked immunosorbent assay), FQ-PCR (realtime fluorescence quantitative PCR) and western blot assays. After treatment with Fructus Ligustri Lucidi for 48h, 72h, 96h, the cell viability was marked increased, on concentration-dependently and time-dependently pattern. High and low concentrations of Fructus Ligustri Lucidi promoted differentiation of cells. Fructus Ligustri Lucidi could up-regulate OPG and RANKL protein in supernatant at 48h and 72h except for highest concentration (10(-1)mg/ml). Fructus Ligustri Lucidi promote OPG and RANKL mRNA expression at 48h and 72h, while the level of promoting at 72 was higher than 48h. 10(-5)mg/ml of Fructus Ligustri Lucidi up-regulates OPG protein expression and down-regulates RANKL protein expression. After treatment with Fructus Ligustri Lucidi water extract, inhibitors, Fructus Ligustri Lucidi water extract with inhibitors for 72h, inhibitors PD 98059, SB 203580, SP600125 and LY 294002 showed Fructus Ligustri Lucidi-induced cell proliferation and the leakage of OPG proteins effects. Fructus Ligustri Lucidi promoted the protein levels of ERK, p-ERK, p-JNK, p38, pp38, AKT and p-AKT, and inhibited the protein levels of JNK. CONCLUSIONS: Fructus Ligustri Lucidi water extract promoted cell proliferation and differentiation, mRNA and protein expression of OPG and RANKL on MC3T3-E1 cells. The effects of cell proliferation and leakage of OPG related to MAPK and AKT signaling pathways in different ways.


Assuntos
Ligustrum/química , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Extratos Vegetais/farmacologia , Células 3T3 , Animais , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Frutas , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Osteoblastos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...