Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
1.
Biometals ; 32(2): 211-226, 2019 04.
Article in English | MEDLINE | ID: mdl-30806852

ABSTRACT

In patients with iron overload disorders, increasing number of reports of renal dysfunction and renal iron deposition support an association between increased iron exposure and renal injury. In systemic iron overload, elevated circulating levels of transferrin-bound (TBI) and non-transferrin-bound iron (NTBI) are filtered to the renal proximal tubules, where they may cause injury. However, the mechanisms of tubular iron handling remain elusive. To unravel molecular renal proximal tubular NTBI and TBI handling, human conditionally immortalized proximal tubular epithelial cells (ciPTECs) were incubated with 55Fe as NTBI and fluorescently labeled holo-transferrin as TBI. Ferrous iron importers ZIP8 and ZIP14 were localized in the ciPTEC plasma membrane. Whereas silencing of either ZIP8 or ZIP14 alone did not affect 55Fe uptake, combined silencing significantly reduced 55Fe uptake compared to control (p < 0.05). Furthermore, transferrin receptor 1 (TfR1) and ZIP14, but not ZIP8, colocalized with early endosome antigen 1 (EEA1). TfR1 and ZIP14 also colocalized with uptake of fluorescently labeled transferrin. Furthermore, ZIP14 silencing decreased 55Fe uptake after 55Fe-Transferrin exposure (p < 0.05), suggesting ZIP14 could be involved in early endosomal transport of TBI-derived iron into the cytosol. Our data suggest that human proximal tubular epithelial cells take up TBI and NTBI, where ZIP8 and ZIP14 are both involved in NTBI uptake, but ZIP14, not ZIP8, mediates TBI-derived iron uptake. This knowledge provides more insights in the mechanisms of renal iron handling and suggests that ZIP8 and ZIP14 could be potential targets for limiting renal iron reabsorption and enhancing urinary iron excretion in systemic iron overload disorders.


Subject(s)
Cation Transport Proteins/metabolism , Epithelial Cells/metabolism , Iron/metabolism , Kidney Tubules, Proximal/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Cells, Cultured , Epithelial Cells/pathology , Humans , Kidney Tubules, Proximal/pathology
2.
Nutr Hosp ; 27(6): 2142-5, 2012.
Article in English | MEDLINE | ID: mdl-23588470

ABSTRACT

INTRODUCTION: Transferrin (Tf) exerts a crucial function in the maintenance of systemic iron homeostasis. The expression of the Tf gene is controlled by transcriptional mechanism, although little is known about genetic factors influence. OBJECTIVE: To study the role of rs3811647 in Tf expression using an in-vitro assay on hepatoma cells. DESIGN AND METHODS: Hep3B cells were co-transfected with constructs containing A (VarA-Tf-luc) and G (VarG-Tf-luc) variants of rs3811647, using luciferase as a surrogate reporter of Tf expression. RESULTS: Luciferase assays showed a higher intrinsic enhancer activity (p < 0.05) in the A compared with the G variant. In silico analysis of SNP rs3811647 showed that the A allele might constitute a binding site for the transcription factor glucocorticoid receptor (GR). CONCLUSION: The A allele of SNP rs3811647 increases Tf expression in a manner that might underlie inter-individual variation in serum transferrin levels observed in different population groups.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Introns/genetics , Polymorphism, Single Nucleotide/genetics , Transferrin/biosynthesis , Transferrin/genetics , Binding Sites , Cell Line, Tumor , Humans , Mutagenesis, Site-Directed , Plasmids/genetics , Transfection
3.
Protein Pept Lett ; 15(1): 63-75, 2008.
Article in English | MEDLINE | ID: mdl-18221016

ABSTRACT

MiRP1 (encoded by the KCNE2 gene) is one of a family of five single transmembrane domain voltage-gated potassium (Kv) channel ancillary subunits currently under intense scrutiny to establish their position in channel complexes and elucidate alpha subunit contact points, but its structure is unknown. MiRP1 mutations are associated with inherited and acquired cardiac arrhythmia. Here, synthetic peptides corresponding to human MiRP1 (full-length and separate domains) were structurally analyzed using FTIR and CD spectroscopy. The N-terminal (extracellular) domain was soluble and predominantly non-ordered in aqueous media, but predominantly alpha-helical in L-alpha-lysophosphatidylcholine (LPC) micelles. The MiRP1 transmembrane domain was predominantly a mixture of alpha-helix and non-ordered structure in LPC micelles, with a minor contribution from non-aggregated beta-strand. The intracellular C-terminal domain was insoluble in aqueous solution; reconstitution into non-aqueous environments resulted in solubility and adoption of increasing amounts of alpha-helix, with the solvent order sodium dodecyl sulphate < dimyristoyl L-alpha-phosphatidylcholine (DMPC) < LPC < trifluoroethanol. Correlation of secondary structure changes with lipid transition temperature during heating suggested that the MiRP1 C-terminus incorporates into DMPC bilayers. Full-length MiRP1 was soluble in SDS micelles and calculated to contain 34% alpha-helix, 23% beta-strand and 43% non-ordered structure in this environment, as determined by CD spectroscopy. Thus, MiRP1 is highly dependent upon hydrophobic interaction via lipid and/or protein contacts for adoption of ordered structure without nonspecific aggregation, consistent with a role as a membrane-spanning subunit within Kv channel complexes. These data will provide a structural framework for ongoing mutagenesis-based in situ structure-function studies of MiRP1 and its relatives.


Subject(s)
Potassium Channels, Voltage-Gated/chemistry , Amino Acid Sequence , Humans , Hydrophobic and Hydrophilic Interactions , Lipid Bilayers , Micelles , Molecular Sequence Data , Peptide Fragments/chemistry , Phospholipids/chemistry , Potassium Channels, Voltage-Gated/isolation & purification , Potassium Channels, Voltage-Gated/metabolism , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/chemistry
4.
Gut ; 57(3): 374-82, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17965061

ABSTRACT

BACKGROUND AND AIMS: Reticulo-endothelial macrophages together with duodenal enterocytes coordinate body iron homeostasis. The aim of this study was to investigate the regulatory actions of the hormone hepcidin on ferroportin expression in these two cell types. METHODS: We investigated the in vitro effects of hepcidin in well-characterised human cell culture models of macrophages (differentiated THP-1 cells) and intestinal epithelial cells (Caco-2 cells). The in vivo effects of hepcidin were also investigated in mice injected with a synthetic hepcidin peptide. RESULTS: Exposure to hepcidin (presented either as conditioned medium from interleukin-6-stimulated HuH7 cells or as a synthetic peptide) resulted in a rapid (within 4 h) decrease in ferroportin expression in THP-1 macrophages but had no effect on ferroportin levels in Caco-2 cells. To determine whether these rapid effects of hepcidin were also evident in vivo we injected mice with a synthetic hepcidin peptide. Four hours post-injection, ferroportin levels in the macrophage-rich red pulp of the spleen were decreased significantly and the hepcidin-treated mice developed hypoferraemia. Interestingly, in the same mice there was no effect of hepcidin on duodenal ferroportin protein expression or duodenal iron transport. CONCLUSIONS: These data suggests that the rapid response to hepcidin is cell type and tissue specific. Upon its release, hepcidin initially targets macrophage iron recycling. The duodenum appears to be less sensitive to this initial rise in hepcidin levels. We believe the fact that macrophages respond more acutely to a hepcidin challenge is fully consistent with their central role in maintaining body iron homeostasis.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Intestinal Mucosa/drug effects , Macrophages/drug effects , Animals , Antimicrobial Cationic Peptides/biosynthesis , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/physiology , Biological Transport/drug effects , Caco-2 Cells , Cation Transport Proteins/metabolism , Coculture Techniques , Culture Media, Conditioned , Duodenum/metabolism , Epithelial Cells/drug effects , Hepcidins , Humans , Intestinal Mucosa/cytology , Iron/blood , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Spleen/metabolism
5.
Diabetologia ; 50(10): 2209-17, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17694297

ABSTRACT

AIMS/HYPOTHESIS: GLUT2 is the main renal glucose transporter upregulated by hyperglycaemia, when it becomes detectable at the brush border membrane (BBM). Since glucose-induced protein kinase C (PKC) activation in the kidney is linked to diabetic nephropathy, we investigated the effect of glycaemic status on the protein levels of PKC isoforms alpha, betaI, betaII, delta and epsilon in the proximal tubule, as well as the relationship between them and changes in GLUT2 production at the BBM. METHODS: Plasma glucose concentrations were modulated in rats by treatment with nicotinamide 15 min prior to induction of diabetes with streptozotocin. Levels of GLUT2 protein and PKC isoforms in BBM were measured by western blotting. Additionally, the role of calcium signalling and PKC activation on facilitative glucose transport was examined by measuring glucose uptake in BBM vesicles prepared from proximal tubules that had been incubated either with thapsigargin, which increases cytosolic calcium, or with the PKC activator phorbol 12-myristate,13-acetate (PMA). RESULTS: Thapsigargin and PMA enhanced GLUT-mediated glucose uptake, but had no effect on sodium-dependent glucose transport. Diabetes significantly increased the protein levels of GLUT2 and PKC-betaI at the BBM. Levels of GLUT2 and PKC-betaI correlated positively with plasma glucose concentration. Diabetes had no effect on BBM levels of alpha, betaII, delta or epsilon isoforms of PKC. CONCLUSIONS/INTERPRETATION: Enhanced GLUT2-mediated glucose transport across the proximal tubule BBM during diabetic hyperglycaemia is closely associated with increased PKC-betaI. Thus, altered levels of GLUT2 and PKC-betaI proteins in the BBM may be important factors in the pathogenic processes underlying diabetic renal injury.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Glucose Transporter Type 2/metabolism , Kidney Tubules, Proximal/metabolism , Microvilli/metabolism , Protein Kinase C/metabolism , Animals , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/enzymology , Enzyme Activation , Glucose Transporter Type 2/drug effects , Kidney/anatomy & histology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/physiology , Male , Microvilli/drug effects , Microvilli/physiology , Niacinamide/pharmacology , Organ Size , Protein Kinase C/drug effects , Protein Kinase C beta , Rats , Rats, Sprague-Dawley , Tetradecanoylphorbol Acetate/pharmacology , Thapsigargin/pharmacology , Weight Gain
6.
Kidney Int ; 72(2): 166-73, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17457376

ABSTRACT

Hyperphosphatemia is an important consequence of chronic renal failure (CRF). Lowering of the plasma phosphate concentration is believed to be critical in the management of patients with CRF, especially those on dialysis. Reports of the effect of CRF on the intestinal handling of phosphate in vitro have been conflicting; but what happens in vivo has not been studied. What effect a reduction in the dietary phosphate intake has on intestinal phosphate absorption in CRF in vivo is unclear. In this study, we have used the in situ intestine loop technique to determine intestinal phosphate absorption in the 5/6-nephrectomy rat model of CRF under conditions of normal and restricted dietary phosphate intake. In this model of renal disease, we found that there is no significant change in the phosphate absorption in either the duodenum or jejunum regardless of the dietary phosphate intake. There was also no change in the expression of the messenger RNA of the major intestinal phosphate carrier the sodium-dependent-IIb transporter. Furthermore, we found no change in the intestinal villus length or in the location of phosphate uptake along the villus. Our results indicate that in CRF, unlike the kidney, there is no reduction in phosphate transport across the small intestine. This makes intestinal phosphate absorption a potential target in the prevention and treatment of hyperphosphatemia.


Subject(s)
Hypophosphatemia/metabolism , Intestinal Absorption , Kidney Failure, Chronic/complications , Phosphates/pharmacokinetics , Animals , Disease Models, Animal , Homeostasis , Hypophosphatemia/etiology , Male , Nephrectomy , Rats , Rats, Sprague-Dawley
7.
Eur J Clin Invest ; 36(1): 35-40, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16403008

ABSTRACT

BACKGROUND: Only 10% of dietary iron is absorbed in the duodenum which implies that 90% (approximately 9 mg day(-1)) reaches the lower small intestine and colon. Therefore the purpose of this study was to assess the iron transport capacity of the rat proximal colon and to determine whether iron absorption is regulated by changes in dietary iron content. MATERIALS AND METHODS: Rats were fed for 14 days on either iron adequate (44 mg Fe kg(-1) diet) or iron-deficient (< 0.5 mg Fe kg(-1) diet) diets. The 59Fe transport across the colonic epithelium and its subsequent appearance in the blood were measured in vivo. In separate studies the colon was excised and used to measure divalent metal transporter expression. RESULTS: Divalent metal transporter (DMT1) was expressed at the apical membrane of the surface epithelium in rat proximal colon. In animals fed an iron-deficient diet, DMT1 mRNA and protein expression were increased. This was accompanied by a significant increase in tissue 59Fe uptake. CONCLUSIONS: The proximal colon can absorb non-haem iron from the intestinal lumen. The purpose of this mechanism remains to be elucidated.


Subject(s)
Colon/metabolism , Iron, Dietary/pharmacokinetics , Animals , Biological Transport , Blotting, Western , Cation Transport Proteins/metabolism , Duodenum/metabolism , Heme , Intestinal Absorption , Intestinal Mucosa/metabolism , Iron/blood , Iron Deficiencies , Iron, Dietary/administration & dosage , Iron-Binding Proteins/metabolism , Male , Rats , Rats, Wistar
8.
Gut ; 54(10): 1391-5, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15914574

ABSTRACT

BACKGROUND: Despite the requirement for increased iron delivery for erythropoiesis during hypoxia, there is very little information on how duodenal iron uptake and its transfer to the blood adapts to this condition. AIMS: To assess the effects of 30 days of chronic hypoxia in rats on luminal iron uptake and transfer of the metal to blood, together with gene expression of hepcidin, a proposed negative regulator of iron transport. METHODS: 59-Fe uptake by isolated duodenum and its transfer to blood by in vivo duodenal segments was measured after exposure of rats to room air or 10% oxygen for four weeks. Liver hepcidin expression was measured by real time reverse transcription-polymerase chain reaction. The effects of hypoxia on hepcidin gene expression by HepG2 cells was also determined. RESULTS: Hypoxia did not affect villus length but enhanced (+192.6%) luminal iron uptake by increasing the rate of uptake by all enterocytes, particularly those on the upper villus. Hypoxia promoted iron transfer to the blood but reduced mucosal iron accumulation in vivo by 66.7%. Hypoxia reduced expression of hepcidin mRNA in both rat liver and HepG2 cells. CONCLUSIONS: Prolonged hypoxia enhances iron transport from duodenal lumen to blood but the process is unable to fully meet the iron requirement for increased erythropoiesis. Reduced secretion of hepcidin may be pivotal to the changes in iron absorption. The processes responsible for suppression of hepcidin expression are unknown but are likely to involve a direct effect of hypoxia on hepatocytes.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Duodenum/metabolism , Hypoxia/metabolism , Iron/pharmacokinetics , Animals , Antimicrobial Cationic Peptides/genetics , Cells, Cultured , Chronic Disease , Disease Models, Animal , Gene Expression Regulation/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Hepcidins , Hypoxia/genetics , Iron/blood , Liver/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
10.
Cells Tissues Organs ; 178(3): 168-79, 2004.
Article in English | MEDLINE | ID: mdl-15655334

ABSTRACT

The effects of extracellular ATP on fluid secretion and reabsorption by renal epithelial cells, as well as its known effects on cell proliferation and death, are potentially important contributory factors in the development and growth of renal cysts. In this study, we have investigated the protein and mRNA expression of several P2Y receptor subtypes (P2Y(1,2,4,6)), as well as the P2X(5) and P2X(7) receptors, in kidney tissue from the Han:SPRD (cy/+) rat model of polycystic kidney disease. All of the P2Y receptors tested for, and the P2X(5) and P2X(7) subtypes, were located on the cyst-lining cells of Han:SPRD (cy/+) rat polycystic kidneys; most immunostaining was cytosolic and we could not confidently localize it to one or other membrane. However, the staining pattern for P2Y(6) was uniquely granular when compared with the other P2 receptors. P2Y(2) and P2Y(6) receptor mRNA was increased in both homozygote (cy/cy) and heterozygote (cy/+) rat kidneys when compared with unaffected littermates. The protein levels of P2Y(2) and P2Y(6) receptors were also increased, being undetectable or at a low level, respectively, in control tissue. Finally, P2X(7) receptor mRNA was increased in cy/+, but not in cy/cy rat kidneys. Our results show that a number of P2Y receptor subtypes, as well as the P2X(5) and P2X(7) receptors, are clearly expressed in cyst-lining cells in the Han:SPRD (cy/+) rat model of renal cystic disease. Furthermore, P2Y(2) and P2Y(6) receptor mRNA and protein levels are markedly increased in cystic rat kidneys compared with normal rats of the same genetic background. Thus, the most consistent findings were an increase in the expression of P2Y(2), P2Y(6) and P2X(7) receptors in cystic tissue. Given the widely reported effects of stimulating these P2 receptor subtypes in epithelial and other renal cells, they could contribute to the development and growth of renal cysts: extracellular ATP and its products 'trapped' in cyst fluid may activate P2 receptors expressed by cyst-lining cells, causing cyst expansion from increased fluid secretion and/or reduced reabsorption, as well as an increase in cell turnover (re-modeling).


Subject(s)
Adenosine Triphosphate/metabolism , Polycystic Kidney, Autosomal Dominant/metabolism , Receptors, Purinergic P2/metabolism , Animals , Apoptosis/genetics , Disease Models, Animal , Heterozygote , Homozygote , Humans , Immunoblotting , Immunohistochemistry , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/pathology , Polymerase Chain Reaction , Proteins/analysis , Proteins/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains
11.
Gut ; 52(4): 510-3, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12631660

ABSTRACT

BACKGROUND: Genetic haemochromatosis is a common hereditary iron loading disorder in humans. The disease is associated with loss of function mutations in the HFE gene. This is thought to change iron stores via increased iron absorption. AIMS: In this study we investigated how adaptation of mucosal reductase activity is engaged in this process and how the changes compare with adaptation seen when an iron deficient diet is fed. METHODS: Duodenal mucosal surface reductase was measured with nitroblue tetrazolium in age matched groups of male Hfe knockout mice (Hfe) and wild- type mice fed a purified diet containing normal (iron adequate), high (iron rich), or low (iron deficient) iron concentrations. RESULTS: Reductase activity increased when mice were fed an iron deficient diet and decreased when they were fed an iron rich diet. Total villus activity, as measured by the average area under the activity curve along the crypt-villus axis, was increased 2.8-2.9-fold by iron deficiency in both genotypes. Approximately half of this difference was attributable to the significantly increased length of the villi in mice on an iron deficient diet (p<0.05). Hfe knockout did not affect villus length but increased mucosal reductase activity near the villus tips. Similar increases (1.3-1.6-fold) were seen on all diets but the increase was significant for iron deficient and iron loaded diets only (p<0.05). CONCLUSION: Hfe gene product and dietary iron downregulate villus reductase activity in mice.


Subject(s)
Duodenum/enzymology , Hemochromatosis/enzymology , Iron, Dietary/administration & dosage , Oxidoreductases/metabolism , Animals , Duodenum/pathology , Hemochromatosis/genetics , Hemochromatosis/pathology , Hemochromatosis Protein , Hemoglobins/analysis , Histocompatibility Antigens Class I , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Iron/analysis , Iron Deficiencies , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Tissue Distribution
12.
Clin Sci (Lond) ; 104(3): 253-8, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12605582

ABSTRACT

Glucagon is known to affect glomerular filtration rate and renal tubular solute and fluid transport, although it is only thought to act directly on the thick ascending limb (TAL) and collecting duct (CD). Indeed, previous studies have detected glucagon-sensitive adenylate cyclase exclusively in these nephron segments, suggesting the presence of glucagon receptors. In the present study, we have demonstrated for the first time that glucagon receptor mRNA is expressed in the rat proximal tubule, as well as in the TAL and CD. By autoradiography, we have also shown that specific binding of glucagon occurs in both the renal cortex and medulla. In addition, using proximal tubule brush-border membrane (BBM) vesicles for studies of glucose transport, we have established that glucagon stimulates glucose uptake via a facilitative GLUT-mediated transport process (by 58%; P < 0.005), whereas cAMP stimulates only the sodium glucose-linked transporter ('SGLT')-mediated glucose uptake (by 53%; P < 0.05). Taken together, these findings suggest that glucagon could have a role in controlling proximal tubular transport function, including glucose reabsorption, but unlike in the TAL and CD, the proximal tubule glucagon receptor might not be coupled primarily to adenylate cyclase.


Subject(s)
Glucagon/physiology , Kidney Tubules, Proximal/metabolism , RNA, Messenger/metabolism , Receptors, Glucagon/genetics , Animals , Biological Transport/physiology , Glucose/metabolism , Kidney/metabolism , Kidney Medulla/metabolism , Male , Microvilli/metabolism , Monosaccharide Transport Proteins/physiology , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, Glucagon/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Transport Vesicles/metabolism
13.
Best Pract Res Clin Haematol ; 15(2): 243-59, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12401306

ABSTRACT

Iron is an essential element playing a vital role in many cellular processes. This requirement is complicated by the fact that environmental iron is invariably present as insoluble Fe(3+) leading to poor bioavailability and toxicity, since even low concentrations of iron catalyse the production of damaging reactive oxygen species. As a result organisms have evolved efficient uptake and transport systems to extract iron from their environment as well as ferritins that store iron in a non-toxic form. In higher organisms, the first membrane barrier encountered is the apical surface of the duodenal enterocyte, a specialized absorptive cell of the intestinal epithelium that undertakes vectorial transport of iron. Iron is initially solubilized by reduction and Fe(2+) is transported across the cell membrane by a carrier-mediated transport process. This is followed by intracellular transfer of iron to the basolateral enterocyte membrane with subsequent transfer and release of iron to transferrin in the portal blood. A second site of iron transport is at the placento-fetal barrier where similar principles operate. In this review we describe recently identified transmembrane transporters and associated accessory proteins responsible for iron transport at these two sites.


Subject(s)
Duodenum/metabolism , Iron/metabolism , Placenta/metabolism , Animals , Biological Transport , Carrier Proteins , Cell Membrane/metabolism , Duodenum/ultrastructure , Female , Humans , In Vitro Techniques , Placenta/ultrastructure , Pregnancy
14.
Eur J Clin Invest ; 32(8): 603-12, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12190960

ABSTRACT

BACKGROUND: HFE knockout mice (C57BL/6 x 129/Ola strain) mimic the functional aberrations of human hereditary haemochromatosis (HH) in short-term experiments. The present study investigates functional and morphological long-term changes. METHODS: HFE(o/o), HFE(+/o) and HFE(+/+) mice were maintained on iron-rich and control diets for 2 weeks, 3, 12 and 18 months. Light microscopic tissue iron distribution, pathomorphological alterations, tissue iron content and oxidative stress were analysed in liver, pancreas, spleen, gastrointestinal tract, kidneys and myocardium. Additionally, duodenal 59Fe absorption and 59Fe whole body loss were measured. RESULTS: Iron distribution between organs and microscopic iron deposition in the tissues resembled the patterns described in HH. After 3 months of iron-rich feeding duodenal 59Fe absorption decreased to approximately 15% of iron-adequate controls but remained about twice as high in HFE(o/o) as in HFE(+/+) mice. Hepatic iron concentrations reached only half the values known to induce hepatic fibrosis in rats and humans, while whole body 59Fe loss was about twice as high. Consequently no hepatic fibrosis developed, although massive hepatocellular iron deposition and indication for oxidative stress were observed. CONCLUSION: C57BL/6 x 129/O1a HFE(o/o) mice mimic HH iron distribution and the regulation of intestinal iron absorption after long-term feeding. However, characteristic morphological late changes in untreated HH are not modelled.


Subject(s)
Hemochromatosis/genetics , Histocompatibility Antigens Class I/genetics , Iron, Dietary/metabolism , Membrane Proteins/genetics , Models, Animal , Animals , Duodenum/metabolism , Hemochromatosis/metabolism , Hemochromatosis Protein , Intestinal Absorption , Iron Radioisotopes , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Time Factors , Tissue Distribution
15.
FEBS Lett ; 507(2): 137-41, 2001 Oct 26.
Article in English | MEDLINE | ID: mdl-11684086

ABSTRACT

Trace metals influence the absorption of each other from the diet and it has been suggested that the divalent metal transporter (DMT1) represents a common uptake pathway for these important micronutrients. However, compelling evidence from our laboratory suggests that DMT1 is predominantly an iron transporter, with lower affinity for other metals. Several studies have shown that increasing dietary iron downregulates DMT1. Interestingly, our current data indicate that zinc upregulates DMT1 protein and mRNA expression and also pH-dependent iron uptake. Transepithelial flux of iron was also increased and was associated with a rise in IREG1 mRNA expression.


Subject(s)
Cation Transport Proteins/genetics , Gene Expression , Iron-Binding Proteins , Zinc/metabolism , Caco-2 Cells , Cation Transport Proteins/physiology , Humans , Intestines/pathology , Iron/metabolism
16.
Biochem Biophys Res Commun ; 285(3): 588-93, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11453632

ABSTRACT

Perfusion of isolated small intestine with the procyanidin dimers B2 and B5 extracted from cocoa indicated that both forms of dimer are transferred to the serosal side of enterocytes but only to a very small extent (<1% of the total transferred flavanol-like compounds). However, perfusion of dimer mainly resulted in large amounts of unmetabolised/unconjugated epicatechin monomer being detected on the serosal side (95.8%). The cleavage of dimer during transfer seemed to be energy-dependent, requiring an intact cell system, as incubation with jejunal homogenates failed to yield epicatechin. Low levels methylated dimer were also detected (3.2%), but no conjugates and metabolites of epicatechin indicating that metabolism of monomer and dimer is limited during dimer cleavage/translocation. The methylation of dimer may be by catechol-O-methyltransferase, however, at high concentrations of dimer COMT activity is reduced leading to an inhibition of both monomer and dimer O-methylation.


Subject(s)
Biflavonoids , Catechin/metabolism , Catechin/pharmacokinetics , Intestine, Small/metabolism , Proanthocyanidins , Animals , Biological Availability , Biological Transport, Active , Biotransformation , Cacao/chemistry , Catechin/analogs & derivatives , Catechin/isolation & purification , Catechol O-Methyltransferase/metabolism , Chromatography, High Pressure Liquid , Dimerization , Enterocytes/metabolism , In Vitro Techniques , Mass Spectrometry , Perfusion , Rats
17.
Biochem J ; 356(Pt 3): 883-9, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11389698

ABSTRACT

Maternal iron deficiency during pregnancy induces anaemia in the developing fetus; however, the severity tends to be less than in the mother. The mechanism underlying this resistance has not been determined. We have measured placental expression of proteins involved in iron transfer in pregnant rats given diets with decreasing levels of iron and examined the effect of iron deficiency on iron transfer across BeWo cell layers, a model for placental iron transfer. Transferrin receptor expression was increased at both mRNA and protein levels. Similarly, expression of the iron-responsive element (IRE)-regulated form of the divalent metal transporter 1 (DMT1) was also increased. In contrast, the non-IRE regulated isoform showed no change in mRNA levels. Protein levels of DMT1 increased significantly. Iron efflux is thought to be mediated by the metal transporter protein, IREG1/ferroportin1/MTP1, and oxidation of Fe(II) to Fe(III) prior to incorporation into fetal transferrin is carried out by the placental copper oxidase. Expression of IREG1 was not altered by iron deficiency, whereas copper oxidase activity was increased. In BeWo cells made iron deficient by treatment with desferrioxamine ('deferioxamine'), iron accumulation from iron-transferrin increased, in parallel with increased expression of the transferrin receptor. At the same time, iron efflux also increased, showing a higher flux of iron from the apical to the basolateral side. The data show that expression of placental proteins of iron transport are up-regulated in maternal iron deficiency, resulting in an increased efficiency of iron flux and a consequent minimization of the severity of fetal anaemia.


Subject(s)
Anemia, Iron-Deficiency/metabolism , Carrier Proteins/metabolism , Cation Transport Proteins , Iron-Binding Proteins , Iron/metabolism , Membrane Proteins/metabolism , Placenta/metabolism , Animals , Base Sequence , Carrier Proteins/genetics , Cell Line , DNA Primers , Female , In Vitro Techniques , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Transferrin/metabolism
18.
Biochem J ; 354(Pt 3): 493-500, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11237853

ABSTRACT

There is considerable current interest in the cytoprotective effects of natural antioxidants against oxidative stress. In particular, epicatechin, a major member of the flavanol family of polyphenols with powerful antioxidant properties in vitro, has been investigated to determine its ability to attenuate oxidative-stress-induced cell damage and to understand the mechanism of its protective action. We have induced oxidative stress in cultured human fibroblasts using hydrogen peroxide and examined the cellular responses in the form of mitochondrial function, cell-membrane damage, annexin-V binding and caspase-3 activation. Since one of the major metabolites of epicatechin in vivo is 3'-O-methyl epicatechin, we have compared its protective effects with that of epicatechin. The results provide the first evidence that 3'-O-methyl epicatechin inhibits cell death induced by hydrogen peroxide and that the mechanism involves suppression of caspase-3 activity as a marker for apoptosis. Furthermore, the protection elicited by 3'-O-methyl epicatechin is not significantly different from that of epicatechin, suggesting that hydrogen-donating antioxidant activity is not the primary mechanism of protection.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Catechin/pharmacology , Fibroblasts/drug effects , Oxidative Stress/drug effects , Caspase 3 , Catechin/analogs & derivatives , Cells, Cultured , Fibroblasts/metabolism , Humans , Hydrogen Peroxide/pharmacology , Lipoproteins, LDL/pharmacology , Methylation , Mitochondria/drug effects
19.
Biochem Biophys Res Commun ; 277(2): 507-12, 2000 Oct 22.
Article in English | MEDLINE | ID: mdl-11032751

ABSTRACT

There is considerable interest in the bioavailability of polyphenols and their bioactivity in vivo. We have studied the absorption and metabolism of catechin and epicatechin in the small intestine and the comparative transfer across the jejunum and ileum. Perfusion of isolated jejunum with the flavanols resulted in glucuronidation ( approximately 45%), O-methylation: 3'-O-Methyl- and 4'-O-methyl- ( approximately 30%), and O-methyl-glucuronidation ( approximately 20% of total flavanols identified) during transfer across the enterocytes to the serosal side. This demonstrates the activity of catechol-O-methyl transferases in the metabolism of flavanols and suggests that these metabolites and conjugates are likely to enter the portal vein. In contrast, in the case of the ileum, the majority of the flavanols appeared on the serosal side unmetabolised and the total percentage of flavanols transferred was higher than that in the jejunum ( approximately fivefold).


Subject(s)
Catechin/chemistry , Intestine, Small/metabolism , Animals , Biological Transport , Catechin/metabolism , Catechol O-Methyltransferase/metabolism , Chromatography, High Pressure Liquid , Gas Chromatography-Mass Spectrometry , Glucuronides/metabolism , Ileum/metabolism , Jejunum/metabolism , Mass Spectrometry , Methylation , Rats
20.
Biochem Biophys Res Commun ; 272(1): 212-7, 2000 May 27.
Article in English | MEDLINE | ID: mdl-10872829

ABSTRACT

We have studied the absorption and metabolism of resveratrol in the jejunum in an isolated rat small intestine model. Only small amounts of resveratrol were absorbed across the enterocytes of the jejunum and ileum unmetabolised. The major compound detected on the serosal side was the glucuronide conjugate of resveratrol (96.5% +/- 4.6 of the amount absorbed) indicating the susceptibility of resveratrol to glucuronidation during transfer across the rat jejunum. The presence of the glucuronide was confirmed using HPLC-PDA and nanoES-MS/MS techniques. These findings suggest that resveratrol is most likely to be in the form of a glucuronide conjugate after crossing the small intestine and entering the blood circulation. This will have important implications for the biological functions of resveratrol in vivo.


Subject(s)
Antioxidants/pharmacokinetics , Intestine, Small/metabolism , Stilbenes/pharmacokinetics , Animals , Antioxidants/chemistry , Biological Transport, Active , Glucuronides/chemistry , Glucuronides/pharmacokinetics , Ileum/metabolism , In Vitro Techniques , Intestinal Absorption , Jejunum/metabolism , Kinetics , Male , Perfusion , Rats , Rats, Sprague-Dawley , Resveratrol , Stilbenes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...