Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
DNA Cell Biol ; 40(6): 776-790, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34029124

RESUMO

Glaesserella parasuis causes porcine Glässer's disease and lipopolysaccharide (LPS) induces acute inflammation and pathological damage. Baicalin has antioxidant, antimicrobial, and anti-inflammatory functions. Long noncoding RNAs (lncRNAs) play key regulatory functions during bacterial infection. However, the role of lncRNAs in the vascular dysfunction induced by a combination of G. parasuis and LPS during systemic inflammation and the effect of baicalin on lncRNA expression induced in porcine aortic vascular endothelial cells (PAVECs) by a combination of G. parasuis and LPS have not been investigated. In this study, we investigated the changes in lncRNA and mRNA expression induced in PAVECs by G. parasuis, LPS, or a combination of G. parasuis and LPS, and the action of baicalin on lncRNA expression induced in PAVECs by the combination of G. parasuis and LPS. Our results showed 133 lncRNAs and 602 genes were differentially expressed when PAVECs were stimulated with the combination of G. parasuis and LPS, whereas 107 lncRNAs and 936 genes were differentially expressed when PAVECs were stimulated with the combination of G. parasuis and LPS after pretreatment with baicalin. The Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed the dominant signaling pathways triggered by the combination of G. parasuis and LPS were the tumor necrosis factor signaling pathway, phosphatidylinositol signaling system, and inositol phosphate metabolism. Protein-protein interaction network analysis showed the differentially expressed target genes of the differentially expressed lncRNAs (DELs) were related to each other. A coexpression analysis indicated the expression levels of the DELs were co-regulated with those of their differentially expressed target genes. This is the first study to systematically compare the changes in lncRNAs and mRNAs in PAVECs stimulated with a combination of G. parasuis and LPS. Our data clarified the mechanisms underlying the vascular inflammation and damage triggered by G. parasuis and LPS, and it may provide novel targets for the treatment of LPS-induced systemic inflammation.


Assuntos
Anti-Inflamatórios , Células Endoteliais , Flavonoides , Inflamação , Infecções por Pasteurellaceae/veterinária , Doenças dos Suínos/tratamento farmacológico , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Linhagem Celular , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Inflamação/tratamento farmacológico , Inflamação/veterinária , Pasteurellaceae , Infecções por Pasteurellaceae/tratamento farmacológico , RNA Longo não Codificante , RNA Mensageiro/genética , Suínos , Doenças dos Suínos/microbiologia , Transcriptoma
2.
DNA Cell Biol ; 40(7): 881-894, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33945308

RESUMO

Baicalin-aluminum regulates the gut microbiome of piglets with diarrhea. However, whether it affects poultry gut microbiome composition and function remains unknown. In this study, we used metagenomic sequencing to explore the effects of baicalin-aluminum on gut microbiome changes in poultry when compared with animals administered colistin sulfate. Our data showed that important gut microbiome components consisted of Ruminococcaceae, Subdoligranulum, Bifidobacterium, Bifidobacterium pseudolongum, and Pseudoflavonifractor when broilers were administered baicalin-aluminum compared with colistin. At the species level, Lactobacillus salivarius, Bacteroides uniformis, Oscillibacter unclassified, Bacteroides fragilis, Ruminococcus torques, and Subdoligranulum unclassified abundance were significantly upregulated upon baicalin-aluminum treatment when compared with colistin administration. In addition, Gene Ontology (GO) enrichment analysis indicated that functional differentially expressed genes, which were in the top 30 GO enrichment terms, were associated with metabolic processes, catalytic activity, and cellular processes. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis demonstrated that ABC transporters, oxidative phosphorylation, and phosphotransferase systems were the dominant signaling pathways in the baicalin-aluminum group when compared with the colistin group. Taken together, our data indicated that baicalin-aluminum modified broiler gut microbiome composition. These observations enhance our physiological insights of baicalin-aluminum-mediated functions in the broiler microbiome and potentially provide a novel therapy to manage both animal and human health.


Assuntos
Galinhas/microbiologia , Flavonoides/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Alumínio/metabolismo , Alumínio/farmacologia , Animais , China , Colistina/farmacologia , Flavonoides/metabolismo , Aves Domésticas/microbiologia
3.
Vet Res ; 51(1): 102, 2020 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-32795339

RESUMO

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.


Assuntos
Anti-Infecciosos/uso terapêutico , Flavonoides/uso terapêutico , Infecções por Haemophilus/veterinária , Haemophilus parasuis/fisiologia , Substâncias Protetoras/uso terapêutico , Doenças dos Suínos/prevenção & controle , Animais , Relação Dose-Resposta a Droga , Infecções por Haemophilus/microbiologia , Infecções por Haemophilus/prevenção & controle , Sus scrofa , Suínos , Doenças dos Suínos/microbiologia
4.
Oxid Med Cell Longev ; 2020: 5803639, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32411329

RESUMO

Oxidative stress can lead to intestinal cell injury as well as the induction of inflammation. It is not clear whether inflammation is an important factor leading to cell injury caused by oxidative stress. The purpose of this study was to investigate the role of inflammation in intestinal injury caused by hydrogen peroxide (H2O2). Our results revealed that H2O2 stimulation significantly decreased the viability of intestinal porcine epithelial cells (IPEC-1), increased lactate dehydrogenase (LDH) activity, and disrupted the distribution of the tight junction protein claudin-1. H2O2 significantly increased the mRNA expression of interleukin-6 (IL-6), IL-8, and tumor necrosis factor-α (TNF-α). H2O2 stimulation also led to increased phosphorylation of p38 and jun N-terminal kinase (JNK), and p65 NF-κB protein translocation into the nucleus of IPEC-1 cells. Cells treated with the NF-κB inhibitor (BAY11-7082), the p38 inhibitor (SB202190), or the JNK inhibitor (PD98059) significantly decreased mRNA and protein expression of IL-6, IL-8, and TNF-α. However, treatment with mitogen-activated protein kinase (MAPK) or NF-κB inhibitors did not prevent the damage effect on cell viability, LDH activity, or the distribution of claudin-1 in cells challenged with H2O2. In summary, our data demonstrate that activation of the NF-κB and MAPK signaling pathways can contribute to the inflammatory response, but not cell injury, in IPEC-1 cells challenged with H2O2.


Assuntos
Peróxido de Hidrogênio/toxicidade , Inflamação/patologia , Sistema de Sinalização das MAP Quinases , NF-kappa B/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Fluorescência , Inflamação/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , L-Lactato Desidrogenase/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Suínos , Proteínas de Junções Íntimas/metabolismo , Fator de Transcrição RelA/metabolismo
5.
DNA Cell Biol ; 39(5): 801-815, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32096672

RESUMO

Haemophilus parasuis can elicit serious inflammatory responses, which contribute to huge economic losses to the swine industry. However, the pathogenic mechanisms underlying inflammation-related damage induced by H. parasuis remain unclear. Accumulating evidence indicates that long non-coding RNAs (lncRNAs) have important functions in the regulation of autoimmune disorders. Baicalin has been shown to have anti-inflammatory, anti-microbial, and anti-oxidant activities. In this study, we investigated whether lncRNAs were involved in the vascular injury or inflammation triggered by H. parasuis and whether baicalin regulated the lncRNA profiles of porcine aortic vascular endothelial cells (PAVECs) infected with H. parasuis. The results showed that the lncRNA and mRNA expression profiles of PAVECs were changed by H. parasuis. Important functions of lncRNAs and mRNAs were predicted. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses demonstrated that the targets of differentially expressed lncRNAs of H. parasuis infected PAVECs were mainly involved in the tumor necrosis factor (TNF) signaling pathway, apoptosis, and N-glycan biosynthesis; whereas nicotinate and nicotinamide metabolism, the cytosolic DNA-sensing pathway, the TNF signaling pathway, and the nuclear factor (NF)-kappa B signaling pathway were enriched in PAVECs pretreated with baicalin. In addition, top hub genes and lncRNAs were identified and validated by quantitative polymerase chain reaction. CCL5, GBP1, and SAMHD1 were significantly upregulated after H. parasuis infection, whereas they were significantly downregulated with baicalin pretreatment. LncRNA ALDBSSCT0000001677, ALDBSSCT0000001353, MSTRG.10724.2, and ALDBSSCT0000010434 had the same expression pattern. Collectively, these data suggested that baicalin could modify changes to the lncRNAs profiles or regulate lncRNAs that participate in inflammation-related signaling pathways, thereby alleviating tissue damage or inflammatory responses induced by H. parasuis. To our best knowledge, this is the first article of H. parasuis stimulating changes to the lncRNA profiles of PAVECs and the capability of baicalin to regulate lncRNA changes in PAVECs infected with H. parasuis, which might provide a novel therapeutic target for the control of H. parasuis infection.


Assuntos
Aorta/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Flavonoides/farmacologia , Haemophilus parasuis/fisiologia , RNA Longo não Codificante/genética , Transcriptoma/efeitos dos fármacos , Animais , Células Endoteliais/microbiologia , RNA Mensageiro/genética , Suínos
6.
FASEB J ; 34(2): 2483-2496, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31909535

RESUMO

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.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Ácido Eicosapentaenoico/farmacologia , Células Epiteliais , Mucosa Intestinal , Necroptose/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Tricotecenos/toxicidade , Animais , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Mucosa Intestinal/lesões , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Suínos
7.
Int J Mol Sci ; 20(10)2019 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-31091773

RESUMO

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.


Assuntos
Anti-Infecciosos/farmacologia , Fezes/microbiologia , Flavonoides/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Alumínio/química , Animais , Anti-Infecciosos/química , Anti-Infecciosos/uso terapêutico , Diarreia/tratamento farmacológico , Diarreia/veterinária , Flavonoides/química , Flavonoides/uso terapêutico , Suínos , Doenças dos Suínos/tratamento farmacológico
8.
BMC Vet Res ; 15(1): 98, 2019 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-30909903

RESUMO

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.


Assuntos
Antibacterianos/uso terapêutico , Flavonoides/uso terapêutico , Haemophilus parasuis/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Monócitos/metabolismo , Doenças dos Suínos/tratamento farmacológico , Animais , Animais Recém-Nascidos/metabolismo , Animais Recém-Nascidos/microbiologia , Antibacterianos/farmacologia , Apoptose/efeitos dos fármacos , Relação Dose-Resposta a Droga , Flavonoides/farmacologia , Infecções por Haemophilus/tratamento farmacológico , Infecções por Haemophilus/metabolismo , Infecções por Haemophilus/veterinária , Monócitos/efeitos dos fármacos , Monócitos/microbiologia , Espécies Reativas de Oxigênio/metabolismo , Suínos , Doenças dos Suínos/metabolismo , Doenças dos Suínos/microbiologia
9.
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
10.
Int J Mol Sci ; 19(5)2018 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-29710817

RESUMO

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.


Assuntos
Células Endoteliais/metabolismo , Infecções por Haemophilus/genética , Transcriptoma , Animais , Células Cultivadas , Células Endoteliais/microbiologia , Endotélio Vascular/citologia , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Infecções por Haemophilus/metabolismo , Haemophilus parasuis/patogenicidade , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Suínos
11.
Amino Acids ; 49(12): 2091-2098, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28929442

RESUMO

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.


Assuntos
Metionina/metabolismo , Estresse Oxidativo/fisiologia , Animais , Doenças Cardiovasculares/fisiopatologia , Humanos , Imunidade Inata , Nefropatias/fisiopatologia , Metabolismo dos Lipídeos , Hepatopatias/fisiopatologia , Neoplasias/fisiopatologia , Doenças do Sistema Nervoso/fisiopatologia
12.
Nutrients ; 9(9)2017 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-28832517

RESUMO

Inflammatory bowel disease (IBD), which includes both ulcerative colitis and Crohn's disease, is a chronic relapsing inflammation of the gastrointestinal tract, and is difficult to treat. The pathophysiology of IBD is multifactorial and not completely understood, but genetic components, dysregulated immune responses, oxidative stress, and inflammatory mediators are known to be involved. Animal models of IBD can be chemically induced, and are used to study etiology and to evaluate potential treatments of IBD. Currently available IBD treatments can decrease the duration of active disease but because of their adverse effects, the search for novel therapeutic strategies that can restore intestinal homeostasis continues. This review summarizes and discusses what is currently known of the effects of amino acids on the reduction of inflammation, oxidative stress, and cell death in the gut when IBD is present. Recent studies in animal models have identified dietary amino acids that improve IBD, but amino acid supplementation may not be adequate to replace conventional therapy. The animal models used in dietary amino acid research in IBD are described.


Assuntos
Aminoácidos/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Colite Ulcerativa/tratamento farmacológico , Doença de Crohn/tratamento farmacológico , Fármacos Gastrointestinais/uso terapêutico , Intestinos/efeitos dos fármacos , Animais , Colite Ulcerativa/imunologia , Colite Ulcerativa/metabolismo , Colite Ulcerativa/fisiopatologia , Doença de Crohn/imunologia , Doença de Crohn/metabolismo , Doença de Crohn/fisiopatologia , Modelos Animais de Doenças , Humanos , Mucosa Intestinal/metabolismo , Intestinos/imunologia , Intestinos/fisiopatologia
13.
Amino Acids ; 49(8): 1277-1291, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28616751

RESUMO

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.


Assuntos
Aminoácidos/metabolismo , Enteropatias/metabolismo , Enteropatias/prevenção & controle , Animais , Humanos
14.
Amino Acids ; 49(12): 1981-1997, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28536843

RESUMO

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.


Assuntos
Estenose da Valva Aórtica , Valva Aórtica/patologia , Calcinose , Modelos Biológicos , Animais , Valva Aórtica/citologia , Valva Aórtica/fisiopatologia , Estenose da Valva Aórtica/tratamento farmacológico , Estenose da Valva Aórtica/fisiopatologia , Biomarcadores Farmacológicos/metabolismo , Calcinose/tratamento farmacológico , Calcinose/fisiopatologia , Descoberta de Drogas , Fibrose/metabolismo , Fibrose/fisiopatologia , Humanos , Inflamação/metabolismo , Inflamação/fisiopatologia , Transdução de Sinais/fisiologia
15.
Amino Acids ; 49(12): 1915-1929, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28271166

RESUMO

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.


Assuntos
Acetilcisteína/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Intestino Delgado/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Transdução de Sinais/efeitos dos fármacos , Sus scrofa , Animais , Suplementos Nutricionais , Receptores ErbB/metabolismo , Sequestradores de Radicais Livres/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Mucosa Intestinal/fisiopatologia , Intestino Delgado/fisiopatologia , Oxirredução/efeitos dos fármacos , Suínos , Desmame
16.
Amino Acids ; 49(12): 1931-1943, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28258325

RESUMO

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.


Assuntos
Acetilcisteína/administração & dosagem , Acetilcisteína/farmacologia , Infecções por Coronavirus/veterinária , Mucosa Intestinal/efeitos dos fármacos , Intestino Delgado/efeitos dos fármacos , Vírus da Diarreia Epidêmica Suína , Doenças dos Suínos/tratamento farmacológico , Animais , Animais Recém-Nascidos , Infecções por Coronavirus/tratamento farmacológico , Suplementos Nutricionais , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Mucosa Intestinal/patologia , Mucosa Intestinal/fisiopatologia , Intestino Delgado/patologia , Intestino Delgado/fisiopatologia , Oxirredução/efeitos dos fármacos , Plasma/efeitos dos fármacos , Plasma/enzimologia , Sus scrofa , Suínos , Aumento de Peso/efeitos dos fármacos
17.
Innate Immun ; 23(1): 34-43, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28064564

RESUMO

Infection and inflammation can result in the rapid loss of muscle mass and myofibrillar proteins (muscle atrophy). In addition, aspartate (Asp) is necessary for protein synthesis in mammalian cells. We hypothesized that Asp could attenuate LPS-induced muscle atrophy in a piglet model. Twenty-four weaning piglets were allotted to four treatments, including non-challenged control, LPS challenged control, LPS+0.5% Asp and LPS+1.0% Asp. On d 21, the piglets were injected with i.p. injection of LPS (100 ug/kg BM) or saline. At 4 h post-injection, blood, gastrocnemius and longissimus dorsi muscles samples were collected for biochemical and molecular analyses. Asp decreased the concentrations of cortisol and glucagon in plasma. In addition, Asp increased protein and RNA contents in muscles, and decreased mRNA expression of muscle atrophy F-box (MAFbx) and muscle RING finger 1 (MuRF1). Moreover, Asp decreased phosphorylation of AMPKα but increased phosphorylation of Akt and Forkhead Box O (FOXO) 1 in the muscles. Our results indicate that Asp suppresses LPS-induced MAFbx and MuRF1 expression via activation of Akt signaling, and inhibition of AMPKα and FOXO1 signaling.


Assuntos
Anti-Inflamatórios/farmacologia , Ácido Aspártico/farmacologia , Proteínas F-Box/metabolismo , Músculo Esquelético/efeitos dos fármacos , Atrofia Muscular/tratamento farmacológico , Músculos Paraespinais/efeitos dos fármacos , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Animais , Células Cultivadas , Modelos Animais de Doenças , Proteínas F-Box/genética , Proteína Forkhead Box O1/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glucagon/sangue , Humanos , Hidrocortisona/sangue , Lipopolissacarídeos/imunologia , Músculo Esquelético/fisiologia , Atrofia Muscular/induzido quimicamente , Proteína Oncogênica v-akt/metabolismo , Músculos Paraespinais/fisiologia , Proteínas Quinases/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Transdução de Sinais/efeitos dos fármacos , Suínos , Proteínas com Motivo Tripartido/genética
18.
J Exp Clin Cancer Res ; 36(1): 8, 2017 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-28069043

RESUMO

BACKGROUND: Sorafenib, a multi-kinase inhibitor, is used as a standard therapy for advanced hepatocellular carcinoma (HCC). However, complete remission has not been achieved and the molecular basis of HCC resistance to sorafenib remains largely unknown. Previous studies have shown that fibroblast growth factor 19 (FGF19) expression correlates with tumor progression and poor prognosis of HCC. Here, we demonstrate the novel role of FGF19 in HCC resistance to sorafenib therapy. METHODS: FGF19 Knockdown cells were achieved by lentiviral-mediated interference, and FGFR4 knockout cells were achieved by CRISPR-Cas9. Protein levels of FGF19, FGFR4 and c-PARP in various HCC cell lines were measured by Western blotting analysis. Cell viability was determined by MTS assay, apoptosis was determined by DAPI nuclear staining and Western blot of c-PRAP, and ROS generation was determined by DCFH-DA staining and electrochemical biosensor. RESULTS: We showed that FGF19, when overexpressed, inhibited the effect of sorafenib on ROS generation and apoptosis in HCC. In contrast, loss of FGF19 or its receptor FGFR4 led to a remarkable increase in sorafenib-induced ROS generation and apoptosis. In addition, knockdown of FGF19 in sorafenib-resistant HCC cells significantly enhanced the sensitivity to sorafenib. Importantly, targeting FGF19/FGFR4 axis by ponatinib, a third-generation inhibitor of chronic myeloid leukemia, overcomes HCC resistance of sorafenib by enhancing ROS-associated apoptosis in sorafenib-treated HCC. CONCLUSION: Our results provide the first evidence that inhibition of FGF19/FGFR4 signaling significantly overcomes sorafenib resistance in HCC. Co-treatment of ponatinib and sorafinib may represent an effective therapeutic approach for eradicating HCC.


Assuntos
Carcinoma Hepatocelular/genética , Resistencia a Medicamentos Antineoplásicos , Fatores de Crescimento de Fibroblastos/genética , Neoplasias Hepáticas/genética , Niacinamida/análogos & derivados , Compostos de Fenilureia/farmacologia , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/genética , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Niacinamida/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Sorafenibe , Regulação para Cima
19.
Oxid Med Cell Longev ; 2017: 1670815, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29348786

RESUMO

Patients suffering from cardiovascular diseases (CVDs) experience a low quality of life and increase pressure on healthcare systems both nationally and globally. DNA methylation, which refers to the pathway by which DNA methyltransferase facilitates the addition of a methyl group to DNA, is of critical importance in this respect primarily because the epigenetic modification is implicated in a range of serious conditions including atherosclerosis, CVDs, and cancer. Research findings indicate that the number of epigenetic alterations can be elicited (both in utero and in adults) through the administration of certain nutritional supplements, including folic acid and methionine; this is partly attributable to the effect employed by methyl-containing nutrients in DNA methylation. Thus, for the purpose of illuminating viable therapeutic measures and preventive strategies for CVDs, research should continue to explore the intricate associations that exist between epigenetic regulation and CVD pathogenesis. This review centers on an exposition of the mechanism by which DNA methylation takes place, the impact it has on a range of conditions, and the potential clinical value of nutrition, driven mainly by the observation that nutritional supplements such as folic acid can affect DNA methylation.


Assuntos
Doenças Cardiovasculares/genética , Doenças Cardiovasculares/patologia , Metilação de DNA , Suplementos Nutricionais , Ácido Fólico/farmacologia , Animais , Epigênese Genética , Humanos , Estado Nutricional
20.
Cancer Med ; 5(11): 3336-3345, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27748080

RESUMO

Autophagy has been linked with melanoma risk and survival, but no polymorphisms in autophagy-related (ATG) genes have been investigated in relation to melanoma progression. We examined five single-nucleotide polymorphisms (SNPs) in three ATG genes (ATG5; ATG10; and ATG16L) with known or suspected impact on autophagic flux in an international population-based case-control study of melanoma. DNA from 911 melanoma patients was genotyped. An association was identified between (GG) (rs2241880) and earlier stage at diagnosis (OR 0.47; 95% Confidence Intervals (CI) = 0.27-0.81, P = 0.02) and a decrease in Breslow thickness (P = 0.03). The ATG16L heterozygous genotype (AG) (rs2241880) was associated with younger age at diagnosis (P = 0.02). Two SNPs in ATG5 were found to be associated with increased stage (rs2245214 CG, OR 1.47; 95% CI = 1.11-1.94, P = 0.03; rs510432 CC, OR 1.84; 95% CI = 1.12-3.02, P = 0.05). Finally, we identified inverse associations between ATG5 (GG rs2245214) and melanomas on the scalp or neck (OR 0.20, 95% CI = 0.05-0.86, P = 0.03); ATG10 (CC) (rs1864182) and brisk tumor infiltrating lymphocytes (TILs) (OR 0.42; 95% CI = 0.21-0.88, P = 0.02), and ATG5 (CC) (rs510432) with nonbrisk TILs (OR 0.55; 95% CI = 0.34-0.87, P = 0.01). Our data suggest that ATG SNPs might be differentially associated with specific host and tumor characteristics including age at diagnosis, TILs, and stage. These associations may be critical to understanding the role of autophagy in cancer, and further investigation will help characterize the contribution of these variants to melanoma progression.


Assuntos
Autofagia/genética , Predisposição Genética para Doença , Variação Genética , Melanoma/epidemiologia , Melanoma/genética , Vigilância da População , Adulto , Idoso , Alelos , Proteínas Relacionadas à Autofagia/genética , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Estadiamento de Neoplasias , Razão de Chances , Polimorfismo de Nucleotídeo Único
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA