Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Oncogene ; 27(16): 2276-88, 2008 Apr 07.
Article in English | MEDLINE | ID: mdl-18391970

ABSTRACT

The FoxO family of Forkhead transcription factors plays an important role in longevity and tumor suppression by upregulating target genes involved in stress resistance, metabolism, cell cycle arrest and apoptosis. FoxO transcription factors translate a variety of environmental stimuli, including insulin, growth factors, nutrients and oxidative stress, into specific gene-expression programs. These environmental stimuli control FoxO activity primarily by regulating their subcellular localization, but also by affecting their protein levels, DNA-binding properties and transcriptional activity. The precise regulation of FoxO transcription factors is enacted by an intricate combination of post-translational modifications (PTMs), including phosphorylation, acetylation and ubiquitination, and binding protein partners. An intriguing possibility is that FoxO PTMs may act as a 'molecular FoxO code' read by selective protein partners to rapidly regulate gene-expression programs. The effective control of FoxO activity in response to environmental stimuli is likely to be critical to prevent aging and age-dependent diseases, including cancer, neurodegenerative diseases and diabetes.


Subject(s)
Forkhead Transcription Factors/physiology , Animals , DNA/metabolism , Forkhead Transcription Factors/metabolism , Humans , Protein Binding , Protein Transport , Subcellular Fractions/metabolism
2.
Clin Sci (Lond) ; 100(6): 627-33, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11352778

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) are effective analgesics but cause gastrointestinal injury. Present prophylactic measures are suboptimal and novel therapies are required. Bovine colostrum is a cheap, readily available source of growth factors, which reduces gastrointestinal injury in rats and mice. We therefore examined whether spray-dried, defatted colostrum could reduce the rise in gut permeability (a non-invasive marker of intestinal injury) caused by NSAIDs in volunteers and patients taking NSAIDs for clinical reasons. Healthy male volunteers (n=7) participated in a randomized crossover trial comparing changes in gut permeability (lactulose/rhamnose ratios) before and after 5 days of 50 mg of indomethacin three times daily (tds) per oral with colostrum (125 ml, tds) or whey protein (control) co-administration. A second study examined the effect of colostral and control solutions (125 ml, tds for 7 days) on gut permeability in patients (n=15) taking a substantial, regular dose of an NSAID for clinical reasons. For both studies, there was a 2 week washout period between treatment arms. In volunteers, indomethacin caused a 3-fold increase in gut permeability in the control arm (lactulose/rhamnose ratio 0.36+/-0.07 prior to indomethacin and 1.17+/-0.25 on day 5, P<0.01), whereas no significant increase in permeability was seen when colostrum was co-administered. In patients taking long-term NSAID treatment, initial permeability ratios were low (0.13+/-0.02), despite continuing on the drug, and permeability was not influenced by co-administration of test solutions. These studies provide preliminary evidence that bovine colostrum, which is already currently available as an over-the-counter preparation, may provide a novel approach to the prevention of NSAID-induced gastrointestinal damage in humans.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Colostrum , Dietary Supplements , Intestinal Diseases/prevention & control , Adult , Aged , Animals , Cattle , Cross-Over Studies , Double-Blind Method , Female , Humans , Indomethacin/adverse effects , Intestinal Absorption/drug effects , Intestinal Diseases/chemically induced , Intestinal Mucosa/metabolism , Male , Middle Aged , Permeability/drug effects
3.
Gut ; 47(5): 622-7, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11034576

ABSTRACT

INTRODUCTION: Epidermal growth factor (EGF) is normally present as EGF(1-53). A variety of C terminal truncated forms have been used in preliminary trials for treating gastrointestinal injury but their relative potency and stability when used in a clinical setting are unclear. Therefore, we compared the biological activity of recombinant EGF(1-53), EGF(1-52), EGF(1-51), and the C terminal peptides EGF(44-53) and EGF(49-53). METHODS: Purity of forms was confirmed by mass spectrometry. Bioactivity of the different EGF forms was determined using [methyl-(3)H] thymidine incorporation into primary rat hepatocytes and their ability to reduce indomethacin (20 mg/kg subcutaneously)/restraint induced gastric injury in rats. Stability of EGF peptides was determined by serial sampling from a syringe driver system containing EGF/4% albumin in saline. RESULTS: Biological activity assays of EGF(1-53), EGF(1-52), and EGF(1-51) gave almost identical thymidine uptake dose-response curves (maximal responses increasing baseline uptake from 4400 (600) cpm (mean (SEM)) to about 22 000 (2000) cpm when EGF was added at 1. 6 nM). EGF(44-53) and EGF(49-53) did not stimulate (3)H thymidine uptake. Control rats had 47 (4) mm(2) damage/stomach, EGF(1-51), EGF(1-52), and EGF(1-53) at 0.16 and 0.80 nmol/kg/h each reduced gastric injury by about 50% and 80%, respectively (both doses p<0.01 compared with control but no significant difference between the different forms). EGF was stable at room temperature for seven days but biological activity decreased by 35% and 40% at two and three weeks, respectively (both p<0.01). Exposure to light did not affect bioactivity. CONCLUSION: EGF(1-51) and EGF(1-52) are as biologically active as full length EGF(1-53) but the C terminal penta- and decapeptides are ineffective. Clinical trials of EGF can probably use infusion systems for at least 48 hours at room temperature and with exposure to light, without reducing biological efficacy.


Subject(s)
Epidermal Growth Factor/chemistry , Peptide Fragments/chemistry , Recombinant Proteins/chemistry , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Drug Stability , Epidermal Growth Factor/therapeutic use , Gastric Mucosa/drug effects , Gastric Mucosa/injuries , Humans , Male , Mass Spectrometry , Peptide Fragments/therapeutic use , Rats , Rats, Sprague-Dawley , Recombinant Proteins/therapeutic use , Stomach Diseases/chemically induced , Stomach Diseases/drug therapy , Stomach Diseases/pathology , Thymidine/pharmacokinetics
4.
Schweiz Med Wochenschr ; Suppl 125: 92S-95S, 2000.
Article in German | MEDLINE | ID: mdl-11141953

ABSTRACT

Nasal polyposis is an inflammatory condition of the nose and the sinuses characterised by a marked infiltration of eosinophils in addition to lymphocytes, mast cells and macrophages. The selective recruitment of eosinophils to inflammatory sites is mediated by CC chemokines such as Eotaxin and Eotaxin-2. In the present study histology, immunohistochemistry and ELISA were performed. The levels of Eotaxin and Eotaxin-2 and for comparison other chemokines RANTES and IL-8 were measured in nasal polyp tissue and in control nasal tissue. On histological examination 6 polyps showed an oedematous structure, one was glandular and one had a fibromatous pattern, while all showed a marked eosinophil infiltration. Immunohistochemistry of the polyps showed that epithelial cells were strongly positive for Eotaxin and IL-8, whereas endothelial cells stained positive for Eotaxin-2. Significantly higher amounts of Eotaxin, Eotaxin-2 and IL-8 were detected in polyp tissue when compared with control middle turbinates. The increased levels of eosinophil-stimulating chemokines, such as Eotaxin and Eotaxin-2 in nasal polyps suggest that they may be important regulators of eosinophil recruitment in this inflammatory disease.


Subject(s)
Chemokines/analysis , Nasal Polyps/pathology , Nasal Polyps/physiopathology , Chemokine CCL11 , Chemokine CCL24 , Chemokine CCL5/analysis , Chemokines/physiology , Chemokines, CC/analysis , Cytokines/analysis , Enzyme-Linked Immunosorbent Assay , Eosinophils/physiology , Humans , Immunohistochemistry , Interleukin-9/analysis , Nasal Mucosa/pathology
5.
J Pathol ; 188(3): 312-7, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10419601

ABSTRACT

TFF1 is a 60-amino acid peptide produced in normal gastric mucosa which forms dimers spontaneously. Tumours of patients with gastric cancer usually have reduced TFF1 levels and disruption of the TFF1 gene causes animals to develop gastric adenomas and carcinomas. The effect of normal sequence human recombinant TFF1 and an analogue (Cys(58)-->Ser(58)), which is unable to dimerize, on the proliferation and morphology of the human gastric adenocarcinoma cell line AGS was therefore investigated. Proliferation, assessed by total cell number and [methyl-(3)H]thymidine incorporation, was reduced by dimeric TFF1 in a dose-dependent manner. Monomeric TFF1 also reduced proliferation but was less potent than the dimeric form. It is concluded that TFF1 may be an important controller of gastric cell proliferation, that dimerization of TFF1 is important in this effect, and that the reduced levels of TFF1 seen in gastric cancer may be of clinical relevance.


Subject(s)
Adenocarcinoma/pathology , Growth Inhibitors/pharmacology , Proteins/pharmacology , Stomach Neoplasms/pathology , Cell Division/drug effects , DNA, Neoplasm/biosynthesis , Dose-Response Relationship, Drug , Female , Humans , Middle Aged , Recombinant Proteins/pharmacology , Trefoil Factor-1 , Tumor Cells, Cultured , Tumor Suppressor Proteins
6.
Gut ; 44(5): 653-8, 1999 May.
Article in English | MEDLINE | ID: mdl-10205201

ABSTRACT

BACKGROUND: Non-steroidal anti-inflammatory drugs (NSAIDs) are effective for arthritis but cause gastrointestinal injury. Bovine colostrum is a rich source of growth factors and is marketed as a health food supplement. AIMS: To examine whether spray dried, defatted colostrum or milk preparations could reduce gastrointestinal injury caused by indomethacin. METHODS: Effects of test solutions, administered orally, were examined using an indomethacin restraint rat model of gastric damage and an indomethacin mouse model of small intestinal injury. Effects on migration of the human colonic carcinoma cell line HT-29 and rat small intestinal cell line RIE-1 were assessed using a wounded monolayer assay system (used as an in vitro model of wound repair) and effects on proliferation determined using [3H]thymidine incorporation. RESULTS: Pretreatment with 0.5 or 1 ml colostral preparation reduced gastric injury by 30% and 60% respectively in rats. A milk preparation was much less efficacious. Recombinant transforming growth factor beta added at a dose similar to that found in the colostrum preparation (12.5 ng/rat), reduced injury by about 60%. Addition of colostrum to drinking water (10% vol/vol) prevented villus shortening in the mouse model of small intestinal injury. Addition of milk preparation was ineffective. Colostrum increased proliferation and cell migration of RIE-1 and HT-29 cells. These effects were mainly due to constituents of the colostrum with molecular weights greater than 30 kDa. CONCLUSIONS: Bovine colostrum could provide a novel, inexpensive approach for the prevention and treatment of the injurious effects of NSAIDs on the gut and may also be of value for the treatment of other ulcerative conditions of the bowel.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Colostrum , Dietary Supplements , Gastrointestinal Diseases/prevention & control , Animals , Cattle , Cell Culture Techniques , Cell Division/drug effects , Cell Movement/drug effects , Female , Gastrointestinal Diseases/chemically induced , Humans , Indomethacin/toxicity , Intestine, Small/drug effects , Intestine, Small/pathology , Mice , Milk , Pregnancy , Rats , Stomach Diseases/chemically induced , Stomach Diseases/prevention & control , Transforming Growth Factor beta/therapeutic use , Tumor Cells, Cultured , Wound Healing
7.
Gut ; 44(5): 709-14, 1999 May.
Article in English | MEDLINE | ID: mdl-10205210

ABSTRACT

BACKGROUND: Dietary lectins can alter the proliferation of colonic cells. Differentiation is regulated by adhesion molecules which, being glycosylated, are targets for lectin binding. AIMS: To examine the effects of dietary lectins on differentiation, adhesion, and proliferation of colorectal cancer cells. METHODS: Differentiation was assessed in three dimensional gels, adhesion by aggregation assay, and proliferation by 3H thymidine incorporation. The role of the epithelial cell adhesion molecule (epCAM) was studied using a specific monoclonal antibody in blocking studies and Western blots. The human colon cancer cell lines LS174T, SW1222, and HT29 were studied. RESULTS: The cell line LS174T differentiated in the presence of Vicia faba agglutinin (VFA) into gland like structures. This was inhibited by anti-epCAM monoclonal antibody. Expression of epCAM itself was unaffected. VFA as well as wheat germ agglutinin (WGA) and the edible mushroom lectin (Agaricus bisporus lectin, ABL) significantly aggregated LS174T cells but peanut agglutinin (PNA) and soybean agglutinin (SBA) did not. All lectins aggregated SW1222 and HT29 cells. Aggregation was blocked by the corresponding sugars. Aggregation of cells by VFA was also inhibited by anti-epCAM. VFA, ABL, and WGL inhibited proliferation of all the cell lines; PNA stimulated proliferation of HT29 and SW1222 cells. In competition studies all sugars blocked aggregation and proliferation of all cell lines, except that the addition of mannose alone inhibited proliferation. CONCLUSION: VFA stimulated an undifferentiated colon cancer cell line to differentiate into gland like structures. The adhesion molecule epCAM is involved in this. Dietary or therapeutic VFA may slow progression of colon cancer.


Subject(s)
Adenocarcinoma/pathology , Colorectal Neoplasms/pathology , Fabaceae/chemistry , Lectins/pharmacology , Plants, Medicinal , Antigens, Neoplasm/metabolism , Blotting, Western , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Differentiation/drug effects , Cell Division/drug effects , Epithelial Cell Adhesion Molecule , Humans , Plant Lectins , Tumor Cells, Cultured/drug effects
8.
J Pathol ; 185(2): 153-8, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9713341

ABSTRACT

Human pS2 (trefoil factor family 1, TFF1), a 60-amino acid member of the trefoil peptide family, forms dimers via Cys58 and may stimulate gut repair. The effects of dimeric pS2-TFF1 and monomeric pS2-TFF1 (Cys58 replaced by Ser58) were compared in models of wound healing. Rats given dimeric pS2-TFF1 at 25 and 50 micrograms/kg per h had 50 per cent and 70 per cent reduction in gastric damage induced respectively by indomethacin (20 mg/kg subcutaneously) and restraint (P < 0.01). Monomeric pS2-TFF1, at the same doses, was significantly less effective at reducing injury (about half the amount of protection, P < 0.01 vs. same doses of dimeric). The rate of migration of cells at the leading edge of wounded monolayers of the human colonic cell line HT29 was increased by addition of dimeric or monomeric forms of pS2-TFF1 (0.65-325 micrograms/ml). Dimeric pS2-TFF1 had a greater effect than the monomeric form at all doses tested (P < 0.05). Cell migration induced by pS2-TFF1 was blocked by a pS2-TFF1 antibody, but not by a transforming growth factor beta neutralizing antibody. pS2-TFF1 did not influence cell proliferation as assessed by thymidine incorporation. The increased biological effects of dimeric pS2-TFF1 might be due to direct interaction of Cys58 with a putative trefoil receptor or, more likely, dimerization of pS2-TFF1 might stabilize the interaction with its receptor. This may involve a bivalent interaction of residues on the surfaces of the two trefoil domains.


Subject(s)
Gastric Mucosa/injuries , Proteins/metabolism , Wound Healing , Analysis of Variance , Animals , Anti-Inflammatory Agents, Non-Steroidal , Cell Division/drug effects , Cell Movement/drug effects , Dimerization , Dose-Response Relationship, Drug , Gastric Mucosa/cytology , Gastric Mucosa/drug effects , HT29 Cells , Humans , In Vitro Techniques , Indomethacin , Male , Proteins/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/pharmacology , Trefoil Factor-1 , Tumor Suppressor Proteins , Wound Healing/drug effects
9.
Hepatology ; 26(1): 49-58, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9214451

ABSTRACT

Following experimental partial hepatectomy of 70% in the rat, there is a semisynchronized surge of hepatocyte proliferation that ceases after 48 to 72 hours. Little is known about the determinants governing the termination of the proliferative phase, although transforming growth factor (TGF) beta has been implicated as an important inhibitor of hepatocyte replication in this model. We previously reported an additional non-TGF-beta inhibitor in medium conditioned by nonparenchymal cells isolated from regenerating liver (CM-NPC-Reg) between 24 and 48 hours after partial hepatectomy, but it was not found in medium conditioned by nonparenchymal cells from unoperated control liver. CM-NPC-Reg suppressed replicative DNA synthesis of primary rat hepatocytes in response to hepatocyte growth factor (HGF), epidermal growth factor (EGF), or TGF-alpha as assessed by 3H-thymidine incorporation. We now present evidence that interleukin (IL)-1 is the major inhibitor of hepatocyte DNA synthesis present in CM-NPC-Reg. IL-1 receptor antagonist abrogated the inhibition, as did antibodies to rat IL-1alpha and -beta; a combination of both antibodies was required, implicating both IL-1alpha and IL-1beta as active constituents in CM-NPC-Reg. To investigate in vivo changes in IL-1 expression, we assessed expression of IL-1alpha messenger RNA (mRNA) in whole rat liver following partial hepatectomy; mRNA was down-regulated at 10 hours in the pre-replicative phase of liver regeneration and up-regulated at 24 hours and 48 hours when proliferation is waning. Rat hepatocytes isolated from liver 24 hours after partial hepatectomy showed increased sensitivity to the inhibitory action of IL-1. Exogenous IL-1beta, administered parenterally to a group of rats at 0 and 12 hours after partial hepatectomy significantly reduced the incorporation of the thymidine analogue, bromodeoxyuridine (BrdU), into hepatocytes at 18 hours. These data indicate that nonparenchymal cells isolated from regenerating rat liver elaborate IL-1, and support the hypothesis that IL-1 plays a role suppressing hepatocyte proliferation and terminating the surge of DNA synthesis induced after partial hepatectomy.


Subject(s)
Interleukin-1/metabolism , Interleukin-1/pharmacology , Liver Regeneration/physiology , Animals , Cell Division/drug effects , Cells, Cultured , Culture Media, Conditioned , DNA/biosynthesis , DNA/drug effects , Dose-Response Relationship, Drug , Epidermal Growth Factor/pharmacology , Insulin/pharmacology , Liver/metabolism , Liver Regeneration/drug effects , Male , RNA, Messenger/metabolism , Rats , Receptors, Interleukin-1/antagonists & inhibitors , Recombinant Proteins/pharmacology , Time Factors
10.
Hepatology ; 21(5): 1405-12, 1995 May.
Article in English | MEDLINE | ID: mdl-7537713

ABSTRACT

Histidinemia in mice and in humans is an autosomal recessive disorder of histidine metabolism that leads to high-histidine levels in both plasma and urine and is caused by a lack of hepatic histidine-alpha-deaminase (histidase). We have used a novel approach to hepatocellular transplantation to effect a complete phenotypic cure of histidinemia in a mouse model. Mice lacking histidase were treated with isolated liver cells (approximately 18 x 10(6) hepatocytes and 9 x 10(6) nonparenchymal cells) from histidase-competent donors transplanted into the peritoneum (active transplant group). Recipient mice showed a dramatic decrease, by more than 75%, in urinary histidine levels from day one throughout the course of the experiment, resulting in levels within the normal range for wild-type mice. In comparison, there was no change in urinary histidine levels in the control group of histidase-deficient mice treated with isolated liver cells from mice lacking histidase (statistical comparison between the two groups, P < .003, two-way ANOVA). Histologically, ectopic liver tissue was seen in the peritoneum in association with abdominal wall, pancreas, and peritoneal connective tissue; immunohistochemical evidence showed expression of histidase in the ectopic liver tissue in the active transplant group. This report is the first to show complete correction of a defective biochemical phenotype achieved by hepatocellular transplantation.


Subject(s)
Cell Transplantation , Histidine/blood , Liver/cytology , Animals , Histidine/deficiency , Histidine/urine , Histidine Ammonia-Lyase/metabolism , Hybridization, Genetic , Immunohistochemistry/methods , Liver/enzymology , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Staining and Labeling
11.
Gastroenterology ; 108(1): 92-101, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7806067

ABSTRACT

BACKGROUND/AIMS: Epidermal growth factor (EGF) is present in gastric juice and has potent mitogenic properties. The stability of EGF in gastric juice under various physiological and pathophysiological conditions was examined. METHODS: Recombinant human EGF1-53 was incubated with HCl containing pepsin. We also determined the forms of EGF present in the gastric juice of patients under basal conditions, patients taking the acid suppressant omeprazole, patients with achlorhydria, and volunteers undergoing intragastric neutralization with NaHCO3 (n = 6 per group). Samples were analyzed using mass spectroscopy and/or high-pressure liquid chromatography followed by radioimmunoassay. The effect of acid and pepsin digestion on EGF bioactivity was determined using an in vitro hepatocyte bioassay and an in vivo cytoprotection assay in the rat stomach. RESULTS: EGF1-53 was digested to the EGF1-49 and EGF1-46 forms in all samples containing pepsin when the pH was < 4. In gastric juice samples with pH > 4, the proportion of intact EGF increased to about 60%. For both methods of bioassay, intact EGF1-53 was about 3-4 times as potent as acid and pepsin-treated EGF. CONCLUSIONS: EGF is produced in the 1-53 form but is rapidly cleaved to smaller, less active forms in acidic gastric juice. In contrast, only a small proportion of the EGF is cleaved if the pH is maintained above 4. This mechanism may be relevant to the healing process of acid suppressants.


Subject(s)
Acids/metabolism , Digestion , Epidermal Growth Factor/metabolism , Gastric Juice/metabolism , Achlorhydria/metabolism , Animals , Bicarbonates/therapeutic use , Drug Stability , Female , Gastric Juice/chemistry , Humans , Indomethacin/pharmacology , Liver/cytology , Liver/metabolism , Male , Omeprazole/therapeutic use , Pepsin A/pharmacology , Rats , Rats, Wistar , Recombinant Proteins , Stomach/drug effects , Stomach/pathology , Thymidine/pharmacokinetics
12.
J Anat ; 185 ( Pt 3): 543-51, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7649790

ABSTRACT

This study reports the tissue distribution of Hepatocyte Growth Factor-Scatter Factor (HGF/SF) in human fetal tissue using Northern analysis and in situ hybridisation techniques. In tissue from fetuses of 9-17 wk gestational age, the 6 kb mRNA transcript for HGF/SF was demonstrated in many tissues but prominently in liver, intestine, gall bladder and spleen. In situ hybridisation demonstrated that HGF/SF expression was not confined to mesenchymal tissues, as suggested by previous studies but was expressed in epithelial tissues, particularly in small intestine, keratinising epithelium of tongue, skin and oesophagus. In the small intestine epithelial expression was strikingly regional, being confined to the crypt region, the site of enterocyte proliferation. Northern analysis of tissues for c-met mRNA, representing expression of the HGF/SF receptor, demonstrated receptor expression in all tissues studied except the thyroid gland.


Subject(s)
Blotting, Northern , Embryonic and Fetal Development/physiology , Hepatocyte Growth Factor/metabolism , In Situ Hybridization , Mesoderm/metabolism , Epithelium/chemistry , Epithelium/embryology , Epithelium/metabolism , Esophagus/chemistry , Esophagus/embryology , Gallbladder/chemistry , Gallbladder/embryology , Hepatocyte Growth Factor/analysis , Humans , Intestines/chemistry , Intestines/embryology , Liver/chemistry , Liver/embryology , Mesoderm/chemistry , Proto-Oncogene Proteins c-met , Receptor Protein-Tyrosine Kinases/metabolism , Skin/chemistry , Skin/embryology , Spleen/chemistry , Spleen/embryology , Tongue/chemistry , Tongue/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...