Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Transl Cancer Res ; 12(4): 743-764, 2023 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-37180648

RESUMO

Background: Ferula sinkiangensis (F. sinkiangensis) is a traditional Chinese medicine that has been used for thousands of years to treat stomach ailments. To identify the main active compounds and explore the mechanisms underlying the therapeutic effect of F. sinkiangensis against gastric cancer (GC) by network pharmacology, molecular docking analysis and cell experiment. Methods: Based on a review of the literature and previous experiments conducted by our research group, the active compounds of F. sinkiangensis were obtained. Active compounds and their target genes were screened from SwissADME, Pubchem, and Pharmmapper databases. GC-related target genes were obtained from GeneCards. The drug-compound-target-disease (D-C-T-D) network and protein-protein interaction (PPI) network were constructed by Cytoscape 3.7.2 and STRING database, and the core target genes and core active compounds were identified. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were conducted using the R package clusterProfiler. The core genes with high expression in GC were screened, which correlated with a poor prognosis using the GEPIA, UALCAN, HPA, and KMplotter databases. KEGG signaling pathway analysis was further conducted to predict the mechanism of F. sinkiangensis during the process of GC inhibition. The AutoDock vina 1.1.2 program was used to verify the molecular docking of the core active compounds and core target genes. MTT, Transwell, and Wound healing assay were used to detect the effects of ethyl acetate extract of F. sinkiangensis on the proliferation, invasion, and apoptosis of GC cells. Results: Final results indicated that the active compounds include Farnesiferol C, Assafoetidin, Lehmannolone, Badrakemone, etc. The identified core target genes were GPI, TKT, GLYCTK, ERBB2, GAPDH, etc. The Glycolysis/Gluconeogenesis pathway and the Pentose Phosphate pathway might play important roles in the treatment of GC with F. sinkiangensis. The data from the study showed that F. sinkiangensis was able to inhibit the proliferation of GC cells. Meanwhile, F. sinkiangensis remarkedly repressed the invasion and migration of GC cells in in vitro experiment. Conclusions: This study revealed that F. sinkiangensis has an antitumor effect in in vitro experiment, and that the mechanism of F. sinkiangensis in GC treatment shows characteristics of multi-components, multi-targets, and multi-pathways, which provides a theoretical basis for its clinical application and subsequent experimental verification.

2.
Artigo em Inglês | MEDLINE | ID: mdl-35392643

RESUMO

Ferula akitschkensis volatile oil (FAVO) has a good inhibitory activity on gastric cancer cell proliferation, but the mechanism of action is not yet clear. In this study, we tested the antigastric cancer efficacy and mechanism of FAVO using both in vivo and in vitro models. The results showed that FAVO effectively inhibited the proliferation, migration, and invasion of human gastric cancer SGC-7901 cells, the formation of small tubules of human umbilical vein endothelial cells as well as zebrafish intersegmental vessel and intestinal vein angiogenesis. In vivo experiments showed that FAVO significantly delayed the growth of SGC-7901 tumor-bearing nude mice and induced higher serum IL-2 and IFN-γ and reduced serum IL-6. Western blot results showed that FAVO reduced the expression of HIF-2α, VEGF, VEGFR2, P-VEGFR2, Akt, and P-Akt in SGC-7901 cells with CoCl2 induced hypoxia. We further clarified the main chemical components of FAVO through GC-MS analysis. In summary, FAVO may inhibit tumor growth and angiogenesis via inhibiting the HIF-2α/VEGF signaling pathway.

3.
Artigo em Inglês | MEDLINE | ID: mdl-34765011

RESUMO

This study identifies the active ingredients of Ferula sinkiangensis and investigates the role and mechanism of episamarcandin in colon cancer cells. The silica gel column chromatography was utilized to separate the chemical components of Ferula sinkiangensis. Sephadex LH-20 and semipreparative HPLC were adopted for further separation and purification. The compound episamarcandin showed good anticolon cancer activity among the 13 monomeric compounds obtained. Its effects on the apoptosis, cell cycle, and invasion and migration of colon cancer HCT 116 cells and PI3K-Akt signaling pathway were further investigated. The results showed that, similar to positive control cisplatin, episamarcandin inhibited the proliferation, promoted the apoptosis, arrested cells at G0/G1 phase, and suppressed migration and invasion of HCT 116 cells. A large number of apoptotic HCT 116 cells were observed under a transmission electron microscope. Fluorescence real-time quantitative PCR and western blot analysis showed that episamarcandin increased the expression of PTEN, p53, and Bax and decreased the expression of P-Akt, Akt, mTOR, Bcl-xl, and Bcl-2. Conclusively, episamarcandin may inhibit cell proliferation, migration, and invasion and promote the apoptosis of human colon cancer HCT 116 cells possibly through the PI3K-Akt signaling pathway.

4.
Br J Nutr ; 123(8): 859-869, 2020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-31524111

RESUMO

Flaxseed oil is rich in α-linolenic acid (ALA), which is the metabolic precursor of EPA and DHA. The present study investigated the effect of flaxseed oil supplementation on lipopolysaccharide (LPS)-induced muscle atrophy and carbohydrate oxidation impairment in a piglet model. Twenty-four weaned pigs were used in a 2 × 2 factorial experiment including dietary treatment (5 % maize oil v. 5 % flaxseed oil) and LPS challenge (saline v. LPS). On day 21 of treatment, the pigs were injected intraperitoneally with 100 µg/kg body weight LPS or sterile saline. At 4 h after injection, blood, gastrocnemius muscle and longissimus dorsi muscle were collected. Flaxseed oil supplementation increased ALA, EPA, total n-3 PUFA contents, protein:DNA ratio and pyruvate dehydrogenase complex quantity in muscles (P < 0·05). In addition, flaxseed oil reduced mRNA expression of toll-like receptor (TLR) 4 and nucleotide-binding oligomerisation domain protein (NOD) 2 and their downstream signalling molecules in muscles and decreased plasma concentrations of TNF-α, IL-6 and IL-8, and mRNA expression of TNF-α, IL-1ß and IL-6 (P < 0·05). Moreover, flaxseed oil inclusion increased the ratios of phosphorylated protein kinase B (Akt) 1:total Akt1 and phosphorylated Forkhead box O (FOXO) 1:total FOXO1 and reduced mRNA expression of FOXO1, muscle RING finger (MuRF) 1 and pyruvate dehydrogenase kinase 4 in muscles (P < 0·05). These results suggest that flaxseed oil might have a positive effect on alleviating muscle protein loss and carbohydrates oxidation impairment induced by LPS challenge through regulation of the TLR4/NOD and Akt/FOXO signalling pathways.


Assuntos
Óleo de Semente do Linho/farmacologia , Lipopolissacarídeos/toxicidade , Proteínas Musculares/metabolismo , Músculo Esquelético/efeitos dos fármacos , Suínos , Animais , Metabolismo dos Carboidratos , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Musculares/genética , Oxirredução , Fosforilação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
5.
Food Funct ; 10(1): 479-489, 2019 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-30638239

RESUMO

The intestinal microbiota plays a vital role in metabolism, pathogen resistance, and immune development in host cells, and is modifiable by dietary change. Lentinan (LNT), a type of mushroom polysaccharide, is known to ameliorate intestinal inflammation with the potential of therapeutic effect on digestive diseases. We hypothesized that LNT could alleviate Escherichia coli lipopolysaccharide (LPS)-induced intestinal injury via regulating the composition and metabolites of intestinal microbiota in a piglet model. Twenty-four weaned piglets were used in a 2 × 2 factorial design, and the main factors included a dietary treatment (basal or LNT diet) and immunological challenge (LPS or saline). After feeding basal or LNT diet for 21 days, pigs were injected with LPS or saline. At 4 h post-injection, pigs were killed and jejunum, ileum and cecal digesta were collected. LNT improved intestinal morphology and barrier function. LNT also inhibited inflammatory signaling pathways (toll-like receptor 4 and nucleotide binding oligomerization domain protein) and pro-inflammatory cytokines (tumor necrosis factor-α, interleukin-1ß and interleukin-6) expression, as well as up-regulated the heat shock protein 70 expression in small intestine. In addition, LNT enhanced the concentrations of propionate, butyrate, isobutyrate and isovalerate in cecal digesta, resulting in a significant increase in histone acetylation without affecting the protein level of G protein-coupled receptor 41 (GPR41), a short chain fatty acid receptor. Bacterial 16S rRNA gene pyrosequencing showed that LNT had a great impact on gut microbiota composition at different taxonomic levels. Moreover, the correlation analysis revealed some potential relationships between cecal metabolites and certain intestinal microbiota. These results indicate that LNT promotes intestinal health, in part, through altering intestinal microbiota composition and increasing the short chain fatty acid synthesis, which subsequently lead to a reduction in inflammation and hyper-acetylation of histones.


Assuntos
Microbioma Gastrointestinal/efeitos dos fármacos , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/microbiologia , Lentinano/administração & dosagem , Lipopolissacarídeos/efeitos adversos , Animais , Bactérias/classificação , Bactérias/efeitos dos fármacos , Bactérias/genética , Bactérias/metabolismo , Ácidos Graxos Voláteis/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Intestino Delgado/imunologia , Intestino Delgado/patologia , Suínos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
6.
J Anim Sci ; 96(9): 3804-3814, 2018 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-30059979

RESUMO

This study was performed to determine if pyruvate, which acts as a critical intermediate in energy metabolism, can substitute the role of glutamate as a metabolic fuel and effectively reduce nitrogen excretion in pigs. First, the experiment in vitro was carried out to investigate the effects of culturing porcine small intestinal epithelial cell line with pyruvate on the oxidation. Then, barrows weighing 40 kg were used in the experiment investigating the changes of nitrogen balance in response to addition of pyruvate to low-protein diets. Last, barrows (40 kg), which were surgically fitted with permanent catheters in the mesenteric vein, portal vein, hepatic vein, and carotid artery, were used to investigate the effects of supplementing low-protein diets with calcium pyruvate on the net portal fluxes of amino acids (AAs) and the consumption of AAs in the liver. The results showed that culturing cells with sodium pyruvate significantly reduced the number of glutamate oxidation (P < 0.05). Addition of calcium pyruvate to low-protein diets significantly reduced urinary nitrogen excretion from 13.2 g/d (18.0% crude protein, CP) to 10.3 g/d (15.0% CP) or 7.80 g/d (13.5% CP) and total nitrogen excretion from 22.5 g/d (18.0% CP) to 17.8 g/d (15.0% CP) or 14.2 g/d (13.5% CP) (P < 0.05), without obviously negative effects on the nitrogen retention (P > 0.05). Addition of calcium pyruvate to low-protein diets significantly decreased essential AA consumption rate in the liver (P < 0.05). This diet modification reduced the net portal fluxes of NH3, glycine, and alanine, as well as urea production rate in the liver (P < 0.05). The results indicated that pyruvate is an effective substitute for glutamate as a supplement in low-protein diets, reducing porcine nitrogen excretion and nitrogen consumption.


Assuntos
Ração Animal/análise , Ácido Glutâmico/farmacologia , Nitrogênio/metabolismo , Ácido Pirúvico/farmacologia , Suínos/metabolismo , Alanina/metabolismo , Aminoácidos/metabolismo , Fenômenos Fisiológicos da Nutrição Animal , Animais , Linhagem Celular , Dieta/veterinária , Dieta com Restrição de Proteínas , Proteínas Alimentares/metabolismo , Suplementos Nutricionais , Metabolismo Energético/fisiologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Ácido Glutâmico/metabolismo , Mucosa Intestinal/citologia , Masculino , Oxirredução , Ureia/metabolismo
7.
Biotechnol Lett ; 40(2): 427-435, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29159512

