Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
Eukaryot Cell ; 11(4): 430-41, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22307976

ABSTRACT

Leishmania double transfectants (DTs) expressing the 2nd and 3rd enzymes in the heme biosynthetic pathway were previously reported to show neogenesis of uroporphyrin I (URO) when induced with delta-aminolevulinate (ALA), the product of the 1st enzyme in the pathway. The ensuing accumulation of URO in DT promastigotes rendered them light excitable to produce reactive oxygen species (ROS), resulting in their cytolysis. Evidence is presented showing that the DTs retained wild-type infectivity to their host cells and that the intraphagolysosomal/parasitophorous vacuolar (PV) DTs remained ALA inducible for uroporphyrinogenesis/photolysis. Exposure of DT-infected cells to ALA was noted by fluorescence microscopy to result in host-parasite differential porphyrinogenesis: porphyrin fluorescence emerged first in the host cells and then in the intra-PV amastigotes. DT-infected and control cells differed qualitatively and quantitatively in their porphyrin species, consistent with the expected multi- and monoporphyrinogenic specificities of the host cells and the DTs, respectively. After ALA removal, the neogenic porphyrins were rapidly lost from the host cells but persisted as URO in the intra-PV DTs. These DTs were thus extremely light sensitive and were lysed selectively by illumination under nonstringent conditions in the relatively ROS-resistant phagolysosomes. Photolysis of the intra-PV DTs returned the distribution of major histocompatibility complex (MHC) class II molecules and the global gene expression profiles of host cells to their preinfection patterns and, when transfected with ovalbumin, released this antigen for copresentation with MHC class I molecules. These Leishmania mutants thus have considerable potential as a novel model of a universal vaccine carrier for photodynamic immunotherapy/immunoprophylaxis.


Subject(s)
Aminolevulinic Acid/pharmacology , Leishmania/genetics , Phagocytes/parasitology , Phagosomes/parasitology , Photosensitizing Agents/pharmacology , Porphyrins/biosynthesis , Vaccination/methods , Animals , Antigen Presentation , Antigens, Protozoan/immunology , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/parasitology , Dendritic Cells/radiation effects , Gene Expression Profiling , Histocompatibility Antigens Class I/metabolism , Leishmania/immunology , Leishmania/radiation effects , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/parasitology , Macrophages, Peritoneal/radiation effects , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Organisms, Genetically Modified/immunology , Photolysis
2.
FEBS J ; 275(23): 5947-59, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19021769

ABSTRACT

Heme is synthesized in all cell types in aerobic organisms. Hydroxymethylbilane synthase (HMBS) and uroporphyrinogen III synthase (UROS) catalyze two consecutive reactions in the heme biosynthetic pathway, generating the first linear and the first cyclic tetrapyrroles, respectively. Each of the HMBS and UROS genes contains the two separate promoters that generate ubiquitous and erythroid-specific mRNAs. Despite the functional significance of HMBS and UROS, regulation of their gene expression remains to be investigated. Here, we showed that hypoxia (1% O(2)) decreased the expression of ubiquitous mRNAs for HMBS and UROS by three- and twofold, respectively, in human hepatic cells (HepG2 and Hep3B), whereas the expression of ubiquitous and erythroid HMBS and UROS mRNAs remained unchanged in erythroid cells (YN-1 and K562). Unexpectedly, hypoxia did not decrease the half-life of HMBS mRNA (8.4 h under normoxia versus 9.1 h under hypoxia) or UROS mRNA (9.0 versus 10.4 h) in hepatic cells. It is therefore unlikely that a change in mRNA stability is responsible for the hypoxia-mediated decrease in the expression levels of these mRNAs. Furthermore, expression levels of HMBS and UROS mRNAs were decreased under normoxia by treatment with deferoxamine or cobalt chloride in hepatic cells, while hypoxia-inducible factor 1alpha was accumulated. Thus, the decrease in the expression of ubiquitous HMBS and UROS mRNAs is associated with accumulation of hypoxia-inducible factor 1alpha protein. In conclusion, the expression of HMBS and UROS mRNAs may be coordinately regulated, which represents a newly identified mechanism that is important for heme homeostasis.


Subject(s)
Heme/biosynthesis , Hydroxymethylbilane Synthase/genetics , Tetrapyrroles/biosynthesis , Uroporphyrinogen III Synthetase/genetics , Blotting, Northern , Blotting, Western , Cell Hypoxia/physiology , Cell Line, Tumor , Cobalt/pharmacology , Deferoxamine/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Hydroxymethylbilane Synthase/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Jurkat Cells , K562 Cells , RNA Stability/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Uroporphyrinogen III Synthetase/metabolism
3.
Eukaryot Cell ; 7(7): 1146-57, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18487349

ABSTRACT

Inherent deficiencies of Leishmania in heme biosynthesis were genetically complemented for delta-aminolevulinate-inducible biosynthesis and accumulation of light-excitable uroporphyrin. The phototoxic flagellar immobilization and cytolysis phenotypes and porphyrin mobilization noted previously were further analyzed biochemically and cytologically to delineate the mechanism of phototoxicity and detoxification in this monoporphyric model. Under optimal conditions of induction for approximately 3 days, cells remained viable but became increasingly uroporphyric, peaking at > or =90% of the population by approximately day 2; thereafter, a small population of less porphyric or aporphyric cells emerged. On exposure to light, the flagella of porphyric cells were immobilized in milliseconds, and singlet oxygen became detectable in their lysates. Both photosensitive phenotypes increased proportionally with the cellular uroporphyric levels and were susceptible to inhibition by azide, but not by D-mannitol. Brief irradiation of the uroporphyric cells produced no appreciable protein degradation but inactivated cytosolic neomycin phosphotransferase and significantly bleached cytosolic green fluorescent protein, which was azide reversible. These cells were irreparably photodamaged, as indicated by their subsequent loss of membrane permeability and viability. This is the first in situ demonstration that early inactivation of functional proteins by singlet oxygen initiates the cytolytic phototoxicity in uroporphyria. Detoxification appears to involve endocytic/exocytic mobilization of uroporphyrin from cytosol to "porphyrinosomes" for its eventual extracellular expulsion. This is proposed as the sole mechanism of detoxification, since it is attributable to the reversion of porphyric to aporphyric cells during uroporphyrinogenesis and repeated cycles of this event plus photolysis selected no resistant mutants, only aporphyric clones of the parental phenotypes. Further characterization of the transport system for uroporphyrin in this model is expected to benefit not only our understanding of the cellular mechanism for disposal of toxic soluble wastes but also potentially the effective management of human uroporphyria and the use of uroporphyric Leishmania for vaccine/drug delivery.


Subject(s)
Aminolevulinic Acid/pharmacology , Cytosol/metabolism , Leishmania/metabolism , Proteins/metabolism , Singlet Oxygen/metabolism , Uroporphyrins/metabolism , Aminolevulinic Acid/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Azides/pharmacology , Biological Transport , Cell Membrane Permeability/radiation effects , Cell Survival/radiation effects , Flagella/drug effects , Flagella/metabolism , Humans , Leishmania/drug effects , Leishmania/genetics , Leishmania/radiation effects , Light , Models, Animal , Phenotype , Photolysis , Porphyrias/chemically induced , Porphyrias/metabolism , Porphyrias/therapy , Transport Vesicles/metabolism , Uroporphyrins/genetics , Uroporphyrins/pharmacokinetics
4.
Exp Parasitol ; 118(4): 629-36, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18164705

ABSTRACT

To further develop the Leishmania model for porphyria based on their deficiencies in heme biosynthesis, three Old World species were doubly transfected as before for Leishmania amazonensis with cDNAs, encoding the 2nd and 3rd enzymes in the pathway. Expression of the transgenes was verified immunologically at the protein level and functionally by uroporphyrin neogenesis that occurs only after exposure of the double-transfectants to delta-aminolevulinate. All species examined were equally deficient in heme biosynthesis, as indicated by the accumulation of uroporphyrin as the sole porphyrin and the production of coproporphyrin upon further transfection of one representative species with the downstream gene. The results obtained thus demonstrate that at least the first five enzymes for heme biosynthesis are absent in all species examined, rendering their transfectants inducible with aminolevulinate to accumulate porphyrins and thus useful as cellular models for human porphyrias.


Subject(s)
Aminolevulinic Acid/pharmacology , Leishmania/metabolism , Porphyrins/biosynthesis , Animals , Blotting, Western , Chromatography, Thin Layer , DNA, Complementary/genetics , Heme/biosynthesis , Heme/genetics , Hydroxymethylbilane Synthase/metabolism , Leishmania/enzymology , Leishmania/genetics , Porphobilinogen Synthase/metabolism , Porphyrins/analysis , Porphyrins/genetics , Spectrometry, Fluorescence , Transfection , Transgenes/physiology , Uroporphyrins/biosynthesis , Uroporphyrins/genetics
6.
Br J Haematol ; 135(3): 281-92, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16956347

ABSTRACT

Recent advances in the molecular understanding of the porphyrias now offer specific diagnosis and precise definition of the types of genetic mutations involved in the disease. Molecular diagnostic testing is powerful and very useful in kindred evaluation and genetic counselling when a disease-responsible mutation has been identified in the family. It is also the only way to properly screen asymptomatic gene carriers, facilitating correct treatment and appropriate genetic counselling of family members at risk. However, it should be noted that DNA-based testing is for the diagnosis of the gene carrier status, but not for the diagnosis of clinical syndrome or severity of the disease, e.g. an acute attack. For the diagnosis of clinically expressed porphyrias, a logical stepwise approach including the analysis of porphyrins and their precursors should not be underestimated, as it is still very useful, and is often the best from the cost-effective point of view.


Subject(s)
Porphyrias/diagnosis , Acute Disease , Chronic Disease , Coproporphyria, Hereditary/diagnosis , Coproporphyria, Hereditary/therapy , Heme/biosynthesis , Humans , Porphobilinogen Synthase/metabolism , Porphyria Cutanea Tarda/diagnosis , Porphyria Cutanea Tarda/therapy , Porphyria, Erythropoietic/diagnosis , Porphyria, Erythropoietic/therapy , Porphyria, Hepatoerythropoietic/diagnosis , Porphyria, Hepatoerythropoietic/therapy , Porphyria, Variegate/diagnosis , Porphyria, Variegate/therapy , Porphyrias/classification , Porphyrias/therapy , Porphyrias, Hepatic/diagnosis , Porphyrias, Hepatic/therapy , Protoporphyria, Erythropoietic/diagnosis , Protoporphyria, Erythropoietic/therapy
7.
Int J Mol Med ; 18(4): 537-46, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16964402

ABSTRACT

Heme oxygenase (HO)-1, the rate-limiting enzyme in heme catabolism, can be induced in response to various oxidative stimuli, and its induction is thought to be critical in the cellular defense against oxidative tissue injuries. Carbon tetrachloride (CCl(4)) treatment of rats causes lipid peroxidation of cell membranes and produces massive hepatic injury. We previously demonstrated that HO-1 induction following CCl(4) treatment is an essential part of the cellular defense against the CCl(4)-inducible toxic changes. As recombinant human interleukin-11 (rhIL-11) has been shown to induce HO-1 in cultured hepatoma cells, we examined the effect of rhIL-11 in vivo in rats on the CCl(4)-induced tissue injury. rhIL-11 treatment of animals by itself markedly induced HO-1 mRNA and its functional protein principally in the liver. rhIL-11 treatment (150 microg/kg) of the CCl(4)-administered (1 ml/kg) animals led to a further increase in HO-1 mRNA, while it markedly suppressed CCl(4)-induced serum alanine transaminase, hepatic malondialdehyde formation, tumor necrosis factor-alpha mRNA, nitric oxide synthase mRNA, nuclear factor-kappaB DNA-binding activity, as well as inflammatory changes of hepatocytes. In contrast, inhibition of HO activity by tin-mesoporphyrin, a competitive specific inhibitor of HO, entirely abolished the cytoprotective effect of rhIL-11. These findings thus demonstrate that rhIL-11 confers significant protection against CCl(4)-induced hepatic injury by virtue of its liver-specific HO-1 induction.


Subject(s)
Heme Oxygenase-1/genetics , Interleukin-11/pharmacology , Liver Diseases/prevention & control , Liver/enzymology , Animals , Blotting, Northern , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury , Dose-Response Relationship, Drug , Gene Expression/drug effects , Humans , Injections, Intravenous , Interleukin-11/administration & dosage , Liver/drug effects , Liver/metabolism , Liver Diseases/enzymology , Male , Malondialdehyde/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/genetics
8.
Br J Haematol ; 132(2): 237-43, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16398658

ABSTRACT

Summary A Caucasian male had symptoms of acute porphyria, with increases in urinary delta-aminolaevulinic acid (ALA), porphobilinogen (PBG) and coproporphyrin that were consistent with hereditary coproporphyria (HCP). However, a greater than expected increase in ALA, compared with PBG, and a substantial increase in erythrocyte zinc protoporphyrin, suggested additional ALA dehydratase (ALAD) deficiency. Nucleotide sequence analysis of coproporphyrinogen oxidase (CPO) cDNA of the patient, but not of the parents, revealed a novel nucleotide transition G835-->C, resulting in an amino acid change, G279R. The mutant CPO protein expressed in Escherichia coli was unstable, and produced about 5% of activity compared with the wild-type CPO. Erythrocyte ALAD activity was 32% of normal in the proband. Nucleotide sequence analysis of cloned ALAD cDNAs from the patient revealed a C36-->G base transition (F12L amino acid change). The F12L ALAD mutation, which was found in the mother and a brother, was previously described, and is known to lack any enzyme activity. This patient thus represents the first case of porphyria where both CPO and ALAD deficiencies were demonstrated at the molecular level.


Subject(s)
Coproporphyria, Hereditary/genetics , Coproporphyrinogen Oxidase/genetics , Porphobilinogen Synthase/genetics , Adult , Coproporphyria, Hereditary/diagnosis , Coproporphyrinogen Oxidase/metabolism , DNA Mutational Analysis/methods , DNA, Complementary/genetics , Erythrocytes/enzymology , Female , Humans , Male , Models, Molecular , Pedigree , Porphobilinogen Synthase/deficiency
9.
Mol Genet Metab ; 87(4): 329-36, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16343966

ABSTRACT

The molecular basis of the enzymatic defect responsible for delta-aminolevulinate dehydratase (ALAD) porphyria (ADP) was investigated in a 14-year-old male who presented clinical and laboratory findings typical of ADP. Nucleotide sequence analysis of ALAD cDNAs from the proband revealed two novel mutations, a 265G to A base transition (C1) and a 394C to T base transition (C2), resulting in amino acid substitutions, Glu89Lys and Cys132Arg, respectively. Both mutations were present within exon 5 of the ALAD gene, and appeared to influence the binding of zinc to the enzyme which is essential for enzyme activity. It was found that the C1 mutation was inherited from his father, while the C2 mutation was from his mother. Expression of these mutant ALAD cDNAs in Chinese hamster ovary cells produced normal ALAD mRNA levels, but markedly decreased ALAD protein and enzyme activity. These results suggest that the combination of the two aberrant ALADs with little enzyme activity accounts for the markedly decreased ALAD activity observed in the proband. This case represents the molecular analysis of the ALAD gene defects in the first case of ADP identified in North America, who is a compound heterozygote for two novel ALAD gene defects.


Subject(s)
Porphobilinogen Synthase/genetics , Porphyrias/genetics , Adolescent , Animals , CHO Cells , Catalytic Domain , Cricetinae , Cricetulus , Erythrocytes/drug effects , Erythrocytes/enzymology , Humans , In Vitro Techniques , Lead/pharmacology , Male , Mitochondria/enzymology , Mutation , Pedigree , Porphobilinogen Synthase/chemistry , Porphyrias/enzymology , Sequence Analysis, DNA , Zinc/metabolism , Zinc/pharmacology
10.
Eur J Haematol ; 76(1): 33-41, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16343269

ABSTRACT

Mutations of the erythroid-specific 5-aminolaevulinate synthase (ALAS2) gene are known to be responsible for X-linked sideroblastic anaemia (XLSA). An amino acid (AA) substitution for arginine at the 452 AA position of the ALAS2 protein is the most frequent mutation, which has been found in approximately one-quarter of patients with XLSA. Despite its high frequency, there has been no report on the enzymatic activity of Arg452 mutant proteins. In this study, we examined enzymatic activity in vitro of two Arg452 mutants, Arg452Cys and Arg452His, which were found in two new pedigrees of XLSA. While these mutations must be responsible for the clinical phenotype of XLSA in patients, the enzymatic activity and stability of these mutant proteins studied in vitro are indistinguishable from those of the wild type protein. These findings suggest that the Arg452 mutation of the ALAS2 gene by itself does not decrease the enzymatic activity or the stability in vitro, and that there may be an additional factor(s) in the bone marrow, which ensures the full ALAS2 activity in vivo.


Subject(s)
5-Aminolevulinate Synthetase/genetics , Amino Acid Substitution , Anemia, Sideroblastic/enzymology , Erythrocytes/enzymology , Genetic Diseases, X-Linked/enzymology , Point Mutation , 5-Aminolevulinate Synthetase/metabolism , Anemia, Sideroblastic/genetics , Arginine/genetics , Bone Marrow/enzymology , Genetic Diseases, X-Linked/genetics , Genetic Linkage , Organ Specificity/genetics , Pedigree , Phenotype
11.
Biochem Biophys Res Commun ; 340(1): 105-10, 2006 Feb 03.
Article in English | MEDLINE | ID: mdl-16356476

ABSTRACT

To identify erythroid-specific heme-regulated genes, we performed differential expression analysis between wild-type and heme-deficient erythroblasts, which had been prepared from wild-type and erythroid-specific delta-aminolevulinate synthase-null mouse ES cells, respectively. Among 8737 clones on cDNA array, 40 cDNA clones, including 34 unknown ESTs, were first selected by their high expression profiles in wild-type erythroblasts, and evaluated further for their erythroid-lineage specificity, expression in hematopoietic tissues in vivo, and heme-dependent expression, which yielded 11, 4, and 4 genes, respectively. Because of the selection strategy employed, the final 4 were considered as the newly identified erythroid-specific heme-regulated genes. These 4 genes were uncoupling protein 2, nucleolar spindle-associated protein, cellular nucleic acid-binding protein, and a novel acetyltransferase-like protein. These findings thus suggest that heme may regulate a wide variety of hitherto unrecognized genes, and further analysis of these genes may clarify their role in erythroid cell differentiation.


Subject(s)
5-Aminolevulinate Synthetase/metabolism , Erythroblasts/metabolism , Gene Expression Regulation, Developmental/physiology , Heme/metabolism , 5-Aminolevulinate Synthetase/deficiency , Animals , Cells, Cultured , Gene Expression Profiling , Mice
12.
Contrib Nephrol ; 148: 70-85, 2005.
Article in English | MEDLINE | ID: mdl-15912028

ABSTRACT

Following ischemia, superoxide is produced during the reperfusion phase in various organs. In renal pathophysiology, excess of free heme, which is released from hemeproteins under these conditions, catalyzes the formation of further reactive oxygen species to accelerate cellular injuries. There is accumulating evidence suggesting that transcriptional activation of heme oxygenase (HO)-1, the rate-limiting enzyme in heme degradation as well as the 32-kDa heat shock protein, participates in the defense against oxidative tissue injuries. Ischemia followed by reperfusion of the rat kidney accompanies significant induction of HO-1 mRNA, protein and enzyme activity, which is in part mediated through a rapid and transient increase in microsomal heme concentration. Inhibition of HO activity by tin mesoporphyrin results in a sustained and enhanced increase in microsomal heme content, and significantly exacerbates renal function. In contrast, SnCl2 treatment, which specifically induces HO-1 mRNA and protein in the proximal tubular epithelial cells, prevents the ischemia-reperfusion-mediated increase in microsomal heme concentration, and ameliorates the ischemic renal injury. In addition to these findings, recent evidence on the role of HO-1 in the kidney pathophysiology is summarized, with a particular emphasis on its protective role in the ischemic acute renal failure.


Subject(s)
Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Cytoprotection/physiology , Heme Oxygenase (Decyclizing)/metabolism , Animals , Heme Oxygenase-1 , Humans , Membrane Proteins
13.
Crit Care Med ; 33(2): 381-90, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15699843

ABSTRACT

OBJECTIVE: The aim of the present study was to investigate whether glutamine pretreatment improves intestinal injury in rats with endotoxemia by its heme oxygenase-1 induction in the lower intestinal tract. DESIGN: Randomized, blinded, controlled animal study. SETTING: University-based animal research facility. SUBJECTS: Sprague-Dawley male rats, weighing 220-250 g (n = 201). INTERVENTIONS: Rats were treated with glutamine (0.75 g/kg) dissolved in lactated Ringer's solution via the tail vein. Endotoxemia was induced in rats by intraperitoneal injection of lipopolysaccharide (10 mg/kg or 20 mg/kg for survival study). Lipopolysaccharide-treated animals were pretreated with glutamine or lactated Ringer's solution 9 hrs before lipopolysaccharide treatment. Some of the glutamine-pretreated animals further received tin mesoporphyrin (1 micromol/kg), a specific inhibitor of heme oxygenase activity, 1 hr before lipopolysaccharide treatment. MEASUREMENTS AND MAIN RESULTS: Glutamine treatment markedly induced heme oxygenase-1 messenger RNA and protein in the mucosal epithelial cells as well as in the lamina propria cells in the ileum and the colon, whereas its expression in the duodenum and the jejunum was not influenced by the treatment. Glutamine treatment before lipopolysaccharide administration significantly ameliorated lipopolysaccharide-induced mucosal injury, inflammation, and apoptotic cell death in the ileum and the colon, as judged by significant decreases in tumor necrosis factor-alpha gene expression, histologic damage scores, and expression of activated caspase-3 and by an increase in gene expression of Bcl-2. In addition, glutamine treatment markedly decreased lipopolysaccharide-induced mortality. In contrast, treatment with tin mesoporphyrin abolished the beneficial effect of glutamine pretreatment. CONCLUSIONS: Glutamine pretreatment significantly ameliorated intestinal tissue injury of rats following lipopolysaccharide treatment. The same treatment also improved the survival of animals from endotoxemia. The protective effect of glutamine is mediated by its lower intestine-specific induction of heme oxygenase-1, since its inhibition by tin mesoporphyrin completely abolished the beneficial effect of glutamine.


Subject(s)
Endotoxemia/enzymology , Glutamine/pharmacology , Heme Oxygenase (Decyclizing)/biosynthesis , Intestines/enzymology , Animals , Apoptosis , Colon/metabolism , Colon/pathology , Endotoxemia/pathology , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Escherichia coli , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase-1 , Ileum/metabolism , Ileum/pathology , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Intestines/pathology , Lipopolysaccharides/toxicity , Male , Metalloporphyrins/pharmacology , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
14.
Antioxid Redox Signal ; 6(5): 819-24, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15345141

ABSTRACT

A large amount of hemoglobin is degraded daily to heme and globin and is replenished by biosynthesis in the bone marrow erythroblasts. "Free heme" can be dissociated from apohemoglobin in vitro and, conversely, native hemoglobin can be renatured from them. Then why does heme need to be degraded to iron, biliverdin IXalpha, and carbon monoxide in vivo? Free heme, i.e., a protein-unbound heme, exists in cells at a very minute concentration and exerts regulatory functions such as the repression of nonspecific delta-aminolevulinate synthase expression and the induction of microsomal heme oxygenase-1 (HO-1). The latter gene expression occurs by way of free heme-mediated derepression of Bach1, a mammalian heme-responsive transcription factor that suppresses the activation of the HO-1 gene. All these events occur at free heme concentrations below 1 microM. In contrast, free heme concentration greater than 1 microM can be toxic because it catalyzes the production of reactive oxygen species. To cope with this problem, the body is equipped with various defense mechanisms against high free heme concentrations. HO is one of the major players in these mechanisms, and it catabolizes free heme to iron, biliverdin IXalpha, and carbon monoxide. These three metabolites of heme by HO reactions have additional important functions and are involved in various critical cellular events. Thus, the breakdown of heme to smaller elements has its own significance in essential cellular metabolism.


Subject(s)
Biliverdine/metabolism , Carbon Monoxide/metabolism , Heme/metabolism , Iron/metabolism , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Hemoglobins/chemistry , Hemoglobins/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism
15.
Antioxid Redox Signal ; 6(5): 867-77, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15345147

ABSTRACT

Oxidative stress, which has been implicated in the pathogenesis of ischemic renal injury, degrades heme proteins, such as cytochrome P450, and causes the elevation in the level of cellular free heme, which can catalyze the formation of reactive oxygen species. Heme oxygenase-1 (HO-1), the rate-limiting enzyme in heme degradation, is induced not only by its substrate, heme, but also by oxidative stress. In various models of oxidative tissue injuries, the induction of HO-1 confers protection on tissues from further damages by removing the prooxidant heme, or by virtue of the antioxidative, antiinflammatory, and/or antiapoptotic actions of one or more of the three products, i.e., carbon monoxide, biliverdin IXalpha, and iron by HO reaction. In contrast, the abrogation of HO-1 induction, or chemical inhibition of HO activity, abolishes its beneficial effect on the protection of tissues from oxidative damages. In this article, we review the protective role of HO-1 in renal ischemic injury, and its potential therapeutic applications. In addition, we summarize recent findings in the regulatory mechanism of ho-1 gene expression.


Subject(s)
Acute Kidney Injury/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Ischemia/metabolism , Kidney Diseases/metabolism , Protective Agents/metabolism , Acute Kidney Injury/pathology , Animals , Gene Expression Regulation, Enzymologic , Heme/metabolism , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1 , Humans , Ischemia/pathology , Kidney Diseases/pathology , Membrane Proteins , Oxidative Stress
16.
Cell ; 118(6): 757-66, 2004 Sep 17.
Article in English | MEDLINE | ID: mdl-15369674

ABSTRACT

FLVCR, a member of the major facilitator superfamily of transporter proteins, is the cell surface receptor for feline leukemia virus, subgroup C. Retroviral interference with FLVCR display results in a loss of erythroid progenitors (colony-forming units-erythroid, CFU-E) and severe anemia in cats. In this report, we demonstrate that human FLVCR exports cytoplasmic heme and hypothesize that human FLVCR is required on developing erythroid cells to protect them from heme toxicity. Inhibition of FLVCR in K562 cells decreases heme export, impairs their erythroid maturation and leads to apoptosis. FLVCR is upregulated on CFU-E, indicating that heme export is important in primary cells at this stage. Studies of FLVCR expression in cell lines suggest this exporter also impacts heme trafficking in intestine and liver. To our knowledge, this is the first description of a mammalian heme transporter.


Subject(s)
Erythrocytes/metabolism , Erythropoiesis/physiology , Hematopoietic Stem Cells/metabolism , Heme/metabolism , Membrane Transport Proteins/metabolism , Protein Transport/physiology , Receptors, Virus/metabolism , Apoptosis/physiology , Cell Differentiation/physiology , Cell Line , Gene Expression Regulation, Developmental/physiology , Humans , Intestinal Mucosa/metabolism , Liver/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/isolation & purification , RNA, Messenger/metabolism , Receptors, Virus/genetics , Receptors, Virus/isolation & purification , Up-Regulation/physiology
17.
Int J Hematol ; 79(5): 448-56, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15239394

ABSTRACT

The first case of porphyria on record in Japan was a patient with congenital erythropoietic porphyria (CEP) reported by Sato and Takahashi in 1920. Since then until the end of December 2002, 827 cases of porphyrias have been diagnosed from characteristic clinical and/or laboratory findings (463 males, 358 females, and 6 of unknown sex). Essentially all inherited porphyrias have been found in Japan, with the incidences and clinical symptoms generally being similar to those reported for other countries. The male-female ratio was approximately 1:1 for CEP, whereas it was higher for erythropoietic protoporphyria. In contrast, preponderances of female patients exist with acute hepatic porphyrias, such as acute intermittent porphyria (AIP), variegate porphyria (VP), and hereditary coproporphyria (HCP), and with undefined acute porphyria. Although porphyria cutanea tarda (PCT) is believed to be increasing recently in women in other countries because of smoking and the use of contraceptives, it is still by far more prominent in males in Japan than in females. The recent increasing contribution of hepatitis C virus infection to PCT in Japan has also been recognized. but there have been no PCT cases in Japan with HFE gene mutations. Familial occurrence and consanguinity were high for CEP, as expected; however, significant consanguinity was also noted in families where CEP, AIP, HCP, VP, or PCT occurred as a single isolated case without a family history of disease. This survey also revealed that as many as 71% of acute hepatic porphyria cases were initially diagnosed as nonporphyria and later revised or corrected to porphyria, indicating the difficulty of diagnosing porphyria in the absence of specific laboratory testing for porphyrins and their precursors in urine, stool, plasma, and erythrocyte samples.


Subject(s)
Porphyrias/pathology , Adolescent , Adult , Aged , Child , Child, Preschool , Consanguinity , DNA Mutational Analysis , Data Collection , Female , History, 20th Century , Humans , Incidence , Infant , Infant, Newborn , Japan , Male , Middle Aged , Pedigree , Porphyrias/genetics , Porphyrias/history , Porphyrias, Hepatic , Sex Factors
18.
Shock ; 21(2): 134-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14752286

ABSTRACT

The liver is one of the major target organs affected in sepsis, and its failure always results in critical consequences. It has been reported that recombinant human interleukin 11 (rhIL-11), a pleiotropic cytokine, may be useful in the treatment of sepsis. However, the effect of IL-11 specifically on the hepatic failure in sepsis has not been evaluated. In the present study, we examined the effect of rhIL-11 on the hepatic injury in a rat endotoxemia model. Acute endotoxemia was induced in male Sprague-Dawley rats by intraperitoneal injection (i.p.) of bacterial lipopolysaccharide (LPS, 20 mg/kg). Immediately after injection of LPS, rats were treated with rhIL-11 (150 microg/kg, i.p.) or the vehicle. LPS treatment induced severe hepatic injury as revealed by marked increases in serum alanine transaminase (ALT) and aspartate transaminase (AST) activities, extensive hepatocyte necrosis, tumor necrosis factor-alpha (TNF-alpha) mRNA, inducible nitric oxide synthase (iNOS) mRNA, and DNA-binding activity of nuclear factor-kappaB (NF-kappaB). In contrast, rhIL-11 treatment significantly ameliorated the LPS-induced hepatic injury, as judged by marked improvement in all these indices. In addition, rhIL-11 treatment markedly decreased LPS-induced mortality. These results indicate that rhIL-11 plays a significant protective role in LPS-induced hepatic injury in acute endotoxemia.


Subject(s)
Endotoxemia/metabolism , Interleukin-11/physiology , Liver/injuries , Liver/microbiology , Acute Disease , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Blotting, Northern , DNA, Complementary/metabolism , Gene Expression Regulation , Hepatocytes/pathology , Interleukin-11/therapeutic use , Lipopolysaccharides/metabolism , Liver/immunology , Male , NF-kappa B/metabolism , Necrosis , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Protein Binding , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/therapeutic use , Sepsis/drug therapy , Time Factors , Tumor Necrosis Factor-alpha/metabolism
19.
Br J Haematol ; 121(4): 657-61, 2003 May.
Article in English | MEDLINE | ID: mdl-12752110

ABSTRACT

Hydroxyurea (HU), an inhibitor of DNA synthesis, can also induce haemoglobinization in certain erythroid cell lines. In this study, we report that intracellular peroxides levels were increased in HU-treated murine erythroleukaemia (MEL) cells and that l-acetyl-N-cysteine (LNAC), a potent reducing reagent, had a significant inhibitory effect on the HU-mediated induction of beta-globin, delta-aminolaevulinate synthase mRNA expression and haemoglobinization of MEL cells. In contrast, the addition of LNAC to dimethyl sulphoxide (DMSO)-treated MEL cells had a much smaller effect on the number of haemoglobinized cells. These findings suggest that oxidative stress is involved in HU-mediated induction of erythroid differentiation and that HU induces MEL cell differentiation by a mechanism different to that involved in DMSO-mediated differentiation. Our findings also suggest that the induction of MEL cell differentiation by HU does not involve RAS-MAP (mitogen-activated protein) kinase signalling.


Subject(s)
Erythroid Precursor Cells/cytology , Hydroxyurea/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Oxidative Stress/physiology , Blotting, Western , Cell Differentiation , Erythroid Precursor Cells/drug effects , Globins/metabolism , Humans , K562 Cells , Mitogen-Activated Protein Kinase Kinases/metabolism , Oxygen/physiology , Peroxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL