Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Neurosci Lett ; 764: 136222, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34500002

ABSTRACT

A growing body of evidence indicates an association between flavin-containing monooxygenase (FMO) and neurodegeneration, including Parkinson's disease (PD); however, the details of this association are unclear. We previously showed that the level of Fmo1 mRNA is decreased in an in vitro rotenone model of parkinsonism. To further explore the potential involvement of FMO1 deficiency in parkinsonism, we generated Fmo1 knockout (KO) mice and examined the survival of dopaminergic neurons and relative changes. Fmo1 KO mice exhibited loss of tyrosine hydroxylase-positive neurons, decreased levels of tyrosine hydroxylase and Parkin proteins, and increased levels of pro-inflammatory cytokines (IL1ß and IL6) in the nigrostriatal region. Moreover, the protein levels of PTEN induced kinase 1 (PINK1) and p62, and the Microtubule associated protein 1 light chain 3 (LC3)-II/I ratio were not significantly altered in Fmo1 KO mice (P > 0.05). FMO1 deficiency promotes neuroinflammation in dopaminergic neurons in mice, thus may plays a potential pathological role in dopaminergic neuronal loss. These findings may provide new insight into the pathogenesis of PD.


Subject(s)
Dopaminergic Neurons/pathology , Neuroinflammatory Diseases/immunology , Oxygenases/deficiency , Parkinson Disease/immunology , Substantia Nigra/pathology , Animals , Dopaminergic Neurons/immunology , Dopaminergic Neurons/metabolism , Humans , Male , Mice , Mice, Knockout , Neuroinflammatory Diseases/pathology , Oxygenases/genetics , Parkinson Disease/pathology , Protein Kinases/analysis , Protein Kinases/metabolism , Sequestosome-1 Protein/analysis , Sequestosome-1 Protein/metabolism , Substantia Nigra/cytology , Substantia Nigra/immunology , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/analysis , Tyrosine 3-Monooxygenase/metabolism , Ubiquitin-Protein Ligases/analysis , Ubiquitin-Protein Ligases/metabolism
2.
Drug Metab Dispos ; 45(9): 982-989, 2017 09.
Article in English | MEDLINE | ID: mdl-28646079

ABSTRACT

We have previously identified flavin-containing monooxygenase 5 (FMO5) as a regulator of metabolic aging. The aim of the present study was to investigate the role of FMO5 in glucose homeostasis and the impact of diet and gut flora on the phenotype of mice in which the Fmo5 gene has been disrupted (Fmo5-/- mice). In comparison with wild-type (WT) counterparts, Fmo5-/- mice are resistant to age-related changes in glucose homeostasis and maintain the higher glucose tolerance and insulin sensitivity characteristic of young animals. When fed a high-fat diet, they are protected against weight gain and reduction of insulin sensitivity. The phenotype of Fmo5-/- mice is independent of diet and the gut microbiome and is determined solely by the host genotype. Fmo5-/- mice have metabolic characteristics similar to those of germ-free mice, indicating that FMO5 plays a role in sensing or responding to gut bacteria. In WT mice, FMO5 is present in the mucosal epithelium of the gastrointestinal tract where it is induced in response to a high-fat diet. In comparison with WT mice, Fmo5-/- mice have fewer colonic goblet cells, and they differ in the production of the colonic hormone resistin-like molecule ßFmo5-/- mice have lower concentrations of tumor necrosis factor α in plasma and of complement component 3 in epididymal white adipose tissue, indicative of improved inflammatory tone. Our results implicate FMO5 as a regulator of body weight and of glucose disposal and insulin sensitivity and, thus, identify FMO5 as a potential novel therapeutic target for obesity and insulin resistance.


Subject(s)
Blood Glucose/metabolism , Gastrointestinal Microbiome/physiology , Oxygenases/metabolism , Age Factors , Animals , Diet, High-Fat , Homeostasis , Insulin/blood , Insulin Resistance/physiology , Intestinal Mucosa/metabolism , Intestines/enzymology , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygenases/deficiency , Oxygenases/genetics , Phenotype , Weight Gain/physiology
3.
Biochem Pharmacol ; 96(3): 267-77, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26049045

ABSTRACT

We report the production and metabolic phenotype of a mouse line in which the Fmo5 gene is disrupted. In comparison with wild-type (WT) mice, Fmo5(-/-) mice exhibit a lean phenotype, which is age-related, becoming apparent after 20 weeks of age. Despite greater food intake, Fmo5(-/-) mice weigh less, store less fat in white adipose tissue (WAT), have lower plasma glucose and cholesterol concentrations and enhanced whole-body energy expenditure, due mostly to increased resting energy expenditure, with no increase in physical activity. An increase in respiratory exchange ratio during the dark phase, the period in which the mice are active, indicates a switch from fat to carbohydrate oxidation. In comparison with WT mice, the rate of fatty acid oxidation in Fmo5(-/-) mice is higher in WAT, which would contribute to depletion of lipid stores in this tissue, and lower in skeletal muscle. Five proteins were down regulated in the liver of Fmo5(-/-) mice: aldolase B, ketohexokinase and cytosolic glycerol 3-phosphate dehydrogenase (GPD1) are involved in glucose or fructose metabolism and GPD1 also in production of glycerol 3-phosphate, a precursor of triglyceride biosynthesis; HMG-CoA synthase 1 is involved in cholesterol biosynthesis; and malic enzyme 1 catalyzes the oxidative decarboxylation of malate to pyruvate, in the process producing NADPH for use in lipid and cholesterol biosynthesis. Down regulation of these proteins provides a potential explanation for the reduced fat deposits and lower plasma cholesterol characteristic of Fmo5(-/-) mice. Our results indicate that disruption of the Fmo5 gene slows metabolic ageing via pleiotropic effects.


Subject(s)
Adipose Tissue, White/enzymology , Aging/genetics , Founder Effect , Gene Expression Regulation , Oxygenases/genetics , Aging/metabolism , Animals , Blood Glucose/metabolism , Body Weight/genetics , Cholesterol/blood , Energy Metabolism/genetics , Fructokinases/genetics , Fructokinases/metabolism , Fructose-Bisphosphate Aldolase/genetics , Fructose-Bisphosphate Aldolase/metabolism , Genotype , Glycerol-3-Phosphate Dehydrogenase (NAD+)/genetics , Glycerol-3-Phosphate Dehydrogenase (NAD+)/metabolism , Hydroxymethylglutaryl-CoA Synthase/genetics , Hydroxymethylglutaryl-CoA Synthase/metabolism , Lipid Metabolism/genetics , Liver/enzymology , Malate Dehydrogenase/genetics , Malate Dehydrogenase/metabolism , Male , Mice , Mice, Knockout , Muscle, Skeletal/enzymology , Oxidation-Reduction , Oxygenases/deficiency , Phenotype
4.
J Lipid Res ; 56(1): 22-37, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25378658

ABSTRACT

We performed silencing and overexpression studies of flavin containing monooxygenase (FMO) 3 in hyperlipidemic mouse models to examine its effects on trimethylamine N-oxide (TMAO) levels and atherosclerosis. Knockdown of hepatic FMO3 in LDL receptor knockout mice using an antisense oligonucleotide resulted in decreased circulating TMAO levels and atherosclerosis. Surprisingly, we also observed significant decreases in hepatic lipids and in levels of plasma lipids, ketone bodies, glucose, and insulin. FMO3 overexpression in transgenic mice, on the other hand, increased hepatic and plasma lipids. Global gene expression analyses suggested that these effects of FMO3 on lipogenesis and gluconeogenesis may be mediated through the PPARα and Kruppel-like factor 15 pathways. In vivo and in vitro results were consistent with the concept that the effects were mediated directly by FMO3 rather than trimethylamine/TMAO; in particular, overexpression of FMO3 in the human hepatoma cell line, Hep3B, resulted in significantly increased glucose secretion and lipogenesis. Our results indicate a major role for FMO3 in modulating glucose and lipid homeostasis in vivo, and they suggest that pharmacologic inhibition of FMO3 to reduce TMAO levels would be confounded by metabolic interactions.


Subject(s)
Atherosclerosis/enzymology , Glucose/metabolism , Lipid Metabolism , Oxygenases/metabolism , Animals , Bile Acids and Salts/metabolism , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Diet, Western , Feces/chemistry , Female , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , Gene Knockout Techniques , Glucose/biosynthesis , Homeostasis , Humans , Insulin/blood , Intestinal Mucosa/metabolism , Kruppel-Like Transcription Factors , Lipogenesis , Lipoproteins/blood , Liver/metabolism , Methylamines/metabolism , Mice , Oxygenases/deficiency , Oxygenases/genetics , PPAR alpha/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Transcription Factors/metabolism
5.
Dermatol Online J ; 20(1): 21260, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24456963

ABSTRACT

Trimethylaminuria is a rare, autosomal recessive, metabolic disorder that results in accumulation of trimethylamine (TMA), which smells like rotten fish. The chemical is excreted in sweat and urine owing to a deficiency in the enzyme flavin monooxygenase 3 (FMO3). We report a case of trimethylaminuria in a 12-year-old girl. The patient failed treatment with diet and hygiene modification, but achieved symptomatic improvement after a four-month course of metronidazole.


Subject(s)
Metabolism, Inborn Errors/diagnosis , Methylamines/urine , Anxiety Disorders/complications , Child , Female , Humans , Hygiene , Hyperhidrosis/complications , Metabolism, Inborn Errors/diet therapy , Metabolism, Inborn Errors/drug therapy , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/urine , Metronidazole/therapeutic use , Oxygenases/deficiency , Oxygenases/genetics
7.
Pharmacogenet Genomics ; 19(4): 289-99, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19262426

ABSTRACT

OBJECTIVES: Many drugs are the subject of multipathway oxidative metabolism catalyzed by one or more cytochromes P450 or flavin-containing monooxygenases (FMOs). This complicates assessment of the role of individual enzymes in metabolizing the drug and, hence, in understanding its pharmacogenetics. To define the role of FMOs in drug metabolism, we produced FMO-deficient mice. METHODS: An Fmo1(-/-), Fmo2(-/-), Fmo4(-/-) mouse line was produced by using chromosomal engineering and Cre-loxP technology. To assess the utility of the mutant mouse line, it was used to investigate the role of FMO in the metabolism of and response to the antidepressant imipramine, which has four major metabolites, three produced by cytochromes P450 and one, imipramine N-oxide, solely by FMO1. RESULTS: On treatment with imipramine, wild-type mice became sedated and produced imipramine N-oxide in the brain and other tissues. In contrast, knockout mice did not produce imipramine N-oxide, but showed exaggerated pharmacological behavioural responses, such as tremor and body spasm, and had a higher concentration of the parent compound imipramine in the serum and kidney and there was an increase in desipramine in the brain. CONCLUSION: The absence of FMO1-mediated N-oxidation of imipramine results in enhanced central nervous system effects of the drug. The results provide insights into the metabolism of imipramine in the brain and may explain the basis of the adverse reactions to the drug seen in some patients. The knockout mouse line will provide a valuable resource for defining the role of FMO1 in the metabolism of drugs and other foreign chemicals.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , Behavior, Animal/drug effects , Embryonic Stem Cells/cytology , Gene Deletion , Imipramine/pharmacology , Oxygenases/genetics , Animals , Antidepressive Agents, Tricyclic/metabolism , Cells, Cultured , Chromosomes/genetics , Female , Heterozygote , Homozygote , Imipramine/analogs & derivatives , Imipramine/metabolism , Integrases/genetics , Male , Mice , Oxygenases/deficiency , Spasm/chemically induced , Transfection , Tremor/chemically induced
8.
Biochemistry ; 48(4): 792-9, 2009 Feb 03.
Article in English | MEDLINE | ID: mdl-19138101

ABSTRACT

Nitrosative stress is induced by pathophysiological levels of nitric oxide (NO) and S-nitrosothiols (e.g., S-nitrosoglutathione, GSNO) and arises, at least in significant part, from the nitrosylation of critical protein Cys thiols (S-nitrosylation) and metallocofactors. However, the mechanisms by which NO and GSNO mediate nitrosative stress are not well understood. Using yeast Saccharomyces cerevisiae strains lacking NO- and/or GSNO-consuming enzymes (flavohemoglobin and GSNO reductase, respectively), we measured the individual and combined effects of NO and GSNO on both cell growth and the formation of protein-bound NO species. Our results suggest an intracellular equilibrium between NO and GSNO, dependent in part on cell-catalyzed release of NO from GSNO (i.e., "SNO-lyase" activity). However, whereas NO induces multiple types of protein-based modifications, levels of which correlate with inhibition of cell growth, GSNO mainly affects protein S-nitrosylation, and the relationship between S-nitrosylation and nitrosative stress is more complex. These data support the idea of multiple classes of protein-SNO, likely reflected in divergent routes of synthesis and degradation. Indeed, a significant fraction of protein S-nitrosylation by NO occurs in the absence of O(2), which is commonly assumed to drive this reaction but instead is apparently dependent in substantial part upon protein-bound transition metals. Additionally, our findings suggest that nitrosative stress is mediated principally via the S-nitrosylation of a subset of protein targets, which include protein SNOs that are stable to cellular glutathione (and thus are not metabolized by GSNO reductase). Collectively, these results provide new evidence for the mechanisms through which NO and GSNO mediate nitrosative stress as well as the cellular pathways of protein S-nitrosylation and denitrosylation involving metalloproteins, SNO lyase(s) and GSNO reductase.


Subject(s)
Aldehyde Oxidoreductases/chemistry , Nitric Oxide/chemistry , Nitric Oxide/genetics , Oxidative Stress/genetics , S-Nitrosothiols/chemistry , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Aldehyde Oxidoreductases/deficiency , Aldehyde Oxidoreductases/genetics , Cysteine/chemistry , Dioxygenases , Glutathione Reductase/chemistry , Glutathione Reductase/deficiency , Glutathione Reductase/genetics , Hemeproteins/deficiency , Hemeproteins/genetics , Hemeproteins/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide/deficiency , Nitrosation/genetics , Oxygenases/deficiency , Oxygenases/genetics , S-Nitrosoglutathione/chemistry , S-Nitrosoglutathione/metabolism , S-Nitrosothiols/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sulfhydryl Compounds/chemistry
9.
J Biol Chem ; 282(46): 33553-33561, 2007 Nov 16.
Article in English | MEDLINE | ID: mdl-17855355

ABSTRACT

Carotenoids are currently investigated regarding their potential to lower the risk of chronic disease and to combat vitamin A deficiency in humans. These plant-derived compounds must be cleaved and metabolically converted by intrinsic carotenoid oxygenases to support the panoply of vitamin A-dependent physiological processes. Two different carotenoid-cleaving enzymes were identified in mammals, the classical carotenoid-15,15'-oxygenase (CMO1) and a putative carotenoid-9',10'-oxygenase (CMO2). To analyze the role of CMO1 in mammalian physiology, here we disrupted the corresponding gene by targeted homologous recombination in mice. On a diet providing beta-carotene as major vitamin A precursor, vitamin A levels fell dramatically in several tissues examined. Instead, this mouse mutant accumulated the provitamin in large quantities (e.g. as seen by an orange coloring of adipose tissues). Besides impairments in beta-carotene metabolism, CMO1 deficiency more generally interfered with lipid homeostasis. Even on a vitamin A-sufficient chow, CMO1(-/-) mice developed a fatty liver and displayed altered serum lipid levels with elevated serum unesterified fatty acids. Additionally, this mouse mutant was more susceptible to high fat diet-induced impairments in fatty acid metabolism. Quantitative reverse transcription-PCR analysis revealed that the expression of peroxisome proliferator-activated receptor gamma-regulated marker genes related to adipogenesis was elevated in visceral adipose tissues. Thus, our study identifies CMO1 as the key enzyme for vitamin A production and provides evidence for a role of carotenoids as more general regulators of lipid metabolism.


Subject(s)
Oxygenases/chemistry , Oxygenases/physiology , Vitamin A/metabolism , Adipose Tissue/metabolism , Animals , Fatty Acids/metabolism , Glucose Tolerance Test , Homeostasis , Humans , Lipids/chemistry , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Genetic , Oxygenases/deficiency , PPAR gamma/metabolism , Recombination, Genetic , beta Carotene/metabolism
10.
Ann Bot ; 98(3): 565-71, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16790464

ABSTRACT

BACKGROUND AND AIMS: Glycinebetaine (GB), a quaternary ammonium compound, is a very effective compatible solute. In higher plants, GB is synthesized from choline (Cho) via betaine aldehyde (BA). The first and second steps in the biosynthesis of GB are catalysed by choline monooxygenase (CMO) and by betaine aldehyde dehydrogenase (BADH), respectively. Rice (Oryza sativa), which has two genes for BADH, does not accumulate GB because it lacks a functional gene for CMO. Rice plants accumulate GB in the presence of exogenously applied BA, which leads to the development of a significant tolerance to salt, cold and heat stress. The goal in this study was to evaluate and to discuss the effects of endogenously accumulated GB in rice. METHODS: Transgenic rice plants that overexpressed a gene for CMO from spinach (Spinacia oleracea) were produced by Agrobacterium-mediated transformation. After Southern and western blotting analysis, GB in rice leaves was quantified by (1)H-NMR spectroscopy and the tolerance of GB-accumulating plants to abiotic stress was investigated. KEY RESULTS: Transgenic plants that had a single copy of the transgene and expressed spinach CMO accumulated GB at the level of 0.29-0.43 micromol g(-1) d. wt and had enhanced tolerance to salt stress and temperature stress in the seedling stage. CONCLUSIONS: In the CMO-expressing rice plants, the localization of spinach CMO and of endogenous BADHs might be different and/or the catalytic activity of spinach CMO in rice plants might be lower than it is in spinach. These possibilities might explain the low levels of GB in the transgenic rice plants. It was concluded that CMO-expressing rice plants were not effective for accumulation of GB and improvement of productivity.


Subject(s)
Betaine/metabolism , Oryza/metabolism , Oxygenases/metabolism , Sodium Chloride/metabolism , Betaine/chemistry , Blotting, Southern , Blotting, Western , Genes, Plant , Magnetic Resonance Spectroscopy , Oryza/genetics , Oxygenases/deficiency , Oxygenases/genetics , Plant Leaves/genetics , Plants, Genetically Modified , Rhizobium/genetics , Spinacia oleracea/enzymology , Spinacia oleracea/genetics , Temperature , Transformation, Genetic
11.
J Inherit Metab Dis ; 29(1): 162-72, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16601883

ABSTRACT

Persistent trimethylaminuria in children is caused by autosomal recessively inherited impairment of hepatic trimethylamine (TMA) oxidation due to deficiency of flavin monooxygenase 3 (FMO3) secondary to mutations in the FMO3 gene. Trimethylaminuria or 'fish odour syndrome' is due to excessive excretion into body fluids and breath of TMA derived from the enterobacterial metabolism of dietary precursors. The disorder is present from birth but becomes apparent as foods containing high amounts of choline or of trimethylamine N-oxide (TMAO) from marine (sea or saltwater) fish are introduced into the diet. In our experience, trimethylaminuria (FMO3 deficiency) in children is rare. We have compared the dynamics and diagnostic efficacy of choline loading with marine fish meals in six children with trimethylaminuria. Loading with a marine fish meal provides a simple and acceptable method for confirmation of diagnosis of suspected trimethylaminuria in children, with the effects being cleared more quickly than with a choline load test. However, oral loading with choline bitartrate allows estimation of residual oxidative capacity in vivo and is a useful adjunct to molecular studies. Patients homozygous for the 'common' P153L mutation in the FMO3 gene showed virtual complete lack of residual TMA N-oxidative capacity, consistent with a nonfunctional or absent FMO3 enzyme, whereas a patient with the M82T mutation showed some residual oxidative capacity. A patient compound heterozygous for two novel mutations, G193E and R483T, showed considerable residual N-oxidative capacity. A further patient, heterozygous for two novel sequence variations in the FMO3 gene, consistently showed malodour and elevated urinary TMA/TMAO ratios under basal conditions but a negative response to both choline and marine fish meal loading. Comparison of the effects of administration of antibiotics (metronidazole, amoxicillin, neomycin) on gut bacterial production of trimethylamine from choline showed they all reduced TMA production to a limited extent, with neomycin being most effective. 'Best-practice' diagnostic and treatment guidelines are summarized.


Subject(s)
Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/therapy , Oxygenases/deficiency , Adult , Case-Control Studies , Child , Child, Preschool , Choline/metabolism , Choline/pharmacology , DNA Mutational Analysis , Female , Fish Products , Humans , Infant , Male , Metabolism, Inborn Errors/genetics , Middle Aged , Mutation
12.
J Inherit Metab Dis ; 26(4): 415-6, 2003.
Article in English | MEDLINE | ID: mdl-12971432

ABSTRACT

Analysis of sweat collected from patients experiencing ichthyohidriosis, and from volunteers in whom this odour phenomenon had been artificially induced, showed that trimethylamine was the major causative factor.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Methylamines/metabolism , Odorants , Oxygenases/metabolism , Sweat/chemistry , Gas Chromatography-Mass Spectrometry , Humans , Methylamines/analysis , Oxygenases/deficiency
13.
Pol Merkur Lekarski ; 14(83): 444-6, 2003 May.
Article in Polish | MEDLINE | ID: mdl-12939823

ABSTRACT

Alkaptonuria is a rare metabolic condition caused by congenital homogentisate oxidase deficiency of recessive inheritance. Homogentisate polymers are accumulated and cause urine darkening, brown pigmentation of connective tissue, articular cartilage pathology. The authors present clinical picture, pathogenesis, diagnostic and therapeutic possibilities in patients with alkaptonuria. Two siblings with alkaptonuria are described.


Subject(s)
Alkaptonuria/diagnosis , Dioxygenases , Alkaptonuria/urine , Homogentisate 1,2-Dioxygenase , Humans , Infant , Male , Oxygenases/deficiency , Oxygenases/urine
15.
Mol Genet Metab ; 77(1-2): 136-42, 2002.
Article in English | MEDLINE | ID: mdl-12359141

ABSTRACT

In alkaptonuria, homogentisate 1,2-dioxygenase deficiency causes tissue accumulation of homogentisic acid (HGA), followed by signs and symptoms of ochronosis. These include massive urinary excretion of HGA, arthritis and joint destruction, pigmentation of cartilage and connective tissue, and cardiac valve deterioration. We describe a 46-year-old man with alkaptonuria and diabetic renal failure whose plasma HGA concentration was twice that of any other alkaptonuria patient, and whose ochronosis progressed much more rapidly than that of his two alkaptonuric siblings. After renal transplantation, the plasma HGA normalized, and the daily urinary excretion of HGA decreased by 2-3g. This case illustrates the critical role of renal tubular secretion in eliminating HGA from the body, and suggests that renal transplantation in a uremic patient not only restores HGA excretion, but may also provide homogentisate 1,2-dioxygenase activity for the metabolism of HGA.


Subject(s)
Alkaptonuria/complications , Alkaptonuria/surgery , Dioxygenases , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/surgery , Kidney Transplantation , Ochronosis/etiology , Adult , Alkaptonuria/enzymology , Alkaptonuria/genetics , Base Sequence , DNA/genetics , DNA Mutational Analysis , Diabetic Nephropathies/complications , Diabetic Nephropathies/surgery , Female , Homogentisate 1,2-Dioxygenase , Homogentisic Acid/metabolism , Humans , Kidney Transplantation/physiology , Male , Middle Aged , Mutation , Ochronosis/pathology , Oxygenases/deficiency , Oxygenases/genetics , Radiography , Spine/diagnostic imaging
16.
An Med Interna ; 19(11): 583-5, 2002 Nov.
Article in Spanish | MEDLINE | ID: mdl-12522896

ABSTRACT

Alkaptonuric ochronosis is rare disorder of tyrosin catabolism with an autosomal recessive trait. Alkaptonuric patients are deficient for homogentisate 1,2-dioxygenase. This enzymatic deficiency leads to the elimination of large amounts of homogentistic acid in the urine (Alkaptonuria) with accumulation of homogentistic acid oxidized pigment in the connective tissue (Ochronosis). The most common clinical features are dark brown discoloration of urine on exposure to air; ocular and cutaneous pigmentation; calcification of the intervertebral disc and cardiovascular ochronosis, especially calcification and stenosis of the aortic valve. The diagnosis is confirmed by detection of homogentistic acid in urine. We report a case of a 87 year old female which has all these clinical features mentioned above and pericardiac calcification, which had not been previously reported, to our knowledge.


Subject(s)
Calcinosis/diagnostic imaging , Dioxygenases , Heart Diseases/diagnosis , Intervertebral Disc/diagnostic imaging , Ochronosis/diagnosis , Pericardium/pathology , Spinal Diseases/diagnostic imaging , Aged , Aged, 80 and over , Alkaptonuria/diagnosis , Alkaptonuria/urine , Female , Homogentisate 1,2-Dioxygenase , Homogentisic Acid/urine , Humans , Ochronosis/urine , Oxygenases/deficiency , Radiography, Abdominal
18.
Ned Tijdschr Geneeskd ; 145(27): 1295, 2001 Jul 07.
Article in Dutch | MEDLINE | ID: mdl-11475020

ABSTRACT

In a 70-year-old woman, in whom ochronosis (alkaptonuria) was diagnosed at the age of 54, bluish discolouration of the cartilage of the ears was observed.


Subject(s)
Dioxygenases , Ear Cartilage/pathology , Homogentisic Acid/metabolism , Ochronosis/metabolism , Aged , Female , Genetic Predisposition to Disease , Homogentisate 1,2-Dioxygenase , Humans , Ochronosis/genetics , Ochronosis/pathology , Oxygenases/deficiency
19.
Am J Hematol ; 65(2): 149-53, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10996833

ABSTRACT

We postulated that a deficiency of flavin monooxygenase (FMO)-a ferrireductase component of cells-could produce sideroblastic anemia. FMO is an intracellular ferrireductase which may be responsible for the obligatory reduction of ferric to ferrous iron so that reduced iron can be incorporated into heme by ferrochelatase. Abnormalities of this mechanism could result in accumulation of excess ferric iron in mitochondria of erythroid cells to produce ringed sideroblasts and impair hemoglobin synthesis. To investigate this hypothesis we obtained blood from patients with sideroblastic anemia and normal subjects. Extracts of peripheral blood lymphocytes were used to measure ferrireduction by utilization of NADPH. Lymphoid precursors are reported to accumulate iron in mitochondria similarly to erythroid precursors. Utilization of lymphoid precursors avoided the need for bone marrow aspirations. We studied three patients with sideroblastic anemia. One patient and his asymptomatic daughter had a significant decrease in ferrireductase activity. They also had markedly diminished concentrations of FMO in lymphocyte protein extracts on Western blots. This was accompanied by increased concentration of mobilferrin in the extracts. These results suggest that abnormalities of FMO and mobilferrin may cause sideroblastic anemia and erythropoietic hemochromatosis in some patients.


Subject(s)
Anemia, Sideroblastic/etiology , Iron-Binding Proteins , Oxygenases/deficiency , Aged , Aged, 80 and over , Anemia, Sideroblastic/blood , Anemia, Sideroblastic/enzymology , Blotting, Western , Carrier Proteins/blood , Carrier Proteins/immunology , Family Health , Female , Genetic Linkage , Humans , Iron/blood , Iron/immunology , Lymphocytes/metabolism , Male , NADH, NADPH Oxidoreductases/blood , NADH, NADPH Oxidoreductases/deficiency , Oxygenases/blood , X Chromosome
SELECTION OF CITATIONS
SEARCH DETAIL