Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
PLoS One ; 12(8): e0182550, 2017.
Article in English | MEDLINE | ID: mdl-28792520

ABSTRACT

The aim of this study was to compare the effect of dietary supplementation with low dose of porous and nano zinc oxide (ZnO) on weaning piglets, and to evaluate the possibility of using them as an alternative to high dose of regular ZnO. Piglets were randomly allocated into four treatment groups fed with four diets: (1) basal diet (NC), (2) NC+ 3000 mg/kg ZnO (PC), (3) NC + 500 mg/kg porous ZnO (HiZ) and (4) NC + 500 mg/kg nano ZnO (ZNP). The result showed that piglets in HiZ group had less diarrhea than ZNP group (P < 0.05). Besides, there was no significant difference between PC, HiZ and ZNP groups in terms of serum malondialdeyhde (MDA) concentration and glutathione peroxidase (GSH-Px) activity (P > 0.05). Analysis of trace metal elements revealed that piglets fed with high dose of regular ZnO had the highest Zn level in kidney (P < 0.05), which may induce kidney stone formation. Additionally, a decrease in ileal crypt depth was observed in PC, HiZ and ZNP group, suggesting an effective protection against intestinal injury. Results of mRNA analysis in intestine showed that ZNP supplementation had better effects on up-regulated trefoil factor 3 (TFF3) and nuclear factor erythroid 2-related factor 2 (Nrf2) levels in duodenum and jejunum than HiZ did (P < 0.05), implying that nano ZnO may possess higher anti-inflammatory capacity than porous ZnO. In conclusion, dietary supplementation with low dose of porous and nano ZnO had similar (even better) effect on improving growth performance and intestinal morphology, reducing diarrhea and intestinal inflammatory as high dose of regular ZnO in weaning piglets. Compared with nano ZnO, porous ZnO had better performance on reducing diarrhea but less effect on up-regulation of intestinal TFF3 and Nrf2.


Subject(s)
Animal Feed , Dietary Supplements , Swine , Zinc Oxide/administration & dosage , Animals , Diarrhea/prevention & control , Diarrhea/veterinary , Glutathione Peroxidase/metabolism , Intestines/anatomy & histology , Intestines/growth & development , Intestines/immunology , Intestines/injuries , Kidney/growth & development , Kidney/metabolism , Kidney Calculi/prevention & control , Kidney Calculi/veterinary , Malondialdehyde/blood , Muscles/metabolism , NF-E2-Related Factor 2/metabolism , Particle Size , Porosity , RNA, Messenger/metabolism , Swine/growth & development , Swine Diseases/prevention & control , Trefoil Factor-3/metabolism , Weaning , Zinc/analysis , Zinc Oxide/chemistry
2.
Biomed Pharmacother ; 85: 280-286, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27876210

ABSTRACT

BACKGROUND: Strengthening of intestinal tight junctions provides an effective barrier from the external environment. Goblet cell-derived trefoil factor 3 (TFF3) increases transepithelial resistance by upregulating the expression of tight junction proteins. Oxyresveratrol (OXY) is a hydroxyl-substituted stilbene found in the roots, leaves, stems, and fruit of many plants and known to have various biological activities. In this study, we investigated the strengthening effect of OXY on intestinal tight junctions through stimulation of TFF production in goblet cells. METHODS: We prepared conditioned medium from LS 174T goblet cells treated with OXY (GCO-CM) and investigated the effect of GCO-CM on strengthening tight junctions of Caco-2 cells. The mRNA and protein expression levels of major tight junction components (claudin-1, occludin, and ZO-1) were measured by quantitative real-time PCR and western blotting, respectively. Transepithelial electric resistance (TEER) was measured using an ohm/V meter. Monolayer permeability was evaluated by paracellular transport of fluorescein isothiocyanate-dextran. RESULTS: OXY showed a strong antioxidant activity. It significantly increased the expression level of TFF3 in LS 174T goblet cells. GCO-CM prepared by treatment with 2.5, 5, and 10µg/ml OXY did not show cytotoxicity in Caco-2 cells. GCO-CM increased the mRNA and protein expression levels of claudin-1, occludin, and ZO-1. It also significantly increased tight junction integrity and reduced permeability in a dose-dependent manner. CONCLUSION: OXY stimulates the expression of TFF3 in goblet cells, which might increase the integrity of the intestinal tight junction barrier.


Subject(s)
Culture Media, Conditioned/metabolism , Gastrointestinal Agents/pharmacology , Goblet Cells/drug effects , Intestinal Mucosa/drug effects , Paracrine Communication/drug effects , Plant Extracts/pharmacology , Stilbenes/pharmacology , Tight Junctions/drug effects , Antioxidants/pharmacology , Caco-2 Cells , Claudin-1/genetics , Claudin-1/metabolism , Dose-Response Relationship, Drug , Electric Impedance , Goblet Cells/metabolism , Humans , Intestinal Mucosa/metabolism , Occludin/genetics , Occludin/metabolism , Permeability , Tight Junctions/metabolism , Time Factors , Trefoil Factor-3/genetics , Trefoil Factor-3/metabolism , Up-Regulation , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
3.
Amino Acids ; 47(5): 1053-63, 2015 May.
Article in English | MEDLINE | ID: mdl-25691144

ABSTRACT

TFF3 is a member of the trefoil factor family (TFF) predominantly secreted by mucous epithelia. Minute amounts are also expressed in the immune system and the brain. In the latter, particularly the hypothalamo-pituitary axis has been investigated in detail in the past. Functionally, cerebral TFF3 has been reported to be involved in several processes such as fear, depression, learning and object recognition, and opiate addiction. Furthermore, TFF3 has been linked with neurodegenerative and neuropsychiatric disorders (e.g., Alzheimer's disease, schizophrenia, and alcoholism). Here, using immunohistochemistry, a systematic survey of the TFF3 localization in the adult human brain is presented focusing on extrahypothalamic brain areas. In addition, the distribution of TFF3 in the developing human brain is described. Taken together, neurons were identified as the predominant cell type to express TFF3, but to different extent; TFF3 was particularly enriched in various midbrain and brain stem nuclei. Besides, TFF3 immunostaining staining was observed in oligodendroglia and the choroid plexus epithelium. The wide cerebral distribution should help to explain its multiple effects in the CNS.


Subject(s)
Choroid Plexus/metabolism , Mesencephalon/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Peptides/genetics , Abortion, Spontaneous , Adult , Amygdala/chemistry , Amygdala/metabolism , Brain Mapping , Cerebellum/chemistry , Cerebellum/metabolism , Cerebral Cortex/chemistry , Cerebral Cortex/metabolism , Choroid Plexus/chemistry , Female , Fetus , Gene Expression , Hippocampus/chemistry , Hippocampus/metabolism , Humans , Hypothalamus/chemistry , Hypothalamus/metabolism , Immunohistochemistry , Male , Mesencephalon/chemistry , Middle Aged , Neurons/chemistry , Oligodendroglia/chemistry , Organ Specificity , Peptides/metabolism , Pituitary Gland/chemistry , Pituitary Gland/metabolism , Pituitary Gland, Posterior/chemistry , Pituitary Gland, Posterior/metabolism , Trefoil Factor-3 , White Matter/chemistry , White Matter/metabolism
4.
Chin J Integr Med ; 19(1): 29-35, 2013 Jan.
Article in English | MEDLINE | ID: mdl-21717156

ABSTRACT

OBJECTIVE: To investigate the synergistic anti-inflammatory effect of Radix Platycodon in combination with herbs for cleaning-heat and detoxification and its mechanism for Fel-targeting. METHODS: Forty Wistar rats were randomly divided into five groups (8 per group): the sham-operated group, model group, Radix Platycodon group, Flos Lonicera and Fructus Forsythia (LF) group, and Radix Platycodon, Flos Lonicera and Fructus Forsythia combination (PLF) group, using a random number table. A rat chronic obstructive pulmonary disease (COPD) model was established by passive smoking and intratracheal instillation of lipopolysaccharide (LPS). The treatments started from the 15th day of passive smoking for a total duration of 14 days. At the end of the treatment, changes in the following measurements were determined: lung histopathology, inflammatory cytokines including tumor necrosis factor α (TNF-α), transforming growth factor ß (TGF-ß) and interleukin IL-1ß (IL-1ß) in bronchoalveolar lavage fluid (BALF), and mRNA expression of endogenous active substance intestinal trefoil factor 3 (TFF3) in the lung tissue. RESULTS: Light microscopy showed that compared with the sham-operated group, rats in the COPD model group had disrupted alveolar structure, collapsed local alveoli, significantly widened or even fused alveolar septa, and massive infiltration of inflammatory cells in the alveolar wall and interstitium. In addition, significant bronchial epithelium hyperplasia, partially shed epithelia, and marked inflammatory cell infiltration in the bronchial wall and its surrounding tissues were noticed. Electron microscopy showed that rats in the model group had degeneration of alveolar type II epithelial cell; reduction, breakage or even loss of cell surface microvilli; swollen mitochondria with disappearing cristae and vacuole-like structure; and, increased secondary lysosomes in alveolar macrophages. The TNF-α, TGF-ß and IL-1ß levels and white blood cell (WBC) count in BALF were significantly increased (P < 0.01 or P < 0.05) and TFF3 mRNA expression in the lung tissue was significantly reduced (P < 0.01). After treatment, the pathological morphology of lung injury was less severe in all three treatment groups. In addition, TGF-ß and IL-1ß and WBC count in BALF were decreased (P < 0.01 or P < 0.05), and TFF3 mRNA expression in the lung tissue was significantly increased in the PLF group (P < 0.01). Compared with the LF group, the IL-1ß in BALF was significantly decreased P < 0.05), and TFF3 mRNA expression was significantly increased (P < 0.05) in the PLF group. CONCLUSIONS: Radix Platycodon synergizes with herbs for cleaning-heat and detoxification in reducing inflammatory injury in a rat model of COPD. The synergistic anti-inflammatory effect is reflected in the improvement in pathological changes and in the reduction of IL-1ß levels in BALF. The mechanism of such synergistic action may be related to its effect on maintaining the TFF3 mRNA expression and Fel-targeting function.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Neuropeptides/metabolism , Phytotherapy/methods , Plant Preparations/therapeutic use , Platycodon , Pulmonary Disease, Chronic Obstructive/drug therapy , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Immunohistochemistry , Lung/drug effects , Lung/pathology , Male , Microscopy, Electron , Neuropeptides/genetics , Polymerase Chain Reaction/methods , Pulmonary Disease, Chronic Obstructive/pathology , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Wistar , Sensitivity and Specificity , Trefoil Factor-3
5.
Article in English | WPRIM | ID: wpr-328480

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the synergistic anti-inflammatory effect of Radix Platycodon in combination with herbs for cleaning-heat and detoxification and its mechanism for Fel-targeting.</p><p><b>METHODS</b>Forty Wistar rats were randomly divided into five groups (8 per group): the sham-operated group, model group, Radix Platycodon group, Flos Lonicera and Fructus Forsythia (LF) group, and Radix Platycodon, Flos Lonicera and Fructus Forsythia combination (PLF) group, using a random number table. A rat chronic obstructive pulmonary disease (COPD) model was established by passive smoking and intratracheal instillation of lipopolysaccharide (LPS). The treatments started from the 15th day of passive smoking for a total duration of 14 days. At the end of the treatment, changes in the following measurements were determined: lung histopathology, inflammatory cytokines including tumor necrosis factor α (TNF-α), transforming growth factor β (TGF-β) and interleukin IL-1β (IL-1β) in bronchoalveolar lavage fluid (BALF), and mRNA expression of endogenous active substance intestinal trefoil factor 3 (TFF3) in the lung tissue.</p><p><b>RESULTS</b>Light microscopy showed that compared with the sham-operated group, rats in the COPD model group had disrupted alveolar structure, collapsed local alveoli, significantly widened or even fused alveolar septa, and massive infiltration of inflammatory cells in the alveolar wall and interstitium. In addition, significant bronchial epithelium hyperplasia, partially shed epithelia, and marked inflammatory cell infiltration in the bronchial wall and its surrounding tissues were noticed. Electron microscopy showed that rats in the model group had degeneration of alveolar type II epithelial cell; reduction, breakage or even loss of cell surface microvilli; swollen mitochondria with disappearing cristae and vacuole-like structure; and, increased secondary lysosomes in alveolar macrophages. The TNF-α, TGF-β and IL-1β levels and white blood cell (WBC) count in BALF were significantly increased (P < 0.01 or P < 0.05) and TFF3 mRNA expression in the lung tissue was significantly reduced (P < 0.01). After treatment, the pathological morphology of lung injury was less severe in all three treatment groups. In addition, TGF-β and IL-1β and WBC count in BALF were decreased (P < 0.01 or P < 0.05), and TFF3 mRNA expression in the lung tissue was significantly increased in the PLF group (P < 0.01). Compared with the LF group, the IL-1β in BALF was significantly decreased P < 0.05), and TFF3 mRNA expression was significantly increased (P < 0.05) in the PLF group.</p><p><b>CONCLUSIONS</b>Radix Platycodon synergizes with herbs for cleaning-heat and detoxification in reducing inflammatory injury in a rat model of COPD. The synergistic anti-inflammatory effect is reflected in the improvement in pathological changes and in the reduction of IL-1β levels in BALF. The mechanism of such synergistic action may be related to its effect on maintaining the TFF3 mRNA expression and Fel-targeting function.</p>


Subject(s)
Animals , Male , Rats , Bronchoalveolar Lavage Fluid , Chemistry , Cell Biology , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Drugs, Chinese Herbal , Therapeutic Uses , Immunohistochemistry , Lung , Pathology , Microscopy, Electron , Neuropeptides , Genetics , Metabolism , Phytotherapy , Methods , Plant Preparations , Therapeutic Uses , Platycodon , Polymerase Chain Reaction , Methods , Pulmonary Disease, Chronic Obstructive , Drug Therapy , Pathology , RNA, Messenger , Metabolism , Random Allocation , Rats, Wistar , Sensitivity and Specificity , Trefoil Factor-3
6.
J Nutr ; 142(12): 2135-40, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23077197

ABSTRACT

A central event in inflammatory bowel disease is the disruption of the mucosal homeostasis. Trefoil peptides [(TFF)] are emerging as key mediators in the defense and repair of the gastrointestinal mucosa. Here, we demonstrate induction of TFF by CLA with therapeutic antiinflammatory effects in a mouse model of inflammatory bowel disease. SW480 cells were treated with linoleic acid or CLA (0-2.5 µmol/L) in the absence or presence of the PPARγ inhibitor GW9662. Cells treated with CLA showed an upregulation of the intestinal trefoil factor, which was prevented by pretreatment with GW9662. Dextran sulfate sodium (2%) was used to induce colitis in mice and they were simultaneously fed with a standard or a CLA-supplemented (100 mg · kg(-1) · d(-1)) diet for 7 d. The CLA-enriched diet prevented the colon shortening induced by DSS and markedly reduced the disease activity index and the colonic expression of inducible NO synthase and NF-κB. Immunohistochemistry revealed an increase in PPARγ and TFF3 expression after CLA administration. Altogether, these results indicate that dietary CLA protects against DSS-induced colitis in a process involving induction of PPARγ and TFF3.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Colitis/drug therapy , Inflammatory Bowel Diseases/drug therapy , Linoleic Acids, Conjugated/administration & dosage , Mucins/physiology , PPAR gamma/physiology , Animals , Dextran Sulfate , Female , Heme Oxygenase-1/physiology , Linoleic Acids, Conjugated/pharmacology , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Mucins/analysis , PPAR gamma/analysis , Trefoil Factor-3
7.
Am J Physiol Gastrointest Liver Physiol ; 303(6): G744-51, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22821948

ABSTRACT

Pomegranate seed oil (PSO), which is the major source of conjugated linolenic acids such as punicic acid (PuA), exhibits strong anti-inflammatory properties. Necrotizing enterocolitis (NEC) is a devastating disease associated with severe and excessive intestinal inflammation. The aim of this study was to evaluate the effects of orally administered PSO on the development of NEC, intestinal epithelial proliferation, and cytokine regulation in a rat model of NEC. Premature rats were divided into three groups: dam fed (DF), formula-fed rats (FF), or rats fed with formula supplemented with 1.5% of PSO (FF + PSO). All groups were exposed to asphyxia/cold stress to induce NEC. Intestinal injury, epithelial cell proliferation, cytokine production, and trefoil factor 3 (Tff3) production were evaluated in the terminal ileum. Oral administration of PSO (FF+PSO) decreased the incidence of NEC from 61 to 26%. Feeding formula with PSO improved enterocyte proliferation in the site of injury. Increased levels of proinflammatory IL-6, IL-8, IL-12, IL-23, and TNF-α in the ileum of FF rats were normalized in PSO-treated animals. Tff3 production in the FF rats was reduced compared with DF but not further affected by the PSO. In conclusion, administration of PSO protects against NEC in the neonatal rat model. This protective effect is associated with an improvement of intestinal epithelial homeostasis and a strong anti-inflammatory effect of PSO on the developing intestinal mucosa.


Subject(s)
Enterocolitis, Necrotizing/drug therapy , Lythraceae/chemistry , Plant Oils/pharmacology , Seeds/chemistry , Animals , Animals, Newborn , Diet , Enterocolitis, Necrotizing/pathology , Female , Gene Expression Regulation , Ileum/pathology , Lipids/chemistry , Mucin-2/genetics , Mucin-2/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Plant Oils/chemistry , Pregnancy , RNA/genetics , RNA/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Trefoil Factor-3
8.
Int J Radiat Oncol Biol Phys ; 84(5): 1151-8, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22516806

ABSTRACT

PURPOSE: Management of locally advanced rectal cancer (RC) consists of neoadjuvant chemoradiotherapy (CRT) with fluoropyrimidines, followed by total mesorectal excision. We sought to evaluate the expression of selected genes, some of which were derived from a previous undirected SAGE (serial analysis of gene expression)-based approach, before and after CRT, to identify mechanisms of resistance. METHODS: This retrospective cohort study included 129 consecutive patients. Quantitative polymerase chain reaction of 53 candidate genes was performed on the biopsy specimen before treatment and on the surgical specimen after CRT. A paired-samples t test was performed to determine genes that were significantly changed after CRT. The result was correlated with patients' disease-free survival. RESULTS: Twenty-two genes were significantly upregulated, and two were significantly downregulated. Several of the upregulated genes have roles in cell cycle control; these include CCNB1IP1, RCC1, EEF2, CDKN1, TFF3, and BCL2. The upregulation of TFF3 was associated with worse disease-free survival on multivariate analyses (hazard ratio, 2.64; P=.027). Patients whose surgical specimens immunohistochemically showed secretion of TFF3 into the lumen of the tumoral microglands had a higher risk of relapse (hazard ratio, 2.51; P=.014). In vitro experiments showed that DLD-1 cells stably transfected with TFF3 were significantly less sensitive to 5-fluorouracil and showed upregulation of genes involved in the transcriptional machinery and in resistance to apoptosis. CONCLUSION: Upregulation of TFF3 after CRT for RC is associated with a higher risk of relapse. The physiological role of TFF3 in restoring the mucosa during CRT could be interfering with treatment efficacy. Our results could reveal not only a novel RC prognostic marker but also a therapeutic target.


Subject(s)
Adenocarcinoma/metabolism , Chemoradiotherapy, Adjuvant , Neoplasm Proteins/metabolism , Neoplasm Recurrence, Local , Peptides/metabolism , Rectal Neoplasms/metabolism , Rectal Neoplasms/therapy , Adenocarcinoma/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Chemoradiotherapy, Adjuvant/methods , Disease-Free Survival , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling/methods , Humans , Male , Middle Aged , Multivariate Analysis , Neoplasm Proteins/genetics , Peptides/genetics , Polymerase Chain Reaction , Prognosis , Protein Array Analysis/methods , Rectal Neoplasms/genetics , Retrospective Studies , Transfection/methods , Trefoil Factor-3 , Up-Regulation , Young Adult
9.
Eur J Gastroenterol Hepatol ; 22(9): 1134-40, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20461009

ABSTRACT

INTRODUCTION: The colonic mucus layer plays an important role in the protection of the intestinal epithelium and mainly consists of mucin glycoproteins (primarily MUC2 in the colon) trefoil factor 3 (TFF3) and secretory IgA. Butyrate is a major end product of fermentation of dietary fibres and is associated with beneficial effects on colonic health. Earlier in-vitro and animal studies showed that butyrate modulates MUC2 and TFF3 expression and mucin secretion, although data from human studies are not yet available. METHODS: Sixteen healthy volunteers and 35 ulcerative colitis (UC) patients in clinical remission self-administered a 60 ml rectal enema containing 100 mmol/l butyrate or placebo once daily for 2 and 3 weeks, respectively. After each treatment, biopsies were taken from the distal sigmoid for quantitative RT-PCR and immunohistochemical analysis of MUC2 and TFF3. In addition, mucosal sections were stained with high iron diamine-alcian blue to distinguish between sialomucins and sulphomucins. To analyse total mucin secretion and secretory IgA concentrations, 24 h faeces were collected during the day before the endoscopic examination. RESULTS: The butyrate intervention did not significantly modulate the expression of MUC2 (fold change: 1.04 and 1.05 in healthy volunteers and ulcerative colitis patients, respectively) or TFF3 (fold change: 0.91 and 0.94 in healthy volunteers and UC patients, respectively). Furthermore, the percentage of sialomucins, mucus secretion and secretory IgA concentrations were not affected by the butyrate intervention in both the groups. CONCLUSION: Butyrate exposure in healthy volunteers and UC patients in remission did not affect the measured parameters of the colonic mucus layer.


Subject(s)
Butyrates/administration & dosage , Colitis, Ulcerative/drug therapy , Colon/drug effects , Enema/methods , Mucin-2/genetics , Peptides/genetics , Adolescent , Adult , Aged , Colitis, Ulcerative/physiopathology , Colon/physiology , Feces/chemistry , Gene Expression/drug effects , Humans , Immunoglobulin A/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/physiology , Middle Aged , Mucin-2/metabolism , Peptides/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sialomucins/metabolism , Trefoil Factor-3 , Young Adult
10.
Aliment Pharmacol Ther ; 21(11): 1357-64, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15932366

ABSTRACT

BACKGROUND: Current treatment of ulcerative colitis is imperfect. Trefoil peptides are known to stimulate repair in many models of injury, including animal models of colitis. AIM: To assess the efficacy of trefoil factor family-3 enema treatment in a clinical trial. METHODS: A total of 16 patients with mild-to-moderate left sided ulcerative colitis were recruited into a double-blind randomized placebo-controlled study. Patients taking steroids or with proctitis only were excluded. Patients received 75 mL enemas containing either human recombinant trefoil factor family-3 (10 mg/mL) or saline alone once a day for 14 days. All patients also received an oral dose-increment of 1.2 g of mesalazine daily above their normal usage. Patients were assessed at 0, 2, 4 and 12 weeks. Remission was defined as Ulcerative Colitis Disease Activity Index of 0 or 1 with no blood in stool. Individual clinical improvement was defined as a Ulcerative Colitis Disease Activity Index reduction of >3. Data was analysed using chi-square test and anova. RESULTS: Median Ulcerative Colitis Disease Activity Index at entry were 8.5 (trefoil factor family-3 group) and 8 (placebo group). Analysed on an intention-to-treat basis, only one patient went into remission (in trefoil factor family-3 group at day 28). Clinical improvement was seen in two trefoil factor family-3 and three placebo patients on day 14 and two patients in each group on day 28. CONCLUSION: Increasing the dose of 5-aminosalicylic acid was moderately effective in reducing the Ulcerative Colitis Disease Activity Index but was insufficient to induce remission. Trefoil factor family-3 enemas were well-tolerated but did not provide additional benefit above that of adding additional 5-aminosalicylic acid alone.


Subject(s)
Colitis, Ulcerative/drug therapy , Mesalamine/administration & dosage , Mucins/therapeutic use , Muscle Proteins/therapeutic use , Administration, Oral , Adult , Double-Blind Method , Drug Therapy, Combination , Enema/methods , Female , Humans , Male , Middle Aged , Patient Compliance , Peptides , Treatment Outcome , Trefoil Factor-3
11.
Peptides ; 25(5): 833-8, 2004 May.
Article in English | MEDLINE | ID: mdl-15177879

ABSTRACT

TFF3 is synthesized in magnocellular oxytocin neurons of the supraoptic (SON) and paraventricular nuclei (PVN) of the rat and human hypothalamus. Here we investigated whether intracerebroventricular (i.c.v.) injection of TFF3 stimulates oxytocin release into the blood and activates Fos protein immunoreactivity in oxytocin neurons of the SON and PVN in rats. The results show that plasma oxytocin concentrations were not altered after i.c.v. injection of TFF3 or vehicle. Fos protein expression was significantly increased in both the SON and PVN after TFF3 injections and double labeling studies showed that the Fos signal was predominantly in oxytocin neurons.


Subject(s)
Hypothalamus/metabolism , Neuropeptides/metabolism , Neuropeptides/pharmacology , Oxytocin/metabolism , Supraoptic Nucleus/metabolism , Animals , Female , Genes, fos/genetics , Hypothalamus/pathology , Neurons/metabolism , Oxytocin/blood , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/pathology , Rats , Rats, Sprague-Dawley , Supraoptic Nucleus/pathology , Trefoil Factor-3
12.
FASEB J ; 15(9): 1517-28, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11427483

ABSTRACT

We have investigated the possible functional relationships between cellular invasion pathways induced by trefoil factors (TFFs), src, and the cyclooxygenases COX-1 and COX-2. Pharmacological inhibitors of the Rho small GTPase (C3 exoenzyme), phospholipase C (U-73122), cyclooxygenases (SC-560, NS-398), and the thromboxane A2 receptor (TXA2-R) antagonist SQ-295 completely abolished invasion induced by intestinal trefoil factor, pS2, and src in kidney and colonic epithelial cells MDCKts.src and PCmsrc. In contrast, invasion was induced by the TXA2-R mimetic U-46619, constitutively activated forms of the heterotrimeric G-proteins Galphaq (AGalphaq), Galpha12, Galpha13 (AGalpha12/13), which are signaling elements downstream of TXA2-R. Ectopic overexpression of pS2 cDNA and protein in MDCKts.src-pS2 cells and human colorectal cancer cells HCT8/S11-pS2 initiate distinct invasion signals that are Rho independent and COX and TXA2-R dependent. We detected a marked induction of COX-2 protein and accumulation of the stable PGH2/TXA2 metabolite TXB2 in the conditioned medium from cells transformed by src. This led to activation of the TXA2-R-dependent invasion pathway, which is monitored via a Rho- and Galpha12/Galpha13-independent mechanism using the Galphaq/PKC signaling cascade. These findings identify a new intracrine/paracrine loop that can be monitored by TFFs and src in inflammatory diseases and progression of colorectal cancers.


Subject(s)
Growth Substances/pharmacology , Mucins , Muscle Proteins , Neuropeptides , Peptides/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Proto-Oncogene Proteins pp60(c-src)/pharmacology , Receptors, Thromboxane/metabolism , Cell Line, Transformed , Cells, Cultured , Culture Media, Conditioned , Cyclooxygenase 1 , Cyclooxygenase 2 , DNA, Complementary/genetics , DNA-Binding Proteins/metabolism , GTP Phosphohydrolases/metabolism , GTP-Binding Protein alpha Subunits, G12-G13 , GTP-Binding Protein alpha Subunits, Gq-G11 , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/enzymology , Isoenzymes/metabolism , Kidney/cytology , Kidney/enzymology , Membrane Proteins , Neoplasm Invasiveness , Proteins/genetics , Proteins/pharmacology , Signal Transduction , Transfection , Trefoil Factor-1 , Trefoil Factor-2 , Trefoil Factor-3 , Tumor Cells, Cultured , Tumor Suppressor Proteins , Type C Phospholipases/metabolism
13.
Gastroenterology ; 118(1): 70-80, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10611155

ABSTRACT

BACKGROUND & AIMS: Trefoil factors (TFFs) are secreted gastrointestinal proteins that have been shown to protect and promote healing of the gastrointestinal tract. Moreover, pS2/TFF1 is essential for normal differentiation of the gastric mucosa because deficient mice develop antropyloric adenomas. To date, it is unclear how TFFs mediate their functions. METHODS: Using the yeast 2-hybrid system, we attempted to identify murine TFF1 interacting proteins by screening a stomach and duodenum complementary DNA (cDNA) expression library. RESULTS: Four positive clones were isolated. Sequence and expression studies showed that they corresponded to the murine counterpart of human cDNA sequences encoding carboxy-terminal fragments of mMuc2 (489 residues) and mMuc5AC (427, 430, and 894 residues) mucin proteins. Mutagenesis experiments showed that TFF1 interacts with the 2 mucins through binding with their VWFC1 and VWFC2 (von Willebrand factor C) cysteine-rich domains. CONCLUSIONS: These results show that the gastrointestinal protective effect of TFF1, and presumably of the other TFFs, is caused at least partially by their participation, via mucin binding, in the correct organization of the mucous layer that protects the apical side of the mucosa from deleterious luminal agents.


Subject(s)
Duodenum/metabolism , Gastric Mucosa/metabolism , Growth Substances/metabolism , Mucins/metabolism , Muscle Proteins , Neuropeptides , Peptides/metabolism , von Willebrand Factor/metabolism , Amino Acid Sequence , Animals , Base Sequence , Clone Cells , Cysteine , Hybrid Cells , Mice , Molecular Sequence Data , Mucins/chemistry , Mucins/genetics , Protein Structure, Tertiary , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Trefoil Factor-2 , Trefoil Factor-3 , Yeasts , von Willebrand Factor/genetics
14.
FASEB J ; 10(13): 1518-23, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8940297

ABSTRACT

Human intestinal trefoil factor, hITF, a secretory polypeptide found mainly in the human gastrointestinal tract, is a member of the newly characterized trefoil factor or P-domain peptide family representing putative growth factors. Here we describe the identification of this gut peptide in the human brain and pituitary. With reverse transcriptase polymerase chain reaction, we were able to isolate and clone the transcript from human hypothalamus. An antibody generated against a synthetic peptide derived from the carboxyl terminus of hITF was used for immunohistochemical studies of appropriate tissue sections. Neurons expressing hITF were identified in two magnocellular hypothalamic nuclei, the paraventricular and periventricular nuclei. hITF polypeptide was also observed in Herring bodies of the neurohypophysis and in secretory cells of the adenohypophysis. Double immunostaining with antigrowth hormone antibody showed partial coexistence in a selected subpopulation of adenohypophysial cells. Localization of hITF in the hypothalamo-neurohypophysial system may suggest a modulatory action on the classical magnocellular nonapeptides vasopressin and oxytocin, and further indicates an adenohypophysial importance of this peptide. It is likely that hITF represents a novel neuropeptide of yet unknown function.


Subject(s)
Growth Substances/biosynthesis , Hypothalamus/metabolism , Mucins , Muscle Proteins , Neuropeptides , Peptide Biosynthesis , Pituitary Gland/metabolism , Adult , Blotting, Western , Cloning, Molecular , Growth Substances/genetics , Humans , Immunoenzyme Techniques , Intestinal Mucosa/metabolism , Male , Peptides/genetics , Polymerase Chain Reaction , Trefoil Factor-2 , Trefoil Factor-3
15.
Eur J Hum Genet ; 4(6): 308-15, 1996.
Article in English | MEDLINE | ID: mdl-9043862

ABSTRACT

Trefoil peptides are small secretory proteins characterized by three intrachain disulfide bonds forming the trefoil motif or P-domain. They are abundantly expressed on mucosal surfaces, especially of the gastrointestinal tract. In pathological conditions such as ulcers, metaplasia and neoplasia, their expression is upregulated. Three human trefoil peptides have been described: the estrogen-inducible pS2 protein, the spasmolytic protein and the intestinal trefoil factor. Recently, their role in the maintenance of surface integrity and ulcer healing was discussed. We already mapped the corresponding three genes (BCEI), SML1, TFF3) to the same genomic region (21q22.3). In this paper, we show that the three genes are clustered in a tandemly orientated fashion within 50 kb on a bacterial artificial chromosome (BAC) recombinant. This cluster is located adjacent to D21S19 and the locus order is cen-D21S212-TFF3-SML1-BCEI-D21S19-tel, whereas transcription of all three genes is directed towards the centromere. The gene structure of SML1 exhibits four exons, two of which encode the two separate trefoil motifs. TFF3 and BCEI, both containing one trefoil motif, are composed of three exons each, suggesting gene duplication and exon-shuffling events during evolution. The 5'-flanking region of SML1 was compared to the corresponding region of other trefoil genes. Two motifs with identical sequence and positions are shared between SML1 and BCEI, thus presenting possible targets for stomach-specific gene regulation. Two other motifs are shared within all known human and rat trefoil genes, suggesting a coordinated regulation and/or a common locus-controlling region. Using RT-PCR, a change in the pattern of trefoil gene expression is detected in tissue samples from normal gastric mucosa, hyperplastic polyps, gastric cancer, and gastric cancer cell lines, respectively.


Subject(s)
Chromosome Mapping , Chromosomes, Human, Pair 21 , Gene Expression Regulation , Growth Substances/genetics , Mucins , Muscle Proteins , Neuropeptides , Peptides/genetics , Proteins , Amino Acid Sequence , Base Sequence , Codon, Initiator , Cysteine , DNA, Complementary , Exons , Gastric Mucosa/metabolism , Humans , Intercellular Signaling Peptides and Proteins , Introns , Molecular Sequence Data , Neoplasm Proteins/genetics , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Trefoil Factor-1 , Trefoil Factor-2 , Trefoil Factor-3 , Tumor Suppressor Proteins
16.
Brain Res Mol Brain Res ; 33(2): 269-76, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8750886

ABSTRACT

P-domain peptides, a new family of secretory polypeptides, have been identified mainly in the gastroenteropancreatic tract of humans, rodents, and amphibians as well as in amphibian skin. In the present study, with PCR and RNA analysis a transcript has been discovered in rat brain termed rP1.B. The deduced polypeptide consists of a single P-domain and its amino acid sequence matches that of rat intestinal trefoil factor (rITF). Thus far, rP1.B is the only P-domain peptide expressed in neuronal cells of the CNS. Immunostained magnocellular perikarya were visible in the paraventricular, supraoptic and periventricular nuclei. Parvocellular rP1.B neurons were found in the arcuate nucleus. Additionally, specific hybridization signals with radiolabeled transcripts were observed in the same regions. rP1.B in the rat hypothalamus may be involved in the control of hypothalamo-hypophysial functions.


Subject(s)
Hypothalamus/metabolism , Neuropeptides/biosynthesis , Amino Acid Sequence , Amphibians , Animals , Base Sequence , DNA Primers , Gene Expression , Humans , Hypothalamo-Hypophyseal System/physiology , Immunohistochemistry , In Situ Hybridization , Molecular Sequence Data , Neuropeptides/analysis , Organ Specificity , Paraventricular Hypothalamic Nucleus/metabolism , Polymerase Chain Reaction , Rats , Rats, Wistar , Sequence Homology, Nucleic Acid , Supraoptic Nucleus/metabolism , Transcription, Genetic , Trefoil Factor-3
SELECTION OF CITATIONS
SEARCH DETAIL