RESUMO

OBJECTIVE: To fabricate in vitro cell-dense, three-dimensional (3D) tumor models by employing a cell sheet technology for testing anti-cancer drug efficacy. RESULTS: The stratified liver tumor models were fabricated by stacking contiguous HepG2 cell sheets. Triple-layer (3L), double-layer (2L), single-layer (1L) cell sheet-based liver tumor models (CSLTMs) demonstrated 106, 96, 85% cell viability, respectively, after 3 days treatment of 10 µM doxorubicin hydrochloride (DOX), while cell viability in two-dimensional (2D) conventional culture (control) was 27%. After 7 days of DOX treatment, the viabilities of 3L, 2L, 1L, control were 24, 14, 3 and 4%, respectively. Probable explanations were blocked diffusion of DOX by the intact and multilayered structure and also hypoxia in the bottom of multilayered cell sheets. CONCLUSION: CSLTMs showed a thickness-dependent cytotoxic efficacy of DOX and greater drug resistance than the control, thereby providing useful information toward the development of improved biomimetic tumor models.


Assuntos
Antineoplásicos/farmacologia , Técnicas de Cultura de Células/métodos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Neoplasias Hepáticas/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Hep G2 , Humanos , Modelos Biológicos
8.
J Mater Chem B ; 1(38): 5003-5013, 2013 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-32261090

RESUMO

Nanoparticles are being increasingly recognized for their potential utility in biological applications including nanomedicine. The aim of this study is to investigate a new strategy to combine the ZnO nanoparticles with graphene for targeting photodynamic therapy (PDT) under visible light irradiation. Folic acid (FA), a targeting agent toward tumor cells, was conjugated onto graphene oxide (GO) via imide linkage. Using a simple and effective chemical precipitation method, a GO-FA-ZnO nanohybrid was then prepared. The combination of ZnO with GO-FA induced a remarkable improvement in tumor targeting, which has been demonstrated by the cellular uptake assay. Due to the high electrical conductivity of graphene, the interaction between graphene and ZnO, and the inhibition of aggregation, the hybrid of GO-FA and ZnO significantly enhances the photodynamic activity. It was noted that the photodynamic activity of the non-cytotoxic GO-FA-ZnO is mediated by reactive oxygen species (ROS) generation under visible light irradiation. Following the ROS generation, GO-FA-ZnO caused a significant decrease in cell viability, mitochondrial membrane potential, superoxide dismutase activity, catalase and glutathione peroxidase, as well as an increase in malonodialdehyde production. Moreover, GO-FA-ZnO induced apoptotic death by elevating the caspase-3 activity. The study presents a novel tumor targeting photosensitizer and a promising strategy in PDT for cancer treatment.

9.
Amino Acids ; 39(3): 633-40, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20162307

RESUMO

Sulfur amino acids (SAA), particularly methionine and cysteine, are critical for the gut to maintain its functions including the digestion, absorption and metabolism of nutrients, the immune surveillance of the intestinal epithelial layer and regulation of the mucosal response to foreign antigens. However, the metabolism of SAA in the gut, specifically the transmethylation of methionine, will result in a net release of homocysteine, which is shown to be associated with cardiovascular disease and stroke. Furthermore, the extensive catabolism of dietary methionine by the intestine or by luminal microbes may result in a decrease in nutritional efficiency. Therefore, the regulation of SAA metabolism in the gut is not only nutritionally relevant, but also relevant to the overall health and well-being. The superiority of DL-2-hydroxy-4-methylthiobutyrate to DL-methionine in decreasing homocysteine production, alleviating stress responses, and reducing the first-pass intestinal metabolism of dietary methionine may provide a promising implication for nutritional strategies to manipulate SAA metabolism and thus to improve the nutrition and health status of animals and perhaps humans.


Assuntos
Aminoácidos Sulfúricos/metabolismo , Mucosa Intestinal/metabolismo , Animais , Transporte Biológico , Humanos , Avaliação Nutricional
10.
Innate Immun ; 15(3): 169-78, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19474210

RESUMO

Our previous study demonstrated mRNA and protein expression of peroxisome proliferator-activated receptor-g (PPAR-g) in the immune system of weaned pigs. In this report, to test the hypothesis that activation of PPAR-g in immune system modulates inflammatory response, and adrenal and somatotropic responses associated with immune challenge, we administered intraperitoneally PPAR-g agonist and/or antagonist in weaned pigs subjected to Escherichia coli lipopolysaccharide (LPS) challenge. Unexpectedly, we found that a single injection of the PPAR-g agonist rosiglitazone (given at 3 mg/kg body weight 30 min before LPS injection) failed to block pro-inflammatory cytokine production induced by LPS injection. Rather, plasma levels of tumor necrosis factor-a (TNF-a) and interleukin-6 (IL-6), mRNA abundance of TNF-a in thymus, spleen, mesenteric lymph node and peripheral white blood cells, mRNA abundance of IL-6 in thymus, protein levels of TNF-a in spleen and mesenteric lymph node, and protein levels of IL-6 in spleen and mesenteric lymph node, were elevated beyond the levels in control pigs injected with LPS. Furthermore, rosiglitazone potentiated the increase of plasma cortisol and prostaglandin E(2) concentrations, and the decrease of plasma insulin-like growth factor-1 concentration induced by LPS injection. Co-administration of the PPAR-g antagonist bisphenol A diglycidyl ether (given 30 mg/kg body weight) 30 min prior to treatment with rosiglitazone antagonized the effect of the PPAR-g agonist, indicating a PPAR-g-dependent effect. Our data indicate that ligand-induced activation of PPAR-g does not ameliorate but enhances pro-inflammatory cytokine production, and further potentiates the adrenal and somatotropic changes in weaned pigs subjected to E. coli LPS challenge, which suggests that PPAR-g activation may not be useful, but potentially harmful, in the treatment of immune challenge in livestock. Our results raise doubts about the prevalently accepted anti-inflammatory role for PPAR-g activation.


Assuntos
Citocinas/biossíntese , Inflamação/imunologia , PPAR gama/agonistas , PPAR gama/imunologia , Animais , Compostos Benzidrílicos , Dimetil Sulfóxido/administração & dosagem , Dimetil Sulfóxido/farmacologia , Dinoprostona/agonistas , Dinoprostona/sangue , Compostos de Epóxi/administração & dosagem , Compostos de Epóxi/farmacologia , Escherichia coli/imunologia , Sequestradores de Radicais Livres/administração & dosagem , Sequestradores de Radicais Livres/farmacologia , Hidrocortisona/agonistas , Hidrocortisona/sangue , Inflamação/microbiologia , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/imunologia , Fator de Crescimento Insulin-Like I/metabolismo , Interleucina-6/agonistas , Interleucina-6/antagonistas & inibidores , Interleucina-6/imunologia , Interleucina-6/metabolismo , Lipopolissacarídeos/imunologia , Masculino , PPAR gama/antagonistas & inibidores , Rosiglitazona , Suínos/imunologia , Suínos/microbiologia , Tiazolidinedionas/administração & dosagem , Tiazolidinedionas/farmacologia , Fator de Necrose Tumoral alfa/agonistas , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Desmame
11.
Vet Immunol Immunopathol ; 124(1-2): 82-92, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18394717

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARgamma), a member of the nuclear hormone receptor superfamily, has been implicated in regulation of immunity and inflammation in rodents and humans. The objective of the current study was to investigate whether the expression of PPARgamma was altered in the immune system of weaned pigs after Escherichia coli lipopolysaccharide (LPS) injection. PPARgamma expression was investigated in the thymus, spleen, mesenteric lymph node and peripheral white blood cells of weaned pigs (8.54+/-0.24 kg BW) after LPS injection (100 microg/kg BW, n=6) and controls (sterile saline, n=6), by using real-time polymerase chain reaction, Western blot analysis, and immunohistochemistry. Plasma pro-inflammatory cytokines and hormones were also assessed. LPS triggered PPARgamma mRNA and protein expression in the thymus (P<0.05, 4.24-fold; P<0.10, 1.46-fold), spleen (P<0.10, 2.75-fold; P<0.05, 1.84-fold), mesenteric lymph node (P<0.05, 4.32-fold; P<0.05, 1.96-fold) and peripheral white blood cells (P<0.001, 24.44-fold; P<0.001, 1.58-fold). The LPS-injected pigs showed an increase in PPARgamma staining in splenic corpuscle and periarterial lymphatic sheath of white pulp (P<0.05) and red pulp (P<0.001) of spleen, and in medullas of thymus lobule of thymus (P<0.05), and in thymus-dependent area of mesenteric lymph node (P<0.05) compared to the control pigs. Concurrent with up-regulation of PPARgamma expression, LPS induced increases in plasma interleukin-6 (P<0.001), tumor necrosis factor-alpha (P<0.001), cortisol (P<0.001), prostaglandin E(2) (P<0.01) and 15-deoxy-Delta(12,14)-prostaglandin J(2) (15 d-PGJ(2)) (P<0.05), and decreases in plasma insulin (P<0.10) and insulin-like growth factor-1 (P<0.001). These results suggest that induction of PPARgamma expression in immune system may be associated with the release of the natural PPARgamma activating ligand 15 d-PGJ(2), and play an important role in host response to immunological stress. Additionally, it is possible that PPARgamma would be a new therapeutic target in treatment of immunological stress of livestock.


Assuntos
Lipopolissacarídeos/farmacologia , PPAR gama/imunologia , Suínos/imunologia , Animais , Western Blotting/veterinária , Dinoprostona/sangue , Hormônio do Crescimento/sangue , Hidrocortisona/sangue , Imuno-Histoquímica/veterinária , Insulina/sangue , Fator de Crescimento Insulin-Like I/análise , Interleucina-6/sangue , Lipopolissacarídeos/imunologia , Tecido Linfoide/imunologia , PPAR gama/biossíntese , PPAR gama/sangue , PPAR gama/genética , Prostaglandina D2/análogos & derivados , Prostaglandina D2/sangue , RNA/química , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Suínos/sangue , Fator de Necrose Tumoral alfa/sangue
12.
Br J Nutr ; 100(3): 552-60, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18275628

RESUMO

This study evaluated whether arginine (Arg) supplementation could attenuate gut injury induced by Escherichia coli lipopolysaccharide (LPS) challenge through an anti-inflammatory role in weaned pigs. Pigs were allotted to four treatments including: (1) non-challenged control; (2) LPS-challenged control; (3) LPS+0.5 % Arg; (4) LPS+1.0 % Arg. On day 16, pigs were injected with LPS or sterile saline. At 6 h post-injection, pigs were killed for evaluation of small intestinal morphology and intestinal gene expression. Within 48 h of challenge, 0.5 % Arg alleviated the weight loss induced by LPS challenge (P = 0.025). In all three intestinal segments, 0.5 or 1.0 % Arg mitigated intestinal morphology impairment (e.g. lower villus height and higher crypt depth) induced by LPS challenge (P < 0.05), and alleviated the decrease of crypt cell proliferation and the increase of villus cell apoptosis after LPS challenge (P < 0.01). The 0.5 % Arg prevented the elevation of jejunal IL-6 mRNA abundance (P = 0.082), and jejunal (P = 0.030) and ileal (P = 0.039) TNF-alpha mRNA abundance induced by LPS challenge. The 1.0 % Arg alleviated the elevation of jejunal IL-6 mRNA abundance (P = 0.053) and jejunal TNF-alpha mRNA abundance (P = 0.003) induced by LPS challenge. The 0.5 % Arg increased PPARgamma mRNA abundance in all three intestinal segments (P < 0.10), and 1.0 % Arg increased duodenal PPARgamma mRNA abundance (P = 0.094). These results indicate that Arg supplementation has beneficial effects in alleviating gut mucosal injury induced by LPS challenge. Additionally, it is possible that the protective effects of Arg on the intestine are associated with decreasing the expression of intestinal pro-inflammatory cytokines through activating PPARgamma expression.


Assuntos
Anti-Inflamatórios/administração & dosagem , Arginina/administração & dosagem , Mucosa Intestinal/imunologia , Intestino Delgado , Suínos/crescimento & desenvolvimento , Animais , Biomarcadores/análise , Suplementos Nutricionais , Duodeno , Escherichia coli , Feminino , Íleo , Interleucina-6/análise , Interleucina-6/genética , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Jejuno , Lipopolissacarídeos , Masculino , PPAR gama/genética , RNA Mensageiro/análise , Suínos/imunologia , Suínos/metabolismo , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/genética , Desmame
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA