Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Molecules ; 26(7)2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33916022

ABSTRACT

Korean Red ginseng (KRG), commonly used in traditional medicine, has anti-inflammatory, anti- oxidative, and anti-tumorigenic properties. Asian sand dust (ASD) is known to aggravate upper and lower airway inflammatory responses. BEAS-2B cells were exposed to ASD with or without KRG or ginsenoside Rg3. Mucin 5AC (MUC5AC), MUC5B, and MUC8 mRNA and protein expression levels were determined using quantitative RT-PCR and enzyme-linked immunosorbent assay. Nuclear factor kappa B (NF-κB), activator protein 1, and mitogen-activated protein kinase expression and activity were determined using western blot analysis. ASD induced MUC5AC, MUC5B, and MUC8 mRNA and protein expression in BEAS-2B cells, which was significantly inhibited by KRG and Rg3. Although ASD-induced mucin expression was associated with NF-κB and p38 mitogen-activated protein kinase (MAPK) activity, KRG and Rg3 significantly suppressed only ASD-induced NF-κB expression and activity. KRG and Rg3 inhibited ASD-induced mucin gene expression and protein production from bronchial epithelial cells. These results suggest that KRG and Rg3 have potential for treating mucus-producing airway inflammatory diseases.


Subject(s)
Dust , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Regulation , Ginsenosides/pharmacology , Mucins/genetics , Panax/chemistry , Sand , Cell Line , Cell Survival/drug effects , Cells, Cultured , Ginsenosides/chemistry , Humans , Molecular Structure , Mucin 5AC/biosynthesis , Mucin 5AC/genetics , Mucin-5B/biosynthesis , Mucin-5B/genetics , Mucins/biosynthesis , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism
2.
Am J Physiol Gastrointest Liver Physiol ; 319(4): G421-G431, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32755385

ABSTRACT

The loss of the intestinal Na+/H+ exchanger isoform 8 (NHE8) results in an ulcerative colitis-like condition with reduction of mucin production and dysbiosis, indicating that NHE8 plays an important role in intestinal mucosal protection. The aim of this study was to investigate the potential rebalance of the altered microbiota community of NHE8-deficient mice via fecal microbiota transplantation (FMT) and feeding probiotic VSL#3. We also aimed to stimulate mucin production by sodium butyrate administration via enema. Data from 16S rRNA sequencing showed that loss of NHE8 contributes to colonic microbial dysbiosis with reduction of butyrate-producing bacteria. FMT increased bacterial adhesion in the colon in NHE8 knockout (NHE8KO) mice. Periodic-acid Schiff reagent (PAS) stain and quantitative PCR showed no changes in mucin production during FMT. In mice treated with the probiotic VSL#3, a reduction of Lactobacillus and segmented filamentous bacteria (SFB) in NHE8KO mouse colon was detected and an increase in goblet cell theca was observed. In NHE8KO mice receiving sodium butyrate (NaB), 1 mM NaB stimulated Muc2 expression without changing goblet cell theca, but 10 mM NaB induced a significant reduction of goblet cell theca without altering Muc2 expression. Furthermore, 5 mM and 10 mM NaB-treated HT29-MTX cells displayed increased apoptosis, while 0.5 mM NaB stimulated Muc2 gene expression. These data showed that loss of NHE8 leads to dysbiosis with reduction of butyrate-producing bacteria and FMT and VSL#3 failed to rebalance the microbiota in NHE8KO mice. Therefore, FMT, VSL#3, and NaB are not able to restore mucin production in the absence of NHE8 in the intestine.NEW & NOTEWORTHY Loss of Na+/H+ exchanger isoform 8 (NHE8), a Slc9 family of exchanger that contributes to sodium uptake, cell volume regulation, and intracellular pH homeostasis, resulted in dysbiosis with reduction of butyrate-producing bacteria and decrease of Muc2 production in the intestine in mice. Introducing fecal microbiota transplantation (FMT) and VSL#3 in NHE8 knockout (NHE8KO) mice failed to rebalance the microbiota in these mice. Furthermore, administration of FMT, VSL#3, and sodium butyrate was unable to restore mucin production in the absence of NHE8 in the intestine.


Subject(s)
Intestinal Mucosa/physiology , Sodium-Hydrogen Exchangers/physiology , Animals , Butyrates/metabolism , Butyric Acid/administration & dosage , Colon/microbiology , Dysbiosis/etiology , Dysbiosis/microbiology , Dysbiosis/therapy , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/physiology , Goblet Cells/drug effects , Goblet Cells/physiology , HT29 Cells , Humans , Lactobacillus/physiology , Mice , Mice, Knockout , Mucins/biosynthesis , Probiotics/administration & dosage , Sodium-Hydrogen Exchangers/deficiency
3.
Toxicol Appl Pharmacol ; 394: 114959, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32201329

ABSTRACT

Arsenic is a ubiquitous environmental toxicant, found in high concentrations worldwide. Although abundant research has dealt with arsenic-induced cancers, studies on mechanisms of non-malignant lung diseases have not been complete. In addition, decades of research have mostly concentrated on high-dose arsenic exposure, which has very limited use in modeling the biological effects of today's low-dose exposures. Indeed, accumulated evidence has shown that low-dose arsenic exposure (i.e. ≤100 ppb) may also alter lung homeostasis by causing host susceptibility to viral infection. However, the underlying mechanism of this alteration is unknown. In this study, we found that low-dose sodium arsenite (As (III)) repressed major airway mucins-MUC5AC and MUC5B at both mRNA and protein levels. We further demonstrated that this repression was not caused by cellular toxicity or mediated by the reduction of a common mucin-inducing pathway-EGFR. Other established mucin activators- dsRNA, IL1ß or IL17 were not able to override As (III)-induced mucin repression. Interestingly, the suppressing effect of As (III) appeared to be partially reversible, and supplementation of all trans retinoic acid (t-RA) doses dependently restored mucin gene expression. Further analyses indicated that As (III) treatment significantly reduced the protein level of retinoic acid receptors (RARα, γ and RXRα) as well as RARE promoter reporter activity. Therefore, our study fills in an important knowledge gap in the field of low-dose arsenic exposure. The interference of RA signaling, and mucin gene expression may be important pathogenic factors in low-dose arsenic induced lung toxicity.


Subject(s)
Arsenic/toxicity , Mucins/biosynthesis , Respiratory Mucosa/metabolism , Signal Transduction/drug effects , Tretinoin , Arsenites/toxicity , Cell Line , Cell Survival/drug effects , ErbB Receptors/antagonists & inhibitors , Gene Expression/drug effects , Gene Expression Regulation/drug effects , Humans , Mucin 5AC/antagonists & inhibitors , Mucin 5AC/genetics , Mucin-5B/antagonists & inhibitors , Mucin-5B/genetics , Respiratory Mucosa/drug effects , Sodium Compounds/toxicity
4.
Biomed Res Int ; 2019: 7084734, 2019.
Article in English | MEDLINE | ID: mdl-30941370

ABSTRACT

Butyrate produced by the intestinal microbiota is essential for proper functioning of the intestinal immune system. Total dependence on parenteral nutrition (PN) is associated with numerous adverse effects, including severe microbial dysbiosis and loss of important butyrate producers. We hypothesised that a lack of butyrate produced by the gut microbiota may be compensated by its supplementation in PN mixtures. We tested whether i.v. butyrate administration would (a) positively modulate intestinal defence mechanisms and (b) counteract PN-induced dysbiosis. Male Wistar rats were randomised to chow, PN, and PN supplemented with 9 mM butyrate (PN+But) for 12 days. Antimicrobial peptides, mucins, tight junction proteins, and cytokine expression were assessed by RT-qPCR. T-cell subpopulations in mesenteric lymph nodes (MLN) were analysed by flow cytometry. Microbiota composition was assessed in caecum content. Butyrate supplementation resulted in increased expression of tight junction proteins (ZO-1, claudin-7, E-cadherin), antimicrobial peptides (Defa 8, Rd5, RegIIIγ), and lysozyme in the ileal mucosa. Butyrate partially alleviated PN-induced intestinal barrier impairment and normalised IL-4, IL-10, and IgA mRNA expression. PN administration was associated with an increase in Tregs in MLN, which was normalised by butyrate. Butyrate increased the total number of CD4+ and decreased a relative amount of CD8+ memory T cells in MLN. Lack of enteral nutrition and PN administration led to a shift in caecal microbiota composition. Butyrate did not reverse the altered expression of most taxa but did influence the abundance of some potentially beneficial/pathogenic genera, which might contribute to its overall beneficial effect.


Subject(s)
Butyrates/pharmacology , Dietary Supplements , Gastrointestinal Microbiome , Intestines/pathology , Parenteral Nutrition , Animals , Biodiversity , Colon/drug effects , Colon/pathology , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation/drug effects , Ileum/drug effects , Ileum/pathology , Intestine, Small/drug effects , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , Models, Animal , Mucins/biosynthesis , Paneth Cells/drug effects , Paneth Cells/metabolism , Peptides/genetics , Peptides/metabolism , Permeability , Phenotype , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Tight Junction Proteins/metabolism
5.
Nutrition ; 59: 182-187, 2019 03.
Article in English | MEDLINE | ID: mdl-30504005

ABSTRACT

OBJECTIVES: The aim of this study was to investigate the effects of dietary l-threonine supplementation on the growth performance, intestinal immune function, mucin synthesis, and goblet cell differentiation in weanling piglets with intrauterine growth retardation (IUGR). METHODS: Eighteen litters of newborn piglets were selected at birth, with one normal birthweight (NBW) and two IUGR piglets in each litter. At weaning, the NBW piglet and one of the IUGR piglets were assigned to groups fed a basal diet (i.e., the NBW-CON and IUGR-CON groups). The other IUGR piglet was assigned to a group fed the basal diet supplemented with 2 g l-threonine per kg of diet (i.e., IUGR-Thr group). Therefore, all piglets were distributed across three groups for a 3-wk feeding trial. RESULTS: Compared with NBW, IUGR decreased growth performance, increased ileal proinflammatory cytokine levels, and reduced ileal mucin 2 (Muc2) content and goblet cell density of weanling piglets. Supplementation of l-threonine increased the feed efficiency of the IUGR-Thr group compared with the IUGR-CON group. The l-threonine-supplemented diet attenuated ileal inflammatory responses of the IUGR-Thr piglets and increased production of Muc2 and secretory immunoglobulin A and density of goblet cells. In addition, L-threonine supplementation downregulated δ-like 1 and hes family bHLH transcription factor 1, whereas growth factor independence 1 and Kruppel-like factor 4 expression levels were upregulated. CONCLUSION: Dietary l-threonine supplementation attenuates inflammatory responses, facilitates Muc2 synthesis, and promotes goblet cell differentiation in the ileum of IUGR piglets.


Subject(s)
Dietary Supplements , Fetal Growth Retardation/diet therapy , Intestines/drug effects , Mucins/biosynthesis , Threonine/pharmacology , Animal Feed , Animals , Fetal Growth Retardation/immunology , Intestinal Mucosa/metabolism , Intestines/immunology , Swine , Weaning
6.
J Anim Sci ; 97(2): 756-767, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30452657

ABSTRACT

An experiment was conducted to test the hypothesis that pigs fed diets supplemented with exogenous phytase reduce mucin synthesis in the small intestine, increase protein hydrolysis in the stomach, increase breakdown of phytate along the gastrointestinal tract, and increase mineral and AA digestibility. A diet based on corn, soybean meal, and canola meal was formulated to meet requirements for growing pigs except for Ca and P, which were lower than requirements. Three additional diets were formulated by adding 750, 1,500, or 3,000 units of phytase (FTU) per kilogram to the basal diet. Eight growing barrows (38.45 ± 3.06 kg) were prepared with a T-cannula in the duodenum and another T-cannula in the distal ileum. Pigs were housed individually and allotted to a replicated 4 × 4 Latin square design with four pigs and four periods in each square. Each period lasted 14 d with the initial 7 d being the adaptation period to the diets. Pigs were fed twice daily in combined amounts equal to 3.2 times the estimated requirement for maintenance energy. Results indicated that the apparent ileal digestibility (AID) and the apparent total tract digestibility (ATTD) of Ca and P increased (linear and quadratic, P ≤ 0.05) as phytase inclusion increased. However, values for AID of Ca and P were not different from values for ATTD of Ca and P, indicating that there is no net absorption of Ca and P in the hindgut. The apparent duodenal digestibility (ADD) of Ca and P was ~30% and 10% to 20%, respectively, indicating some digestion in the stomach of both Ca and P. A quadratic increase (P < 0.05) of the AID of GE was observed with the breakpoint around 1,500 FTU, but there was a negative linear (P ≤ 0.001) effect of dietary phytase on the ATTD of GE. Phytase did not affect mucin synthesis in the small intestine, protein hydrolysis in the stomach, or ileal digestibility of dispensable and indispensable AA. However, degradation of higher phytate esters (IP6 and IP5) into lower phytate esters (IP4 and IP3) and inositol increased as dietary phytase increased, indicating that it is possible to completely degrade dietary phytate if microbial phytase is included by at least 3,000 FTU in the diet. In conclusion, supplementing diets with phytase resulted in increased degradation of phytate and phytate esters and improved digestibility of Ca and P, but phytase did not change intestinal mucin synthesis, gastric protein hydrolysis, or the AID of AA.


Subject(s)
6-Phytase/pharmacology , Digestion/drug effects , Gastrointestinal Tract/drug effects , Mucins/biosynthesis , Phytic Acid/metabolism , Swine/growth & development , 6-Phytase/administration & dosage , Animal Feed/analysis , Animals , Diet/veterinary , Dietary Proteins/metabolism , Gastrointestinal Tract/physiology , Hydrolysis , Male , Phosphorus, Dietary/metabolism , Swine/physiology
7.
Food Chem Toxicol ; 118: 880-888, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29935245

ABSTRACT

The intestinal mucus layer plays an important role in the management of inflammatory bowel disease. The aim of this study was to investigate the effects of oxyresveratrol (OXY), an antioxidant, on the stimulation of mucin production in human LS 174T goblet cells and the underlying mechanism thereof. OXY increased MUC2 expression at both the mRNA and protein levels. By performing two-dimensional gel electrophoresis, we found that the expression of nicotinic acid phosphoribosyltransferase1 (NaPRT1) in OXY-treated LS 174T cells was greatly increased compared with that in negative control cells. In addition, the NAD+/NADH ratio was increased in proportion to OXY in LS 174T cells. The expression of NAD+-synthesis enzymes, NaPRT1, nicotinamide riboside kinase1 (NRK1) and nicotinamide mononucleotide adenylyltransferase1 (Nmnat1) was significantly increased at both the mRNA and protein levels in OXY-treated LS 174T cells. The inhibition of NaPRT1 and NRK1 did not decrease MUC2 expression after inhibiting by small interfering RNA (siRNA)-NaPRT1 and siRNA-NRK1, respectively; however, inhibition of Nmnat by an Nmnat inhibitor decreased MUC2 expression in a dose-dependent manner. In conclusion, OXY increases NAD+ levels, resulting in the stimulation of MUC2 expression in LS 174T cells. These findings present a novel role for NAD+ in stimulation of MUC2 expression.


Subject(s)
Goblet Cells/metabolism , Intestinal Mucosa/metabolism , Mucins/biosynthesis , NAD/metabolism , Plant Extracts/pharmacology , Stilbenes/pharmacology , Cell Line , Electrophoresis, Gel, Two-Dimensional , Humans , Intestinal Mucosa/cytology , Mucin-2/metabolism , N-Acetylgalactosaminyltransferases/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Polypeptide N-acetylgalactosaminyltransferase
8.
J Agric Food Chem ; 65(22): 4472-4483, 2017 Jun 07.
Article in English | MEDLINE | ID: mdl-28502165

ABSTRACT

Airway epithelial cells are often infected by respiratory syncytial virus (RSV), one of the most common causes of asthma, bronchiolitis, chronic obstructive pulmonary disease, and pneumonia. During the infection process, excessive mucins instigate airway inflammation. However, the mechanism underlying RSV-induced airway hyper-responsiveness and inflammation is poorly understood. Furthermore, no reliable vaccines or drugs for antiviral therapy are available. In this study, the effect of the natural compound grape seed proanthocyanidin (GSP) on RSV-infected human airway epithelial cells A549 was evaluated. After pretreatment of the cells with or without exposure to RSV with 5-10 µg GSP/mL, the expression of various mucins (MUC1, MUC2, MUC5AC, MUC5B, and MUC8) was evaluated by real-time polymerase chain reaction, enzyme-linked immunosorbent assay, and Western blotting, as well as confocal microscopy. We found that GSP significantly decreased RSV-induced mucin synthesis at the mRNA and protein levels. In addition, GSP suppressed the RSV-induced signaling pathways, including extracellular signal-regulated kinase, c-Jun N-terminal kinase, and p38, together with nuclear factor kappa B (NF-κB) and activating protein-1 family members (c-Jun and c-Fos). Concomitantly, GSP inhibited the replication of RSV within A549 cells. Taken together, all our results suggest that GSP could be a potent therapeutic agent to suppress excessive mucus production and viral replication in RSV-induced airway inflammatory disorders.


Subject(s)
Grape Seed Extract/pharmacology , MAP Kinase Signaling System/drug effects , Mucins/biosynthesis , Proanthocyanidins/pharmacology , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/physiology , A549 Cells , Humans , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/virology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Virus Replication , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Int J Mol Sci ; 18(2)2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28216579

ABSTRACT

Proteinase inhibitors have been associated with anti-inflammatory and antioxidant activities and may represent a potential therapeutic treatment for emphysema. Our aim was to evaluate the effects of a plant Kunitz proteinase inhibitor, Enterolobium contortisiliquum trypsin inhibitor (EcTI), on several aspects of experimental elastase-induced pulmonary inflammation in mice. C57/Bl6 mice were intratracheally administered elastase (ELA) or saline (SAL) and were treated intraperitoneally with EcTI (ELA-EcTI, SAL-EcTI) on days 1, 14 and 21. On day 28, pulmonary mechanics, exhaled nitric oxide (ENO) and number leucocytes in the bronchoalveolar lavage fluid (BALF) were evaluated. Subsequently, lung immunohistochemical staining was submitted to morphometry. EcTI treatment reduced responses of the mechanical respiratory system, number of cells in the BALF, and reduced tumor necrosis factor-α (TNF-α), matrix metalloproteinase-9 (MMP-9), matrix metalloproteinase-12 (MMP-12), tissue inhibitor of matrix metalloproteinase (TIMP-1), endothelial nitric oxide synthase (eNOS) and inducible nitric oxide synthase (iNOS)-positive cells and volume proportion of isoprostane, collagen and elastic fibers in the airways and alveolar walls compared with the ELA group. EcTI treatment reduced elastase induced pulmonary inflammation, remodeling, oxidative stress and mechanical alterations, suggesting that this inhibitor may be a potential therapeutic tool for chronic obstructive pulmonary disease (COPD) management.


Subject(s)
Airway Remodeling/drug effects , Fabaceae/chemistry , Pancreatic Elastase/metabolism , Plant Extracts/pharmacology , Pneumonia/metabolism , Pneumonia/physiopathology , Protease Inhibitors/pharmacology , Animals , Biomarkers , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Extracellular Matrix/metabolism , Male , Mice , Mucins/biosynthesis , Oxidative Stress , Pneumonia/drug therapy , Pneumonia/pathology
10.
Nihon Rinsho Meneki Gakkai Kaishi ; 39(6): 522-527, 2016.
Article in Japanese | MEDLINE | ID: mdl-28049961

ABSTRACT

The intestinal epithelium constitutes a physical barrier between inner and outer side of our body. It also functions as a "hub" which connects factors that determine the development of inflammatory bowel disease, such as microbiota, susceptibility genes, and host immune response. Accordingly, recent studies have implicated and further featured the role of intestinal epithelial cell dysfunction in the pathophysiology of inflammatory bowel disease. For example, mucin producing goblet cells are usually "depleted" in ulcerative colitis patients. Studies have shown that those goblet cells exhibit various immune-regulatory functions in addition to mucin production, such as antigen presentation or cytokine production. Paneth cells are another key cell lineage that has been deeply implicated in the pathophysiology of Crohn's disease. Several susceptibility genes for Crohn's disease may lead to impairment of anti-bacterial peptide production and secretion by Paneth cells. Also, other susceptibility genes may determine the survival of Paneth cells, which leads to reduced Paneth cell function in the patient small intestinal mucosa. Further studies may reveal other unexpected roles of the intestinal epithelium in the pathophysiology of inflammatory bowel disease, and may help to develop alternative therapies targeted to intestinal epithelial cell functions.


Subject(s)
Epithelial Cells/physiology , Inflammatory Bowel Diseases/etiology , Intestines/cytology , Antigen Presentation , Antimicrobial Cationic Peptides/metabolism , Cytokines/biosynthesis , Gastrointestinal Microbiome , Genetic Predisposition to Disease/genetics , Goblet Cells/metabolism , Goblet Cells/pathology , Goblet Cells/physiology , Humans , Immunity , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/therapy , Molecular Targeted Therapy , Mucins/biosynthesis , Paneth Cells/metabolism , Paneth Cells/physiology
11.
Inflamm Bowel Dis ; 21(7): 1541-52, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25901972

ABSTRACT

BACKGROUND: The role of hnRNP A1 in the onset of intestinal inflammation remains unclear. This study investigated the function of hnRNP A1 in mice enteritis models. METHODS: C57Bl6/J mice were intraperitoneally injected with anti-CD3 antibodies to develop enteritis. In the DSS-induced colitis group, the mice were allowed free access to 3% DSS solution in their drinking water for 5 days. 3H-mannitol flux and complementary DNA array tests were used to assess the intestinal barrier function and messenger RNA (mRNA) expression, respectively. Real-time PCR was performed after immunoprecipitation with anti-hnRNP antibodies to determine the specific mRNA binding of hnRNP A1. RESULTS: The hnRNP A1 expression was increased in the intestine of the mouse at 24 hours after treatment with anti-CD3 antibodies and 5 days after starting DSS administration. Small interfering RNA (siRNA) against hnRNP A1 exacerbated the intestinal injuries in both models. According to the microarray analysis, trefoil factor 2 (TFF2) was identified as a candidate molecule targeted by hnRNP A1 in the anti-CD3 antibody-induced enteritis group. Moreover, the binding between hnRNP A1 and TFF2 mRNA significantly increased in the enteritis mice, and the administration of siRNA against either hnRNP A1 or TFF2 exacerbated the degree of intestinal injury. In the DSS-induced colitis group, treatment with the siRNA of hnRNP A1 worsened the intestinal injury, while the expression of TFF3 did not change. CONCLUSIONS: hnRNP A1 improves intestinal injury in anti-CD3 antibody-induced enteritis mice through the upregulation of TFF2, which regulates apoptosis and enhances epithelial restoration, whereas this molecule ameliorates DSS-induced colitis through a different pathway.


Subject(s)
Apoptosis , Enteritis/genetics , Gene Expression Regulation , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Mucins/genetics , Muscle Proteins/genetics , Peptides/genetics , RNA, Messenger/genetics , Animals , Blotting, Western , CD3 Complex/immunology , Cells, Cultured , Disease Models, Animal , Enteritis/immunology , Enteritis/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/biosynthesis , In Situ Nick-End Labeling , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mucins/biosynthesis , Muscle Proteins/biosynthesis , Real-Time Polymerase Chain Reaction , Trefoil Factor-2
12.
Int J Mol Sci ; 15(4): 5907-15, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24717412

ABSTRACT

In this study, the effect of Codonopsis lanceolata fermented by lactic acid on controlling gene expression levels related to obesity was observed in an oligonucleotide chip microarray. Among 8170 genes, 393 genes were up regulated and 760 genes were down regulated in feeding the fermented C. lanceolata (FCL). Another 374 genes were up regulated and 527 genes down regulated without feeding the sample. The genes were not affected by the FCL sample. It was interesting that among those genes, Chytochrome P450, Dmbt1, LOC76487, and thyroid hormones, etc., were mostly up or down regulated. These genes are more related to lipid synthesis. We could conclude that the FCL possibly controlled the gene expression levels related to lipid synthesis, which resulted in reducing obesity. However, more detailed protein expression experiments should be carried out.


Subject(s)
Codonopsis/metabolism , Obesity/drug therapy , Obesity/genetics , Plant Extracts/pharmacology , Animals , Calcium-Binding Proteins , Cytochrome P-450 Enzyme System/biosynthesis , DNA-Binding Proteins , Fermentation , Gene Expression Profiling , Gene Expression Regulation , Lactic Acid/chemistry , Lipids/biosynthesis , Liver/enzymology , Liver/metabolism , Male , Mice , Mice, Inbred ICR , Mucins/biosynthesis , Oligonucleotide Array Sequence Analysis , Plants, Medicinal/metabolism , Thyroid Hormones/biosynthesis , Tumor Suppressor Proteins
13.
Free Radic Biol Med ; 68: 101-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24333275

ABSTRACT

Neutrophil-mediated tissue injury is a shared pathogenesis of both chronic pulmonary diseases and acute responses to pathogens, allergens, and airborne pollutants. Interventions to minimize toxic effects of neutrophil-derived oxidants and proteases are usually limited to corticosteroids, which can have adverse side effects. We used a rodent model of endotoxin-induced lung injury to test the hypothesis that the dietary supplement γ-tocopherol (γT), a natural form of vitamin E with antioxidant and novel anti-inflammatory properties, will protect from adverse nasal and pulmonary inflammatory responses induced by endotoxin (lipopolysaccharide; LPS). Male Fisher F344 rats were intranasally (i.n.) instilled with LPS for 2 consecutive days. Beginning 2 days before i.n. LPS, the rats were gavaged daily with 30mg/kg γT. Twenty-four hours after the last i.n. LPS, bronchoalveolar lavage fluid (BALF) was collected, and pulmonary and nasal tissues were analyzed for gene expression and morphometric analyses of neutrophils and intraepithelial mucosubstances (IM). LPS caused increased BALF total cells (70% increase), neutrophils (300%), protein (35%), PGE2 (500%), and secreted mucins (75%). Robust increases in neutrophils and IM were detected in conducting airways. Pulmonary expression of MUC5AC, MIP-2, CINC-1, and MCP-1 was elevated three- to eightfold by LPS. Treatment with γT inhibited LPS-induced increases in BALF total cells, neutrophils, protein, PGE2, and secreted mucins, as well as IM and tissue neutrophil influx. Furthermore γT induced the expression of the regulatory cytokines IL-10 and IFN-γ while decreasing MUC5AC, MIP-2, CINC-1, and MCP-1. These data demonstrate novel therapeutic effects of the dietary vitamin E γT promoting anti-inflammatory pathways to protect from neutrophil-mediated lung injury.


Subject(s)
Lung Injury/drug therapy , Respiratory Mucosa/drug effects , gamma-Tocopherol/administration & dosage , Animals , Bronchoalveolar Lavage Fluid , Dietary Supplements , Gene Expression Regulation/drug effects , Lipopolysaccharides/toxicity , Lung Injury/chemically induced , Lung Injury/pathology , Mucins/biosynthesis , Mucins/genetics , Nasal Mucosa/drug effects , Nasal Mucosa/metabolism , Neutrophils , Rats , Respiratory Mucosa/metabolism
14.
Phytomedicine ; 21(2): 172-6, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24060215

ABSTRACT

In the present study, we investigated whether aqueous extract of Liriope Tuber, ophiopogonin D and spicatoside A derived from Liriope Tuber affect basal or phorbol ester (phorbol 12-myristate 13-acetate, PMA)-induced airway mucin production and secretion from airway epithelial cells. Confluent NCI-H292 cells were treated with each agent for 24 h (basal production) or pretreated with each agent for 30 min and then stimulated with PMA for 24 h (PMA-induced production and secretion), respectively. MUC5AC airway mucin production and secretion were measured by ELISA. The results were as follows: (1) aqueous extract of Liriope Tuber stimulated basal mucin production and did not inhibit but increased PMA-induced mucin production; (2) ophiopogonin D and spicatoside A stimulated basal mucin production and did not inhibit but increased PMA-induced mucin production; (3) two compounds increased PMA-induced mucin secretion. These results suggest that ophiopogonin D and spicatoside A can increase mucin production and secretion, by directly acting on airway epithelial cells and, at least in part, explain the traditional use of aqueous extract of Liriope Tuber as expectorants in diverse inflammatory pulmonary diseases.


Subject(s)
Epithelial Cells/drug effects , Liriope Plant/chemistry , Mucins/metabolism , Plant Extracts/pharmacology , Respiratory Mucosa/drug effects , Saponins/metabolism , Saponins/pharmacology , Spirostans/pharmacology , Cell Line , Epithelial Cells/metabolism , Humans , Mucins/biosynthesis , Phorbol Esters/pharmacology , Respiratory Mucosa/metabolism
15.
Clin Exp Allergy ; 43(9): 1071-82, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23957343

ABSTRACT

BACKGROUND: Asthma is a chronic disease characterized by airways hyperresponsiveness, inflammation and airways remodelling involving reversible bronchial obstruction. Omega-3 fatty acids and their derivatives are known to reduce inflammation in several tissues including lung. OBJECTIVES: The effects of eicosapentaenoic acid monoacylglyceride (MAG-EPA), a newly synthesized EPA derivative, were determined on the resolution of lung inflammation and airway hyperresponsiveness in an in vivo model of allergic asthma. METHODS: Ovalbumin (OVA)-sensitized guinea-pigs were treated or not with MAG-EPA administered per os. Isometric tension measurements, histological analyses, homogenate preparation for Western blot experiments or total RNA extraction for RT-PCR were performed to assess the effect of MAG-EPA treatments. RESULTS: Mechanical tension measurements revealed that oral MAG-EPA treatments reduced methacholine (MCh)-induced bronchial hyperresponsiveness in OVA-sensitized guinea-pigs. Moreover, MAG-EPA treatments also decreased Ca(2+) hypersensitivity of bronchial smooth muscle. Histological analyses and leucocyte counts in bronchoalveolar lavages revealed that oral MAG-EPA treatments led to less inflammatory cell recruitment in the lung of OVA-sensitized guinea-pigs when compared with lungs from control animals. Results also revealed a reduction in mucin production and MUC5AC expression level in OVA-sensitized animals treated with MAG-EPA. Following MAG-EPA treatments, the transcript levels of pro-inflammatory markers such as IL-5, eotaxin, IL-13 and IL-4 were markedly reduced. Moreover, per os MAG-EPA administrations reduced COX2 over-expression in OVA-sensitized animals. CONCLUSION AND CLINICAL RELEVANCE: We demonstrate that MAG-EPA reduces airway hyperresponsiveness and lung inflammation in OVA-sensitized animals, a finding consistent with a decrease in IL-4, IL-5, IL-13, COX-2 and MUC5AC expression levels in the lung. The present data suggest that MAG-EPA represents a new potential therapeutic strategy for resolving inflammation in allergic asthma.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Asthma/drug therapy , Monoglycerides/pharmacology , Allergens/immunology , Animals , Anti-Asthmatic Agents/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Asthma/chemically induced , Asthma/metabolism , Asthma/pathology , Bronchial Hyperreactivity/drug therapy , Bronchial Hyperreactivity/immunology , Cyclooxygenase 2/metabolism , Disease Models, Animal , Fatty Acids/blood , Fatty Acids/metabolism , Female , Guinea Pigs , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Monoglycerides/administration & dosage , Mucins/biosynthesis , Ovalbumin/adverse effects , Receptors, Chemokine/metabolism
17.
J Gastroenterol Hepatol ; 27(11): 1745-51, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22849613

ABSTRACT

BACKGROUND AND AIM: The increasing prevalence of cholesterol gallstone (CG) disease has become an economic burden to the healthcare system. Ursodeoxycholic acid (UDCA) is the only established medical agent used to dissolve gallstones. In investigating novel therapeutics for CG, we assessed the preventive effects of n-3 polyunsaturated fatty acids (n-3PUFA) on the formation of CG induced by feeding a lithogenic diet (LD) containing high cholesterol levels to mice. METHODS: Mice were divided into the following six groups: (A) regular diet (RD); (B) RD+n-3PUFA; (C) LD; (D) LD+n-3PUFA; (E) LD+UDCA; (F) LD+n-3PUFA+UDCA. After RD/LD feeding for 2 weeks, n-3PUFA or UDCA was administered orally and the diet maintained for 8 weeks. The levels of phospholipids and cholesterol in bile, CG formation, gallbladder wall thickness, MUC gene expression in gallbladder were analyzed. RESULTS: No stone or sludge was evident in the RD groups (Groups A, B). Mice in the n-3PUFA treatment (Groups D, F) showed significantly lower stone formation than the other LD groups (Groups C, E). The combination treatment of n-3PUFA and UDCA suppressed stone formation more than mono-therapy with n-3PUFA or UDCA. Bile phospholipid levels were significantly elevated in the Group F. Hypertrophy of the gallbladder wall was evident in mice fed LD. MUC 2, 5AC, 5B and 6 mRNA expression levels were significantly elevated in the LD-fed group, and this was suppressed by n-3PUFA with or without UDCA. CONCLUSIONS: N-3PUFA attenuated gallstone formation in mouse, through increasing the levels of bile phospholipids and suppressing bile mucin formation.


Subject(s)
Cholesterol, Dietary/therapeutic use , Fatty Acids, Omega-3/therapeutic use , Gallstones/prevention & control , Mucins/biosynthesis , Animals , Bile/metabolism , Cholagogues and Choleretics/pharmacology , Cholagogues and Choleretics/therapeutic use , Cholesterol/metabolism , Cholesterol, Dietary/pharmacology , Drug Therapy, Combination , Fatty Acids, Omega-3/pharmacology , Gallbladder/pathology , Gene Expression/drug effects , Male , Mice , Mice, Inbred C57BL , Mucin 5AC/genetics , Mucin-2/genetics , Mucin-5B/genetics , Mucin-6/genetics , Mucins/drug effects , Mucins/genetics , Mucous Membrane/pathology , Phospholipids/metabolism , RNA, Messenger/metabolism , Statistics, Nonparametric , Ursodeoxycholic Acid/pharmacology , Ursodeoxycholic Acid/therapeutic use
18.
Curr Eye Res ; 37(1): 33-42, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22029601

ABSTRACT

PURPOSE: Retinoic acid (RA) is essential for epithelial differentiation and maintenance of the mucous phenotype. This study investigated the effect of RA on corneal epithelial differentiation and mucin expression in a primary human corneal limbal epithelial cell (HCLEC) culture model. METHODS: HCLECs were grown in RA-supplemented media at various concentrations (0, 10(-9) to 10(-6) M). Stratified HCLECs were examined using immunohistochemical or immunofluorescent staining for p63, ABCG2, CK3, CK19, and Western blotting for ABCG2 and CK12 to assess differentiation. Ultrastructural morphology was investigated using scanning and transmission electron microscopy. They were incubated with rose bengal dye to examine barrier function. The effects of RA on the expression of MUC1, -4, and -16 were analyzed by immunohistochemistry, quantitative real-time PCR and Western blot analysis. RESULTS: HCLEC grown without RA showed hyperkeratosis, whereas those grown with 10(-8) to 10(-7) M RA induced non-keratinized stratified epithelium with a normal appearance. Under these conditions, p63, ABCG2, CK3, CK19, MUC1, -4, and -16 staining patterns were similar to in vivo limbal epithelium. A higher concentration (10(-6) M) of RA resulted in abnormal differentiation. HCLECs grown with RA were tightly apposed and maintained intact barrier function against dye penetration. In addition, MUC1, -4, and -16 expressions were highly associated with RA concentrations. CONCLUSIONS: This study showed that cultured HCLEC could mimic physiologic and functional phenotypes by controlling RA concentrations in medium. Also, our results suggested modulating effect of RA on differentiation and mucin expression in corneal epithelium.


Subject(s)
Cell Differentiation/drug effects , Epithelium, Corneal/ultrastructure , Gene Expression Regulation/drug effects , Limbus Corneae/drug effects , Mucins/biosynthesis , RNA/genetics , Tretinoin/pharmacology , Blotting, Western , Cells, Cultured , Epithelium, Corneal/drug effects , Epithelium, Corneal/metabolism , Humans , Immunohistochemistry , Keratolytic Agents/pharmacology , Limbus Corneae/metabolism , Limbus Corneae/ultrastructure , Microscopy, Electron, Scanning Transmission , Mucins/genetics , Reverse Transcriptase Polymerase Chain Reaction
19.
Phytother Res ; 25(8): 1196-200, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21305630

ABSTRACT

This study investigated whether prunetin significantly affects the secretion, production and gene expression of mucin from cultured airway epithelial cells. Confluent primary rat tracheal surface epithelial (RTSE) cells were pretreated with adenosine triphosphate (ATP) for 5 min and then chased for 30 min in the presence of prunetin to assess the effect on mucin secretion using enzyme-linked immunosorbent assay (ELISA). At the same time, confluent NCI-H292 cells were pretreated with prunetin for 30 min and then stimulated with epidermal growth factor (EGF) or phorbol 12-myristate 13-acetate (PMA) for 24 h, respectively. The MUC5AC mucin gene expression and mucin protein production were measured by reverse transcription-polymerase chain reaction (RT-PCR) and ELISA. The results were as follows: (1) prunetin significantly suppressed ATP-induced mucin secretion from cultured RTSE cells; (2) prunetin inhibited the production of MUC5AC mucin protein induced by EGF or PMA from NCI-H292 cells; (3) prunetin also inhibited the expression of MUC5AC mucin gene induced by EGF or PMA from NCI-H292 cells. This result suggests that prunetin can regulate the secretion, production and gene expression of mucin, by directly acting on airway epithelial cells.


Subject(s)
Epithelial Cells/drug effects , Isoflavones/pharmacology , Mucins/antagonists & inhibitors , Respiratory Mucosa/drug effects , Adenosine Triphosphate/pharmacology , Animals , Cell Line, Tumor , Cells, Cultured , Epithelial Cells/metabolism , Gene Expression/drug effects , Glycyrrhiza/chemistry , Humans , Male , Mucin 5AC/antagonists & inhibitors , Mucin 5AC/biosynthesis , Mucin 5AC/genetics , Mucins/biosynthesis , Mucins/genetics , Mucins/metabolism , Rats , Rats, Sprague-Dawley , Respiratory Mucosa/cytology , Tetradecanoylphorbol Acetate/pharmacology , Trachea/metabolism
20.
Br Poult Sci ; 51(5): 658-66, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21058070

ABSTRACT

1. An experiment was conducted to evaluate the interactive effects of manno-oligosaccharides (MOS; Bio-MOS®) and dietary threonine on the growth performance in relation to intestinal mucin dynamics in broiler chickens from 1 to 21 and from 22 to 35 d of age. Two concentrations of MOS (0 or 2 g/kg for d 1 to 21; and 0 or 1 g/kg for d 22 to 35) and three concentrations of threonine (0.0, 1.0 and 1.2 of National Research Council (NRC), 1994, recommendations) were included in the experimental diets for each age group. 2. Body weight gain was significantly lower in threonine-deficient birds compared with those fed on adequate or excess threonine diets. Positive interaction between MOS and threonine supplementation on body weight gain was apparent in all phases of growth due mainly to the significantly poorer performance of birds given excess threonine in the absence of MOS. 3. The duodenal and ileal adherent mucous thickness were reduced at 14 and 28 d in threonine-deficient birds. Nevertheless, MOS significantly increase duodenal adherent mucous thickness at 14 d and ileal mucous thickness at 14 and 28 d. At 14 d, a significant MOS and threonine interaction on the jejunal adherent mucous thickness was also noted in that there was no difference between adequate and excess threonine groups in the absence of MOS, but a significant increase with excess threonine and MOS supplementation. 4. Dietary threonine greatly influenced mucin synthesis at the translational stage with no effect on jejunal MUC2 gene expression. Conversely, MOS modulated the transcriptional stage of intestinal mucin synthesis by consistently up-regulating jejunal MUC2 gene expression which was independent of dietary threonine concentration. There were no significant interactions between threonine and MOS on all the goblet cell densities. However, there was a MOS and threonine interaction on the staining intensities of jejunal sulphomucins due mainly to the significantly lower staining intensities in birds fed excess threonine in the absence of MOS. 5. The ameliorative effect of MOS on the growth-suppressive effects of excess threonine is likely to be linked to its modulating effects on the intestinal mucin dynamics.


Subject(s)
Chickens/metabolism , Gastrointestinal Tract/metabolism , Mucins/biosynthesis , Oligosaccharides/pharmacology , Threonine/pharmacology , Animal Feed , Animals , Chickens/growth & development , Dietary Supplements , Gene Expression Regulation , Male , Oligosaccharides/metabolism , Threonine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL