Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 34(2): 2483-2496, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31909535

RESUMEN

Deoxynivalenol (DON) is one of the most common mycotoxins that contaminates food or feed and cause intestinal damage. Long-chain n-3 polyunsaturated fatty acids (PUFA) such as EPA and DHA exert beneficial effects on intestinal integrity in animal models and clinical trials. Necroptosis signaling pathway plays a critical role in intestinal cell injury. This study tested the hypothesis that EPA and DHA could alleviate DON-induced injury to intestinal porcine epithelial cells through modulation of the necroptosis signaling pathway. Intestinal porcine epithelial cell 1 (IPEC-1) cells were cultured with or without EPA or DHA (6.25-25 µg/mL) in the presence or absence of 0.5 µg/mL DON for indicated time points. Cell viability, cell number, lactate dehydrogenase (LDH) activity, cell necrosis, transepithelial electrical resistance (TEER), fluorescein isothiocyanate-labeled dextran 4kDa (FD4) flux, tight junction protein distribution, and protein abundance of necroptosis related signals were determined. EPA and DHA promoted cell growth indicated by higher cell viability and cell number, and inhibited cell injury indicated by lower LDH activity in the media. EPA and DHA also improved intestinal barrier function, indicated by higher TEER and lower permeability of FD4 flux as well as increased proportions of tight junction proteins located in the plasma membrane. Moreover, EPA and DHA decreased cell necrosis demonstrated by live cell imaging and transmission electron microscopy. Finally, EPA and DHA downregulated protein expressions of necroptosis related signals including tumor necrosis factor receptor (TNFR1), receptor interacting protein kinase 1 (RIP1), RIP3, phosphorylated mixed lineage kinase-like protein (MLKL), phosphoglycerate mutase family 5 (PGAM5), dynamin-related protein 1 (Drp1), and high mobility group box-1 protein (HMGB1). EPA and DHA also inhibited protein expression of caspase-3 and caspase-8. These results suggest that EPA and DHA prevent DON-induced intestinal cell injury and enhance barrier function, which is associated with inhibition of the necroptosis signaling pathway.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/farmacología , Células Epiteliales , Mucosa Intestinal , Necroptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tricotecenos/toxicidad , Animales , Células Epiteliales/metabolismo , Células Epiteliales/patología , Mucosa Intestinal/lesiones , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Porcinos
2.
Molecules ; 26(5)2021 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-33652818

RESUMEN

Glaesserella parasuis (G. parasuis) causes inflammation and damage to piglets. Whether polyserositis caused by G. parasuis is due to tight junctions damage and the protective effect of baicalin on it have not been examined. Therefore, this study aims to investigate the effects of baicalin on peritoneal tight junctions of piglets challenged with G. parasuis and its underlying molecular mechanisms. Piglets were challenged with G. parasuis and treated with or without baicalin. RT-PCR was performed to examine the expression of peritoneal tight junctions genes. Immunofluorescence was carried out to detect the distribution patterns of tight junctions proteins. Western blot assays were carried out to determine the involved signaling pathways. Our data showed that G. parasuis infection can down-regulate the tight junctions expression and disrupt the distribution of tight junctions proteins. Baicalin can alleviate the down-regulation of tight junctions mRNA in peritoneum, prevent the abnormalities and maintain the continuous organization of tight junctions. Our results provide novel evidence to support that baicalin has the capacity to protect peritoneal tight junctions from G. parasuis-induced inflammation. The protective mechanisms of baicalin could be associated with inhibition of the activation of PKC and MLCK/MLC signaling pathway. Taken together, these data demonstrated that baicalin is a promising natural agent for the prevention and treatment of G. parasuis infection.


Asunto(s)
Flavonoides/farmacología , Infecciones por Pasteurellaceae/tratamiento farmacológico , Pasteurellaceae/efectos de los fármacos , Enfermedades de los Porcinos/tratamiento farmacológico , Animales , Pasteurellaceae/genética , Pasteurellaceae/patogenicidad , Infecciones por Pasteurellaceae/genética , Infecciones por Pasteurellaceae/microbiología , Infecciones por Pasteurellaceae/veterinaria , Peritoneo/efectos de los fármacos , Peritoneo/microbiología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , Porcinos , Enfermedades de los Porcinos/microbiología , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/genética , Uniones Estrechas/microbiología
3.
Mol Cell Biochem ; 472(1-2): 45-56, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32519231

RESUMEN

Glässer's disease, caused by Haemophilus parasuis (H. parasuis), is associated with vascular damage and vascular inflammation in pigs. Therefore, early assessment and treatment are essential to control the inflammatory disorder. MicroRNAs have been shown to be involved in the vascular pathology. Baicalin has important pharmacological functions, including anti-inflammatory, antimicrobial and antioxidant effects. In this study, we investigated the changes of microRNAs in porcine aortic vascular endothelial cells (PAVECs) induced by H. parasuis and the effect of baicalin in this model by utilizing high-throughput sequencing. The results showed that 155 novel microRNAs and 76 differentially expressed microRNAs were identified in all samples. Subsequently, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of the target genes of the differentially expressed microRNAs demonstrated that regulation of actin cytoskeleton, focal adhesion, ECM-receptor interaction, bacterial invasion of epithelial cells, and adherens junction were the most interesting pathways after PAVECs were infected with H. parasuis. In addition, when the PAVECs were pretreated with baicalin, mismatch repair, peroxisome, oxidative phosphorylation, DNA replication, and ABC transporters were the most predominant signaling pathways. STRING analysis showed that most of the target genes of the differentially expressed microRNAs were associated with each other. The expression levels of the differentially expressed microRNAs were negatively co-regulated with their target genes' mRNA following pretreatment with baicalin in the H. parasuis-induced PAVECs using co-expression networks analysis. This is the first report that microRNAs might have key roles in inflammatory damage of vascular tissue during H. parasuis infection. Baicalin regulated the microRNAs changes in the PAVECs following H. parasuis infection, which may represent useful novel targets to prevent or treat H. parasuis infection.


Asunto(s)
Aorta/metabolismo , Endotelio Vascular/metabolismo , Flavonoides/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Infecciones por Haemophilus/microbiología , MicroARNs/genética , Transcriptoma/efectos de los fármacos , Animales , Animales Recién Nacidos , Antiinflamatorios no Esteroideos/farmacología , Aorta/citología , Aorta/microbiología , Endotelio Vascular/citología , Endotelio Vascular/microbiología , Haemophilus parasuis/aislamiento & purificación , Porcinos
4.
Vet Res ; 51(1): 102, 2020 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-32795339

RESUMEN

Glaesserella parasuis (G. parasuis) causes porcine vascular inflammation and damage. Baicalin is reported to have antioxidant and anti-inflammatory functions. However, whether baicalin protects piglets against G. parasuis challenge and the potential protective mechanism have not been investigated. Therefore, in this study, we comprehensively examined the protective efficacy of baicalin in piglets challenged with G. parasuis and the possible protective mechanism. Our results show that baicalin attenuated the release of the inflammation-related cytokines interleukin (IL) 1ß, IL6, IL8, IL10, and tumour necrosis factor α (TNF-α) and reduced high mobility group box 1 (HMGB1) production and cell apoptosis in piglets infected with G. parasuis. Baicalin also inhibited the activation of the mitogen-activated protein kinase (MAPK) signalling pathway and protected piglets against G. parasuis challenge. Taken together, our data suggest that baicalin could protect piglets from G. parasuis by reducing HMGB1 release, attenuating cell apoptosis, and inhibiting MAPK signalling activation, thereby alleviating the inflammatory response induced by the bacteria. Our results suggest that baicalin has utility as a novel therapeutic drug to control G. parasuis infection.


Asunto(s)
Antiinfecciosos/uso terapéutico , Flavonoides/uso terapéutico , Infecciones por Haemophilus/veterinaria , Haemophilus parasuis/fisiología , Sustancias Protectoras/uso terapéutico , Enfermedades de los Porcinos/prevención & control , Animales , Relación Dosis-Respuesta a Droga , Infecciones por Haemophilus/microbiología , Infecciones por Haemophilus/prevención & control , Sus scrofa , Porcinos , Enfermedades de los Porcinos/microbiología
5.
BMC Vet Res ; 15(1): 98, 2019 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-30909903

RESUMEN

BACKGROUND: Haemophilus parasuis (HPS) is the causative agent of Glässer's disease, characterized by arthritis, fibrinous polyserositis and meningitis, and resulting in worldwide economic losses in the swine industry. Baicalin (BA), a commonly used traditional Chinese medication, has been shown to possess a series of activities, such as anti-bacterial, anti-viral, anti-tumor, anti-oxidant and anti-inflammatory activities. However, whether BA has anti-apoptotic effects following HPS infection is unclear. Here, we investigated the anti-apoptotic effects and mechanisms of BA in HPS-induced apoptosis via the protein kinase C (PKC)-mitogen-activated protein kinase (MAPK) pathway in piglet's mononuclear phagocytes (PMNP). RESULTS: Our data demonstrated that HPS could induce reactive oxygen species (ROS) production, arrest the cell cycle and promote apoptosis via the PKC-MAPK signaling pathway in PMNP. Moreover, when BA was administered, we observed a reduction in ROS production, suppression of cleavage of caspase-3 in inducing apoptosis, and inhibition of activation of the PKC-MAPK signaling pathway for down-regulating p-JNK, p-p38, p-ERK, p-PKC-α and PKC-δ in PMNP triggered by HPS. CONCLUSIONS: Our data strongly suggest that BA can reverse the apoptosis initiated by HPS through regulating the PKC-MAPK signaling pathway, which represents a promising therapeutic agent in the treatment of HPS infection.


Asunto(s)
Antibacterianos/uso terapéutico , Flavonoides/uso terapéutico , Haemophilus parasuis/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Monocitos/metabolismo , Enfermedades de los Porcinos/tratamiento farmacológico , Animales , Animales Recién Nacidos/metabolismo , Animales Recién Nacidos/microbiología , Antibacterianos/farmacología , Apoptosis/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Flavonoides/farmacología , Infecciones por Haemophilus/tratamiento farmacológico , Infecciones por Haemophilus/metabolismo , Infecciones por Haemophilus/veterinaria , Monocitos/efectos de los fármacos , Monocitos/microbiología , Especies Reactivas de Oxígeno/metabolismo , Porcinos , Enfermedades de los Porcinos/metabolismo , Enfermedades de los Porcinos/microbiología
6.
Int J Mol Sci ; 20(10)2019 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-31091773

RESUMEN

The gut microbiome has important effects on gastrointestinal diseases. Diarrhea attenuation functions of baicalin (BA) is not clear. Baicalin-aluminum complexes (BBA) were synthesized from BA, but the BBA's efficacy on the diarrhea of piglets and the gut microbiomes have not been explored and the mechanism remains unclear. This study has explored whether BBA could modulate the composition of the gut microbiomes of piglets during diarrhea. The results showed that the diarrhea rate reduced significantly after treatment with BBA. BBA altered the overall structure of the gut microbiomes. In addition, the Gene Ontology (GO) enrichment analysis indicated that the functional differentially expressed genes, which were involved in the top 30 GO enrichments, were associated with hydrogenase (acceptor) activity, nicotinamide-nucleotide adenylyltransferase activity, and isocitrate lyase activity, belong to the molecular function. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that flagellar assembly, bacterial chemotaxis, lipopolysaccharide biosynthesis, ATP-binding cassette transporters (ABC) transporters, biosynthesis of amino acids, and phosphotransferase system (PTS) were the most enriched during BBA treatment process. Taken together, our results first demonstrated that BBA treatment could modulate the gut microbiomes composition of piglets with diarrhea, which may provide new potential insights on the mechanisms of gut microbiomes associated underlying the antimicrobial efficacy of BBA.


Asunto(s)
Antiinfecciosos/farmacología , Heces/microbiología , Flavonoides/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Aluminio/química , Animales , Antiinfecciosos/química , Antiinfecciosos/uso terapéutico , Diarrea/tratamiento farmacológico , Diarrea/veterinaria , Flavonoides/química , Flavonoides/uso terapéutico , Porcinos , Enfermedades de los Porcinos/tratamiento farmacológico
7.
Int J Mol Sci ; 19(5)2018 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-29702580

RESUMEN

Haemophilus parasuis (H. parasuis) can cause Glässer’s disease in pigs. However, the molecular mechanism of the inflammation response induced by H. parasuis remains unclear. The high-mobility group box 1 (HMGB1) protein is related to the pathogenesis of various infectious pathogens, but little is known about whether H. parasuis can induce the release of HMGB1 in piglet peripheral blood monocytes. Baicalin displays important anti-inflammatory and anti-microbial activities. In the present study, we investigated whether H. parasuis can trigger the secretion of HMGB1 in piglet peripheral blood monocytes and the anti-inflammatory effect of baicalin on the production of HMGB1 in peripheral blood monocytes induced by H. parasuis during the inflammation response. In addition, host cell responses stimulated by H. parasuis were determined with RNA-Seq. The RNA-Seq results showed that H. parasuis infection provokes the expression of cytokines and the activation of numerous pathways. In addition, baicalin significantly reduced the release of HMGB1 in peripheral blood monocytes induced by H. parasuis. Taken together, our study showed that H. parasuis can induce the release of HMGB1 and baicalin can inhibit HMGB1 secretion in an H. parasuis-induced peripheral blood monocytes model, which may provide a new strategy for preventing the inflammatory disorders induced by H. parasuis.


Asunto(s)
Antiinfecciosos/farmacología , Antiinflamatorios/farmacología , Flavonoides/farmacología , Proteína HMGB1/metabolismo , Enfermedades de los Porcinos/tratamiento farmacológico , Animales , Antiinfecciosos/uso terapéutico , Antiinflamatorios/uso terapéutico , Secuencia de Bases , Citocinas/genética , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Flavonoides/uso terapéutico , Proteína HMGB1/genética , Infecciones por Haemophilus/tratamiento farmacológico , Infecciones por Haemophilus/veterinaria , Haemophilus parasuis/patogenicidad , Inflamación/tratamiento farmacológico , Inflamación/veterinaria , Monocitos/efectos de los fármacos , Cultivo Primario de Células , Porcinos
8.
Int J Mol Sci ; 19(5)2018 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-29710817

RESUMEN

Haemophilus parasuis is the causative agent of Glässer’s disease in pigs. H. parasuis can cause vascular damage, although the mechanism remains unclear. In this study, we investigated the host cell responses involved in the molecular pathway interactions in porcine aortic vascular endothelial cells (PAVECs) induced by H. parasuis using RNA-Seq. The transcriptome results showed that when PAVECs were infected with H. parasuis for 24 h, 281 differentially expressed genes (DEGs) were identified; of which, 236 were upregulated and 45 downregulated. The 281 DEGs were involved in 136 KEGG signaling pathways that were organismal systems, environmental information processing, metabolism, cellular processes, and genetic information processing. The main pathways were the Rap1, FoxO, and PI3K/Akt signaling pathways, and the overexpressed genes were determined and verified by quantitative reverse transcription polymerase chain reaction. In addition, 252 genes were clustered into biological processes, molecular processes, and cellular components. Our study provides new insights for understanding the interaction between bacterial and host cells, and analyzed, in detail, the possible mechanisms that lead to vascular damage induced by H. parasuis. This may lead to development of novel therapeutic targets to control H. parasuis infection.


Asunto(s)
Células Endoteliales/metabolismo , Infecciones por Haemophilus/genética , Transcriptoma , Animales , Células Cultivadas , Células Endoteliales/microbiología , Endotelio Vascular/citología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Infecciones por Haemophilus/metabolismo , Haemophilus parasuis/patogenicidad , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Porcinos
9.
Amino Acids ; 49(1): 203-207, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27761755

RESUMEN

ß-Conglycinin (ß-CG) is well known for inducing intestinal allergies and dysfunction in neonates and young pigs. However, the underlying mechanisms are largely unknown. In this study, to clarify the role of autophagy in ß-CG-induced cytotoxicity, we investigated the effects of ß-CG on cell viability and autophagy activity in porcine enterocytes (IPEC-1 cells). The results indicated that the cell viability was decreased with the increasing levels of ß-CG. ß-CG treatment enhanced the eGFP-LC3 puncta per cells and LC3-II/LC3-I, and the latter was further increased in IPEC-1 cells cultured with bafilomycin A1. We conclude that ß-CG enhances autophagy activity in enterocytes.


Asunto(s)
Alérgenos/farmacología , Antígenos de Plantas/farmacología , Autofagia/efectos de los fármacos , Globulinas/farmacología , Glycine max/química , Proteínas de Almacenamiento de Semillas/farmacología , Proteínas de Soja/farmacología , Alérgenos/aislamiento & purificación , Alimentación Animal/análisis , Animales , Antígenos de Plantas/aislamiento & purificación , Autofagia/genética , Línea Celular , Supervivencia Celular/efectos de los fármacos , Enterocitos , Regulación de la Expresión Génica , Globulinas/aislamiento & purificación , Macrólidos/farmacología , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Almacenamiento de Semillas/aislamiento & purificación , Proteínas de Soja/aislamiento & purificación , Porcinos
10.
Amino Acids ; 49(8): 1277-1291, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28616751

RESUMEN

Animal models are needed to study and understand a human complex disease. Because of their similarities in anatomy, structure, physiology, and pathophysiology, the pig has proven its usefulness in studying human gastrointestinal diseases, such as inflammatory bowel disease, ischemia/reperfusion injury, diarrhea, and cancer. To understand the pathogenesis of these diseases, a number of experimental models generated in pigs are available, for example, through surgical manipulation, chemical induction, microbial infection, and genetic engineering. Our interests have been using amino acids as therapeutics in pig and human disease models. Amino acids not only play an important role in protein biosynthesis, but also exert significant physiological effects in regulating immunity, anti-oxidation, redox regulation, energy metabolism, signal transduction, and animal behavior. Recent studies in pigs have shown that specific dietary amino acids can improve intestinal integrity and function under normal and pathological conditions that protect the host from different diseases. In this review, we summarize several pig models in intestinal diseases and how amino acids can be used as therapeutics in treating pig and human diseases.


Asunto(s)
Aminoácidos/metabolismo , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/prevención & control , Animales , Humanos
11.
Amino Acids ; 49(12): 2091-2098, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28929442

RESUMEN

Methionine is an aliphatic, sulfur-containing, essential amino acid, and a precursor of succinyl-CoA, homocysteine, cysteine, creatine, and carnitine. Recent research has demonstrated that methionine can regulate metabolic processes, the innate immune system, and digestive functioning in mammals. It also intervenes in lipid metabolism, activation of endogenous antioxidant enzymes such as methionine sulfoxide reductase A, and the biosynthesis of glutathione to counteract oxidative stress. In addition, methionine restriction prevents altered methionine/transmethylation metabolism, thereby decreasing DNA damage and carcinogenic processes and possibly preventing arterial, neuropsychiatric, and neurodegenerative diseases. This review focuses on the role of methionine in metabolism, oxidative stress, and related diseases.


Asunto(s)
Metionina/metabolismo , Estrés Oxidativo/fisiología , Animales , Enfermedades Cardiovasculares/fisiopatología , Humanos , Inmunidad Innata , Enfermedades Renales/fisiopatología , Metabolismo de los Lípidos , Hepatopatías/fisiopatología , Neoplasias/fisiopatología , Enfermedades del Sistema Nervioso/fisiopatología
12.
Amino Acids ; 49(12): 1915-1929, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28271166

RESUMEN

This study determined whether N-acetylcysteine (NAC) could improve intestinal function through phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR), epithelial growth factor receptor (EGFR), toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB), adenosine 5'-monophosphate-activated protein kinase (AMPK), and type I interferon (IFN) signaling pathways in a piglet model of lipopolysaccharides (LPS) challenge. Thirty-two piglets (24-day-old) were randomly allocated to one of four treatments, with eight replicates per treatment and one piglet per replicate. The experiment consisted of four treatments in a 2 × 2 factorial arrangement with two diets (supplemented with 0 or 500 mg NAC/kg diet) and saline or LPS administration. On day 20 of the trial, piglets in the LPS and LPS + NAC groups were intraperitoneally injected with 0 (saline) or 100 µg LPS/kg BW. Blood samples were obtained at 3 h and intestinal mucosae were collected at 6 h post LPS or saline injection. The growth performance was not affected by dietary NAC. LPS induced intestinal dysfunction, as indicated by: (1) reductions in the small-intestinal glutathione concentrations and plasma D-xylose levels; (2) elevations in plasma diamine oxidase activity, mucosal MMP3 mRNA levels and caspase-3 protein abundance; (3) reduced the activities of the small-intestinal mucosal maltase, sucrase and lactase. The adverse effects of LPS on porcine intestinal function and redox status were mitigated by NAC supplementation through the activation of multiple signaling pathways involving PI3K/Akt/mTOR, EGFR, TLR4/NF-κB, AMPK, and type I IFN. Our findings provide novel mechanisms for beneficial effects of NAC in protecting the intestine from inflammation in animals.


Asunto(s)
Acetilcisteína/farmacología , Mucosa Intestinal/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Lipopolisacáridos/toxicidad , Transducción de Señal/efectos de los fármacos , Sus scrofa , Animales , Suplementos Dietéticos , Receptores ErbB/metabolismo , Depuradores de Radicales Libres/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Mucosa Intestinal/fisiopatología , Intestino Delgado/fisiopatología , Oxidación-Reducción/efectos de los fármacos , Porcinos , Destete
13.
Amino Acids ; 49(12): 1931-1943, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28258325

RESUMEN

Porcine epidemic diarrhea virus (PEDV) infects the intestine of young pigs, but effective measures for prevention and treatment are lacking. N-Acetylcysteine (NAC) has been shown to reduce endotoxin-induced intestinal dysfunction. This study was conducted with the PEDV-infected neonatal piglet model to determine the effect of NAC supplementation on intestinal function. Thirty-two 7-day-old piglets were randomly allocated to one of four treatments in a 2 × 2 factorial design consisting of two liquid diets (0 or 50 mg/kg BW NAC supplementation) and oral administration of 0 or 104.5 TCID50 (50% tissue culture infectious dose) PEDV. On day 7 of the trial, half of the pigs (n = 8) in each dietary treatment received either sterile saline or PEDV (Yunnan province strain) solution at 104.5 TCID50 per pig. On day 10 of the trial, D-xylose (0.1 g/kg BW) was orally administrated to all pigs. One hour later, jugular vein blood samples were collected, and then all pigs were killed to obtain the small intestine. PEDV infection increased diarrhea incidence, while reducing ADG. PEDV infection also decreased plasma D-xylose concentration, small intestinal villus height, mucosal I-FABP and villin mRNA levels but increased mucosal MX1 and GCNT3 mRNA levels (P < 0.05). Dietary NAC supplementation ameliorated the PEDV-induced abnormal changes in all the measured variables. Moreover, NAC reduced oxidative stress, as indicated by decreases in plasma and mucosal H2O2 levels. Collectively, these novel results indicate that dietary supplementation with NAC alleviates intestinal mucosal damage and improves the absorptive function of the small intestine in PEDV-infected piglets.


Asunto(s)
Acetilcisteína/administración & dosificación , Acetilcisteína/farmacología , Infecciones por Coronavirus/veterinaria , Mucosa Intestinal/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Virus de la Diarrea Epidémica Porcina , Enfermedades de los Porcinos/tratamiento farmacológico , Animales , Animales Recién Nacidos , Infecciones por Coronavirus/tratamiento farmacológico , Suplementos Dietéticos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/fisiopatología , Intestino Delgado/patología , Intestino Delgado/fisiopatología , Oxidación-Reducción/efectos de los fármacos , Plasma/efectos de los fármacos , Plasma/enzimología , Sus scrofa , Porcinos , Aumento de Peso/efectos de los fármacos
14.
Amino Acids ; 49(12): 1945-1954, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28299479

RESUMEN

Dietary glutamine (Gln) or arginine (Arg) supplementation is beneficial for intestinal health; however, whether Gln or Arg may confer protection against Enterotoxigenic Escherichia coli (ETEC) infection is not known. To address this, we used an ETEC-infected murine model to investigate the protective effects of Gln and Arg. Experimentally, we pre-treated mice with designed diet of Gln or Arg supplementation prior to the oral ETEC infection and then assessed mouse mortality and intestinal bacterial burden. We also determined the markers of intestinal innate immunity in treated mice, including secretory IgA response (SIgA), mucins from goblet cells, as well as antimicrobial peptides from Paneth cells. ETEC colonized in mouse small intestine, including duodenum, jejunum, and ileum, and inhibited the mRNA expression of intestinal immune factors, such as polymeric immunoglobulin receptor (pIgR), cryptdin-related sequence 1C (CRS1C), and Reg3γ. We found that dietary Gln or Arg supplementation decreased bacterial colonization and promoted the activation of innate immunity (e.g., the mRNA expression of pIgR, CRS1C, and Reg3γ) in the intestine of ETEC-infected mice. Our results suggest that dietary arginine or glutamine supplementation may inhibit intestinal ETEC infection through intestinal innate immunity.


Asunto(s)
Arginina/farmacología , Infecciones por Escherichia coli/tratamiento farmacológico , Glutamina/farmacología , Inmunidad Innata/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Animales , Antiinfecciosos/metabolismo , Carga Bacteriana/efectos de los fármacos , Suplementos Dietéticos , Escherichia coli Enterotoxigénica/efectos de los fármacos , Infecciones por Escherichia coli/inmunología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Íleon/efectos de los fármacos , Íleon/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/fisiopatología , Yeyuno/efectos de los fármacos , Yeyuno/inmunología , Ratones , Sustancias Protectoras/farmacología , Transducción de Señal/efectos de los fármacos
15.
Amino Acids ; 49(12): 1981-1997, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28536843

RESUMEN

Calcific aortic valve disease is a common, severe heart condition that is currently with no proven, effective drug treatment and requires a surgical valve replacement or an entire heart explanation. Thus, developing novel, targeted therapeutic approaches becomes a major goal for cardiovascular disease research. To achieve this goal, isolated heart valve interstitial cells could be an advanced model to explore molecular mechanisms and measure drug efficacy. Based on this progress, molecular mechanisms that harbor components of  inflammation and fibrosis coupled with proteins, for example, BMP-2, TLRs, RANKL, Osteoprotegerin, have been proposed. Small molecules or antibodies targeting these proteins have shown promising efficacy for either reversing or slowing down calcification development in vitro. In this review, we summarize these potential therapeutics with some highlights of interstitial cellular models.


Asunto(s)
Estenosis de la Válvula Aórtica , Válvula Aórtica/patología , Calcinosis , Modelos Biológicos , Animales , Válvula Aórtica/citología , Válvula Aórtica/fisiopatología , Estenosis de la Válvula Aórtica/tratamiento farmacológico , Estenosis de la Válvula Aórtica/fisiopatología , Biomarcadores Farmacológicos/metabolismo , Calcinosis/tratamiento farmacológico , Calcinosis/fisiopatología , Descubrimiento de Drogas , Fibrosis/metabolismo , Fibrosis/fisiopatología , Humanos , Inflamación/metabolismo , Inflamación/fisiopatología , Transducción de Señal/fisiología
16.
Amino Acids ; 49(12): 1999-2007, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28534176

RESUMEN

Pregnane X receptor (PXR, NR1I2), a member of the nuclear receptor superfamily, is a crucial regulator of nutrient metabolism and metabolic detoxification such as metabolic syndrome, xenobiotic metabolism, inflammatory responses, glucose, cholesterol and lipid metabolism, and endocrine homeostasis. Notably, much experimental and clinical evidence show that PXR senses xenobiotics and triggers the detoxification response to prevent diseases such as diabetes, obesity, intestinal inflammatory diseases and liver fibrosis. In this review we summarize recent advances on remarkable metabolic and regulatory versatility of PXR, and we emphasizes its role and potential implication as an effective modulator of self-detoxification in animals and humans.


Asunto(s)
Enfermedades Metabólicas/metabolismo , Receptor Cross-Talk/fisiología , Receptores de Esteroides/metabolismo , Transducción de Señal/fisiología , Xenobióticos/metabolismo , Animales , Biomarcadores Farmacológicos , Humanos , Inactivación Metabólica , Terapia Molecular Dirigida , Receptor X de Pregnano , Procesamiento Proteico-Postraduccional , Receptores de Esteroides/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología
18.
Can J Microbiol ; 63(4): 312-320, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28177794

RESUMEN

Haemophilus parasuis can cause a severe membrane inflammation disorder. It has been documented that superoxide dismutase (SOD) is a potential target to treat systemic inflammatory diseases. Therefore, we constructed an experimental H. parasuis subunit vaccine SOD and determined the protective efficacy of SOD using a lethal dose challenge against H. parasuis serovar 4 strain MD0322 and serovar 5 strain SH0165 in a mouse model. The results demonstrated that SOD could induce a strong humoral immune response in mice and provide significant immunoprotection efficacy against a lethal dose of H. parasuis serovar 4 strain MD0322 or serovar 5 strain SH0165 challenge. IgG subtype analysis indicated SOD protein could trigger a bias toward a Th1-type immune response and induce the proliferation of splenocytes and secretion of IL-2 and IFN-γ of splenocytes. In addition, serum in mice from the SOD-immunized group could inhibit the growth of strain MD0322 and strain SH0165 in the whole-blood killing bacteria assay. This is the first report that immunization of mice with SOD protein could provide protective effect against a lethal dose of H. parasuis serovar 4 and serovar 5 challenge in mice, which may provide a novel approach against heterogeneous serovar infection of H. parasuis in future.


Asunto(s)
Vacunas contra Haemophilus/inmunología , Haemophilus parasuis/inmunología , Superóxido Dismutasa/inmunología , Animales , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Haemophilus parasuis/enzimología , Inmunización , Ratones , Ratones Endogámicos BALB C , Vacunas Sintéticas/inmunología
19.
J Biol Chem ; 290(11): 7234-46, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25616664

RESUMEN

Autophagy is an intracellular degradation pathway and is considered to be an essential cell survival mechanism. Defects in autophagy are implicated in many pathological processes, including inflammatory bowel disease. Among the innate defense mechanisms of intestinal mucosa, a defective tight junction (TJ) barrier has been postulated as a key pathogenic factor in the causation and progression of inflammatory bowel disease by allowing increased antigenic permeation. The cross-talk between autophagy and the TJ barrier has not yet been described. In this study, we present the novel finding that autophagy enhances TJ barrier function in Caco-2 intestinal epithelial cells. Nutrient starvation-induced autophagy significantly increased transepithelial electrical resistance and reduced the ratio of sodium/chloride paracellular permeability. Nutrient starvation reduced the paracellular permeability of small-sized urea but not larger molecules. The role of autophagy in the modulation of paracellular permeability was confirmed by pharmacological induction as well as pharmacological and genetic inhibition of autophagy. Consistent with the autophagy-induced reduction in paracellular permeability, a marked decrease in the level of the cation-selective, pore-forming TJ protein claudin-2 was observed after cell starvation. Starvation reduced the membrane presence of claudin-2 and increased its cytoplasmic, lysosomal localization. Therefore, our data show that autophagy selectively reduces epithelial TJ permeability of ions and small molecules by lysosomal degradation of the TJ protein claudin-2.


Asunto(s)
Autofagia , Claudina-2/metabolismo , Células Epiteliales/citología , Mucosa Intestinal/citología , Proteolisis , Uniones Estrechas/metabolismo , Células CACO-2 , Células Epiteliales/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Permeabilidad
20.
Am J Physiol Regul Integr Comp Physiol ; 311(2): R365-73, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27225947

RESUMEN

Pro-inflammatory cytokines play a critical role in the pathophysiology of muscle atrophy. We hypothesized that glycine exerted an anti-inflammatory effect and alleviated lipopolysaccharide (LPS)-induced muscle atrophy in piglets. Pigs were assigned to four treatments including the following: 1) nonchallenged control, 2) LPS-challenged control, 3) LPS+1.0% glycine, and 4) LPS+2.0% glycine. After receiving the control, 1.0 or 2.0% glycine-supplemented diets, piglets were treated with either saline or LPS. At 4 h after treatment with saline or LPS, blood and muscle samples were harvested. We found that 1.0 or 2.0% glycine increased protein/DNA ratio, protein content, and RNA/DNA ratio in gastrocnemius or longissimus dorsi (LD) muscles. Glycine also resulted in decreased mRNA expression of muscle atrophy F-box (MAFbx) and muscle RING finger 1 (MuRF1) in gastrocnemius muscle. In addition, glycine restored the phosphorylation of Akt, mammalian target of rapamycin (mTOR), eukaryotic initiation factor 4E binding protein 1 (4E-BP1), and Forkhead Box O 1 (FOXO1) in gastrocnemius or LD muscles. Furthermore, glycine resulted in decreased plasma tumor necrosis factor-α (TNF-α) concentration and muscle TNF-α mRNA abundance. Moreover, glycine resulted in decreased mRNA expresson of Toll-like receptor 4 (TLR4), nucleotide-binding oligomerization domain protein 2 (NOD2), and their respective downstream molecules in gastrocnemius or LD muscles. These results indicate glycine enhances muscle protein mass under an inflammatory condition. The beneficial roles of glycine on the muscle are closely associated with maintaining Akt-mTOR-FOXO1 signaling and suppressing the activation of TLR4 and/or NOD2 signaling pathways.


Asunto(s)
Glicina/administración & dosificación , Atrofia Muscular/tratamiento farmacológico , Atrofia Muscular/fisiopatología , Proteína Adaptadora de Señalización NOD2/metabolismo , Proteína Oncogénica v-akt/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Femenino , Proteína Forkhead Box O1/metabolismo , Glicina/farmacología , Lipopolisacáridos , Masculino , Proteínas Musculares/biosíntesis , Atrofia Muscular/inducido químicamente , Tamaño de los Órganos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Porcinos , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA