Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
3.
Med Hypotheses ; 72(5): 504-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19195795

ABSTRACT

This communication explores the temporal link between the age-associated increase in body iron stores and the age-related incidence of Alzheimer's disease (AD), the most prevalent cause of senile dementia. Body iron stores that increase with age could be pivotal to AD pathogenesis and progression. Increased stored iron is associated with common medical conditions such as diabetes and vascular disease that increase risk for development of AD. Increased stored iron could also promote oxidative stress/free radical damage in vulnerable neurons, a critical early change in AD. A ferrocentric model of AD described here forms the basis of a rational, easily testable experimental therapeutic approach for AD, which if successful, would be both widely applicable and inexpensive. Clinical studies have shown that calibrated phlebotomy is an effective way to reduce stored iron safely and predictably without causing anemia. We hypothesize that reducing stored iron by calibrated phlebotomy to avoid iron deficiency will improve cerebrovascular function, slow neurodegenerative change, and improve cognitive and behavioral functions in AD. The hypothesis is eminently testable as iron reduction therapy is useful for chronic diseases associated with iron excess such as nonalcoholic steatohepatitis (NASH), atherosclerosis, hereditary hemochromatosis and thalassemia. Testing this hypothesis could provide valuable insight into the causation of AD and suggest novel preventive and treatment strategies.


Subject(s)
Alzheimer Disease/therapy , Iron/isolation & purification , Phlebotomy , Humans , Iron/blood , Models, Theoretical
4.
Hepatology ; 46(6): 1927-834, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17854053

ABSTRACT

UNLABELLED: Porphyria cutanea tarda is a liver disease characterized by elevated hepatic iron and excessive production of uroporphyrin (URO). Phlebotomy is an effective treatment that probably acts by reducing hepatic iron. Here we used Hfe(-/-) mice to compare the effects on hepatic URO accumulation of two different methods of hepatic iron depletion: iron chelation using deferiprone (L1) versus iron-deficient diets. Hfe(-/-) mice in a 129S6/SvEvTac background were fed 5-aminolevulinic acid (ALA), which results in hepatic URO accumulation, and increasing doses of L1 in the drinking water. Hepatic URO accumulation was completely prevented at low L1 doses, which partially depleted hepatic nonheme iron. By histological assessment, the decrease in hepatic URO accumulation was associated with greater depletion of iron from hepatocytes than from Kupffer cells. The L1 treatment had no effect on levels of hepatic cytochrome P4501A2 (CYP1A2). L1 also effectively decreased hepatic URO accumulation in C57BL/6 Hfe(-/-) mice treated with ALA and a CYP1A2 inducer. ALA-treated mice maintained on defined iron-deficient diets, rather than chow diets, did not develop uroporphyria, even when the animals were iron-supplemented either directly in the diet or by iron dextran injection. CONCLUSION: The results suggest that dietary factors other than iron are involved in the development of uroporphyria and that a modest depletion of hepatocyte iron by L1 is sufficient to prevent URO accumulation.


Subject(s)
Iron Chelating Agents/therapeutic use , Iron Deficiencies , Porphyria Cutanea Tarda/diet therapy , Porphyria Cutanea Tarda/drug therapy , Pyridones/therapeutic use , Animals , Deferiprone , Disease Models, Animal , Liver/chemistry , Male , Mice , Uroporphyrins/analysis
5.
Hepatology ; 45(1): 187-94, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17187429

ABSTRACT

UNLABELLED: Excess hepatic iron is known to enhance both porphyria cutanea tarda (PCT) and experimental uroporphyria. Since previous studies have suggested a role for ascorbate (AA) in suppressing uroporphyria in AA-requiring rats (in the absence of excess iron), the present study investigated whether AA could suppress uroporphyria produced by excess hepatic iron. Hepatic URO accumulation was produced in AA-requiring Gulo(-/-) mice by treatment with 3,3',4,4',5-pentachlorbiphenyl, an inducer of CYP1A2, and 5-aminolevulinic acid. Mice were administered either sufficient AA (1000 ppm) in the drinking water to maintain near normal hepatic AA levels or a lower intake (75 ppm) that resulted in 70 % lower hepatic AA levels. The higher AA intake suppressed hepatic URO accumulation in the absence of administered iron, but not when iron dextran (300-500 mg Fe/kg) was administered. This effect of iron was not due to hepatic AA depletion since hepatic AA content was not decreased. The effect of iron to prevent AA suppression of hepatic URO accumulation was not observed until a high hepatic iron threshold was exceeded. At both low and high AA intakes, hepatic malondialdehyde (MDA), an indicator of oxidative stress, was increased three-fold by high doses of iron dextran. MDA was considerably increased even at low iron dextran doses, but without any increase in URO accumulation. The level of hepatic CYP1A2 was unaffected by either AA intake. CONCLUSION: In this mouse model of PCT, AA suppresses hepatic URO accumulation at low, but not high hepatic iron levels. These results may have implications for the management of PCT.


Subject(s)
Ascorbic Acid Deficiency/metabolism , Ascorbic Acid/pharmacology , Iron/pharmacology , Porphyria Cutanea Tarda/metabolism , Uroporphyrins/metabolism , Aminolevulinic Acid , Animals , Ascorbic Acid/genetics , Ascorbic Acid/metabolism , Ascorbic Acid Deficiency/genetics , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Dietary Supplements , Disease Models, Animal , Dose-Response Relationship, Drug , Iron/metabolism , Iron-Dextran Complex/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/drug effects , Polychlorinated Biphenyls , Porphyria Cutanea Tarda/chemically induced , Porphyria Cutanea Tarda/drug therapy
6.
Arch Biochem Biophys ; 439(1): 1-11, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15946643

ABSTRACT

Primary cultures of chick embryo hepatocytes have been used to study the mechanisms by which various drugs and other chemicals cause accumulation of porphyrin intermediates of the heme pathway. When these cultures are incubated with the heme precursor, 5-aminolevulinic acid (ALA), there is a major accumulation of protoporphyrin. However, in the presence of ALA, addition of insulin caused a striking increase in accumulation of uroporphyrin I and coproporphyrin III, whereas addition of glucagon mainly caused an increase in uroporphyrin I. Treatment with both insulin and glucagon resulted in additive increases in uroporphyrin, but not coproporphyrin. Antioxidants abolished the uroporphyrin I accumulation and increased coproporphyrin III. Insulin caused an increase in uptake of ALA and an increase in porphobilinogen accumulation, suggesting that the accumulation of uroporphyrin I is due to increased flux through the heme pathway. Apparently, this increased flux could particularly affect the utilization of the intermediate hydroxymethylbilane, which would result in accumulation of uroporphyrin I.


Subject(s)
Aminolevulinic Acid/metabolism , Coproporphyrins/biosynthesis , Gastrointestinal Agents/pharmacology , Glucagon/pharmacology , Hepatocytes/metabolism , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Uroporphyrins/biosynthesis , Aminolevulinic Acid/pharmacology , Animals , Cells, Cultured , Chick Embryo , Hepatocytes/drug effects , Photosensitizing Agents/pharmacology
7.
Hepatology ; 40(4): 942-50, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15382179

ABSTRACT

Two major risk factors for porphyria cutanea tarda (PCT) are alcohol consumption and homozygosity for the C282Y mutation in the hereditary hemochromatosis gene (HFE). We recently described an animal model for alcohol-induced uroporphyria, using Hfe(-/-) mice. In the present study we show that this effect is dependent on genetic background and ethanol dose. In the 129S6/SvEvTac (129) strain, treatment with 15% ethanol in the drinking water for 6.5 months produced an accumulation of hepatic uroporphyrin (URO) 4-fold higher than that observed with 10% ethanol, a 90% decrease in uroporphyrinogen decarboxylase activity (UROD), and further increased the activities of hepatic 5-aminolevulinate synthase (ALAS) and CYP1A2. Hepatic nonheme iron (NHFe) and hepatocyte iron staining were not further increased by 15% compared to 10% ethanol. Treatment of C57BL/6 Hfe(-/-) mice with 15% ethanol for 6.5 months did not increase hepatic URO. Although NHFe was increased by ethanol, the resulting level was only half that of ethanol-treated 129 Hfe(-/-) mice. ALAS induction was similar in both Hfe(-/-) strains. In wild-type 129 mice treated with ethanol for 6 to 7 months, administration of iron dextran increased hepatic URO accumulation and decreased UROD activity. In conclusion, this study demonstrates a strong effect of genetic background on ethanol-induced uroporphyria, which is probably due to a greater effect of ethanol on iron metabolism in the susceptible strain.


Subject(s)
Alcohol Drinking/genetics , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Histocompatibility Antigens Class I/genetics , Membrane Proteins/genetics , Porphyria Cutanea Tarda/genetics , 5-Aminolevulinate Synthetase/metabolism , Alcohol Drinking/metabolism , Animals , Cytochrome P-450 CYP1A2/metabolism , Hemochromatosis Protein , Iron/metabolism , Iron-Dextran Complex/pharmacology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Porphyria Cutanea Tarda/etiology , Porphyria Cutanea Tarda/metabolism , Uroporphyrins/metabolism
8.
Hepatology ; 37(2): 351-8, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12540785

ABSTRACT

Two major risk factors for the development of porphyria cutanea tarda (PCT) are alcohol consumption and homozygosity for the C282Y mutation in the hereditary hemochromatosis gene (HFE). To develop an animal model, Hfe knockout mice were treated continuously with 10% ethanol in drinking water. By 4 months, uroporphyrin (URO) was detected in the urine. At 6 to 7 months, hepatic URO was increased and hepatic uroporphyrinogen decarboxylase (UROD) activity was decreased. Untreated Hfe(-/-) mice or wild-type mice treated with or without ethanol did not show any of these biochemical changes. Treatment with ethanol increased hepatic nonheme iron and hepatic 5-aminolevulinate synthase activity in Hfe(-/-) but not wild-type mice. The increases in nonheme iron in Hfe(-/-) mice were associated with diffuse increases in iron staining of parenchymal cells but without evidence of significant liver injury. In conclusion, the results of this study suggest that the uroporphyrinogenic effect of ethanol is mediated by its effects on hepatic iron metabolism. Ethanol-treated Hfe(-/-) mice seem to be an excellent model for studies of alcohol-mediated PCT.


Subject(s)
Ethanol/pharmacology , Membrane Proteins/deficiency , Porphyria Cutanea Tarda/chemically induced , Porphyria Cutanea Tarda/metabolism , Uroporphyrins/urine , 5-Aminolevulinate Synthetase/metabolism , Animals , Cytochrome P-450 CYP1A2/metabolism , Disease Models, Animal , Hemochromatosis Protein , Histocompatibility Antigens Class I/genetics , Iron/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout/genetics , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...