Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biochemistry ; 40(33): 9821-7, 2001 Aug 21.
Article in English | MEDLINE | ID: mdl-11502175

ABSTRACT

The terminal step in heme biosynthesis, the insertion of ferrous iron into protoporphyrin IX to form protoheme, is catalyzed by the enzyme ferrochelatase (EC 4.99.1.1). A number of highly conserved residues identified from the crystal structure of human ferrochelatase as being in the active site were examined by site-directed mutagenesis. The mutants Y123F, Y165F, Y191H, and R164L each had an increased K(m) for iron without an altered K(m) for porphyrin. The double mutant R164L/Y165F had a 6-fold increased K(m) for iron and a 10-fold decreased V(max). The double mutant Y123F/Y191F had low activity with an elevated K(m) for iron, and Y123F/Y165F had no measurable activity. The mutants H263A/C/N, D340N, E343Q, E343H, and E343K had no measurable enzyme activity, while E343D, E347Q, and H341C had decreased V(max)s without significant alteration of the K(m)s for either substrate. D340E had near-normal kinetic parameters, while D383A and H231A had increased K(m)s for iron. On the basis of these data and the crystal structure of human ferrochelatase, it is proposed that residues E343, H341, and D340 form a conduit from H263 in the active site to the protein exterior and function in proton extraction from the porphyrin macrocycle. The role of H263 as the porphyrin proton-accepting residue is central to catalysis since metalation only occurs in conjunction with proton abstraction. It is suggested that iron is transported from the exterior of the enzyme at D383/H231 via residues W227 and Y191 to the site of metalation at residues R164 and Y165 which are on the opposite side of the active site pocket from H263. This model should be general for mitochondrial membrane-associated eucaryotic ferrochelatases but may differ for bacterial ferrochelatases since the spatial orientation of the enzyme within prokaryotic cells may differ.


Subject(s)
Ferrochelatase/chemistry , Ferrochelatase/metabolism , Iron/chemistry , Protons , Amino Acid Sequence , Amino Acids/chemistry , Arginine/chemistry , Binding Sites , Cobalt/chemistry , Cobalt/metabolism , Histidine/chemistry , Humans , Intracellular Membranes/enzymology , Iron/metabolism , Kinetics , Mitochondria/enzymology , Models, Molecular , Molecular Sequence Data , Mutation , Porphyrins/chemistry , Porphyrins/metabolism , Protein Binding , Sequence Homology, Amino Acid
2.
Nat Struct Biol ; 8(2): 156-60, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11175906

ABSTRACT

Human ferrochelatase (E.C. 4.99.1.1) is a homodimeric (86 kDa) mitochondrial membrane-associated enzyme that catalyzes the insertion of ferrous iron into protoporphyrin to form heme. We have determined the 2.0 A structure from the single wavelength iron anomalous scattering signal. The enzyme contains two NO-sensitive and uniquely coordinated [2Fe-2S] clusters. Its membrane association is mediated in part by a 12-residue hydrophobic lip that also forms the entrance to the active site pocket. The positioning of highly conserved residues in the active site in conjunction with previous biochemical studies support a catalytic model that may have significance in explaining the enzymatic defects that lead to the human inherited disease erythropoietic protoporphyria.


Subject(s)
Ferrochelatase/chemistry , Ferrochelatase/metabolism , Heme/biosynthesis , Amino Acid Sequence , Binding Sites , Catalysis , Crystallography, X-Ray , Detergents/metabolism , Dimerization , Ferrochelatase/genetics , Humans , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Metals/metabolism , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Protein Structure, Secondary , Sequence Alignment
3.
J Biol Chem ; 275(49): 38135-8, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-11027676

ABSTRACT

The transferrin receptor (TfR) interacts with two proteins important for iron metabolism, transferrin (Tf) and HFE, the protein mutated in hereditary hemochromatosis. A second receptor for Tf, TfR2, was recently identified and found to be functional for iron uptake in transfected cells (Kawabata, H., Germain, R. S., Vuong, P. T., Nakamaki, T., Said, J. W., and Koeffler, H. P. (2000) J. Biol. Chem. 275, 16618-16625). TfR2 has a pattern of expression and regulation that is distinct from TfR, and mutations in TfR2 have been recognized as the cause of a non-HFE linked form of hemochromatosis (Camaschella, C., Roetto, A., Cali, A., De Gobbi, M., Garozzo, G., Carella, M., Majorano, N., Totaro, A., and Gasparini, P. (2000) Nat. Genet. 25, 14-15). To investigate the relationship between TfR, TfR2, Tf, and HFE, we performed a series of binding experiments using soluble forms of these proteins. We find no detectable binding between TfR2 and HFE by co-immunoprecipitation or using a surface plasmon resonance-based assay. The affinity of TfR2 for iron-loaded Tf was determined to be 27 nm, 25-fold lower than the affinity of TfR for Tf. These results imply that HFE regulates Tf-mediated iron uptake only from the classical TfR and that TfR2 does not compete for HFE binding in cells expressing both forms of TfR.


Subject(s)
HLA Antigens/chemistry , HLA Antigens/metabolism , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/metabolism , Membrane Proteins , Receptors, Transferrin/chemistry , Receptors, Transferrin/metabolism , Transferrin/chemistry , Transferrin/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Cell Line , Dimerization , Hemochromatosis/genetics , Hemochromatosis Protein , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
4.
Blood ; 96(3): 1113-8, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10910930

ABSTRACT

Hypotransferrinemic (Trf(hpx/hpx)) mice have a severe deficiency in serum transferrin (Trf) as the result of a spontaneous mutation linked to the murine Trf locus. They are born alive, but before weaning, die from severe anemia if they are not treated with exogenous Trf or red blood cell transfusions. We have determined the molecular basis of the hpx mutation. It results from a single point mutation, which alters an invariable nucleotide in the splice donor site after exon 16 of the Trf gene. No normal Trf messenger RNA (mRNA) is made from the hpx allele. A small amount of mRNA results from the usage of cryptic splice sites within exon 16. The predominant cryptic splice site produces a Trf mRNA carrying a 27-base pair (bp), in-frame deletion. Less than 1% of normal levels of a Trf-like protein is found in the serum of Trf(hpx/hpx) mice, most likely resulting from translation of the internally deleted mRNA. Despite their severe Trf deficiency, however, Trf(hpx/hpx) mice initially treated with transferrin injections can survive after weaning without any further treatment. They have massive tissue iron overload develop in all nonhematopoietic tissues, while they continue to have severe iron deficiency anemia. Their liver iron burden is 100-fold greater than that of wild-type mice and 15- to 20-fold more than that of mice lacking the hemochromatosis gene, Hfe. Trf(hpx/hpx) mice thus provide an additional model with a defined molecular defect for the study of genetic iron disorders.


Subject(s)
Alleles , Mutation , Transferrin/deficiency , Transferrin/genetics , Animals , Mice
5.
Acta Crystallogr D Biol Crystallogr ; 55(Pt 6): 1201-3, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10329785

ABSTRACT

Ferrochelatase (protoheme ferrolyase, E.C. 4.99.1.1), the terminal enzyme in the heme biosynthetic pathway, catalyzes the insertion of ferrous iron into protoporphyrin IX to form protoheme. In eukaryotes, the protein is associated with the inner surface of the inner mitochondrial membrane, and in higher animals the enzyme contains a [2Fe-2S] cluster. This cluster is highly sensitive to NO and is coordinated by four Cys residues whose spacing in the primary sequence is unique. Ferrochelatase from Drosophila melanogaster has been expressed in Escherichia coli with an amino-terminal six-histidine tag and purified to homogeneity. The protein has been crystallized with the [2Fe-2S] cluster intact. The crystals belong to space group I422, with unit-cell dimensions a = b = 158.1, c = 171.2 A and two molecules in the asymmetric unit, and diffract to 3. 0 A resolution.


Subject(s)
Drosophila melanogaster/enzymology , Ferrochelatase/isolation & purification , Animals , Catalysis , Crystallography, X-Ray , Ferrochelatase/chemistry , Ferrochelatase/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
6.
J Biol Chem ; 273(35): 22311-6, 1998 Aug 28.
Article in English | MEDLINE | ID: mdl-9712849

ABSTRACT

In a previous study, site-directed mutagenesis experiments identified three of the four ligands to the [2Fe-2S] cluster in animal ferrochelatase as conserved cysteines in the COOH-terminal extension, Cys-403, Cys-406, and Cys-411 in human ferrochelatase (Crouse, B. R., Sellers, V. M., Finnegan, M. G., Dailey, H. A. & Johnson, M. K. (1996) Biochemistry 35, 16222-16229). The nature of the fourth ligand was left unresolved, and spectroscopic studies raised the possibility of one noncysteinyl, oxygenic ligand. In this work, we report two lines of evidence that strongly suggest the fourth ligand is a cysteine residue. Cysteine at position 196 in human recombinant ferrochelatase when changed to a serine results in an inactive enzyme that is lacking the [2Fe-2S] cluster. Furthermore, whole cell EPR studies demonstrate that in the C196S mutant the cluster fails to assemble. Additionally, the cloning and expression of Drosophila melanogaster ferrochelatase has allowed the identification, by EPR and UV-visible spectroscopy, of a [2Fe-2S]2+ cluster with properties analogous to those of animal ferrochelatases. The observation that Drosophila ferrochelatase contains only four conserved cysteines at positions 196, 403, 406, and 411, is in accord with the proposal that these residues function as cluster ligands. In the case of the ferrochelatase iron-sulfur cluster ligands, NH2-Cys-X206-Cys-X2-Cys-X4-Cys-COOH, the position distant from other ligands may lead to a spatial positioning of the cluster near the enzyme active site or at the interface of two domains, thereby explaining the loss of enzyme activity that accompanies cluster degradation and reinforcing the idea that the cluster functions as a regulatory switch.


Subject(s)
Cysteine/metabolism , Drosophila melanogaster/enzymology , Ferrochelatase/metabolism , Iron-Sulfur Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Chromatography, High Pressure Liquid , Cloning, Molecular , Cysteine/chemistry , DNA, Complementary , Electron Spin Resonance Spectroscopy , Ferrochelatase/chemistry , Ferrochelatase/genetics , Humans , Ligands , Mass Spectrometry , Molecular Sequence Data , Mutagenesis, Site-Directed , Sequence Homology, Amino Acid
7.
Blood ; 91(10): 3980-5, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9573038

ABSTRACT

Ferrochelatase (E.C. 4.99.1.1), the enzyme that catalyzes the terminal step in the heme biosynthetic pathway, is the site of defect in the human inherited disease erythropoietic protoporphyria (EPP). Previously it has been demonstrated that patients with EPP may have missense mutations leading to amino acid substitutions, early chain termination, or exon deletions. While it has been clearly demonstrated that two missense mutations result in lowered enzyme activity, it has never been shown what effect specific exon deletions may have. In the current work, recombinant human ferrochelatase has been engineered to have individual exon deletions corresponding to exons 3 through 11. When expressed in Escherichia coli, none of these possesses significant enzyme activity and all lack the [2Fe-2S] cluster. One of the human missense mutations, F417S, and a series of amino acid replacements at this site (ie, F417W, F417Y, and F417L) were examined. With the exception of F417L, all lacked enzyme activity and did not contain the [2Fe-2S] cluster in vivo or as isolated in vitro.


Subject(s)
Ferrochelatase/genetics , Mutation , Porphyria, Hepatoerythropoietic/genetics , Amino Acid Sequence , Base Sequence , DNA Mutational Analysis , Escherichia coli , Exons/genetics , Ferrochelatase/metabolism , Humans , Iron/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Point Mutation , Protoporphyria, Erythropoietic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion
9.
Biochemistry ; 35(50): 16222-9, 1996 Dec 17.
Article in English | MEDLINE | ID: mdl-8973195

ABSTRACT

The five cysteines closest to the carboxyl terminus of human ferrochelatase have been individually mutated to serine, histidine, or aspartate residues in an attempt to identify the protein ligands to the [2Fe-2S] cluster. Mutations of cysteines at positions 403, 406, and 411 (C403D, C403H, C406D, C406H, C406S, C411H, and C411S mutants) all resulted in inactive enzyme that failed to assemble the [2Fe-2S] cluster as judged by whole-cell EPR studies. In contrast, mutation of the cysteines at positions 360 and 395 to serines (C360S and C395S mutants) did not affect the enzymatic activity, and the resulting enzyme assembled a [2Fe-2S] cluster that was spectroscopically indistinguishable from the wild-type enzyme. The results indicate that three of the conserved cysteines in the 30-residue C-terminal extension of mammalian ferrochelatase are involved in ligating the [2Fe-2S] cluster. Resonance Raman and variable-temperature magnetic circular dichroism studies of heme-free preparations of human ferrochelatase are reported, and the spectra are best interpreted in terms of one non-cysteinyl, oxygenic ligand for the [2Fe-2S] cluster. Such anomalous coordination could account for the cluster lability compared to similar clusters with complete cysteinyl ligation and hence may be intrinsic to the proposed regulatory role for this cluster in mammalian ferrochelatases.


Subject(s)
Cysteine , Ferrochelatase/chemistry , Iron-Sulfur Proteins/chemistry , Protein Conformation , Amino Acid Sequence , Animals , Cattle , Cloning, Molecular , Dithionite , Electron Spin Resonance Spectroscopy , Escherichia coli , Ferrochelatase/biosynthesis , Ferrochelatase/isolation & purification , Humans , Iron-Sulfur Proteins/biosynthesis , Iron-Sulfur Proteins/isolation & purification , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Oxidation-Reduction , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Homology, Amino Acid , Spectrum Analysis, Raman
10.
Biochemistry ; 35(8): 2699-704, 1996 Feb 27.
Article in English | MEDLINE | ID: mdl-8611576

ABSTRACT

Ferrochelatase (E.C. 4.99.1.1) is the terminal enzyme of the heme biosynthetic pathway, catalyzing the insertion of ferrous iron into protoporphyrin. In mammals the enzyme contains a labile [2Fe-2S] center. Although this cluster is absent in all prokaryotic, plant, and yeast ferrochelatases, its destruction or elimination from the mammalian enzyme results in loss of enzyme activity. In the current study we present data which clearly demonstrate that mammalian ferrochelatase is strongly inhibited by nitric oxide and that this effect is mediated via destruction of the [2Fe-2S] cluster. Carbon monoxide has no inhibitory effect, and yeast ferrochelatase, which lacks the [2Fe-2S] cluster, is not affected by NO (or CO). EPR and UV-visible absorption of purified recombinant human ferrochelatase provides evidence that NO is targeting the [2Fe-2S] center. UV-visible absorption spectroscopy of both human and murine recombinant ferrochelatase incubated with NO or the NO donor, S-nitroso-N-acetylpenicillamine (SNAP), indicate a rapid loss of the visible absorption spectrum of the [2Fe-2S] cluster. EPR studies of the resulting samples reveal the characteristic axial S = 1/2 resonance, g perpendicular = 2.033, and g parallel = 2.014 of a cysteinyl-coordinated monomeric iron-dinitrosyl cluster degradation product. Parallel spectroscopic studies of spinach ferredoxin, which also contains a [2Fe-2S] cluster, gave no indication of NO-induced cluster degradation under the same experimental conditions. Exposure of DMSO-induced murine erythroleukemia cells exposed to SNAP results in an initial decrease in heme production, suggesting that in vivo the cluster is rapidly destroyed. The potential physiological relevance of these data to the anemias that are found in individuals with chronic infections is discussed.


Subject(s)
Ferrochelatase/chemistry , Animals , Binding Sites , Electron Spin Resonance Spectroscopy , Ferrochelatase/genetics , Ferrochelatase/metabolism , Heme/biosynthesis , Humans , In Vitro Techniques , Iron/chemistry , Mice , Molecular Sequence Data , Molecular Structure , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Sequence Homology, Amino Acid , Spectrophotometry , Spectrophotometry, Ultraviolet , Sulfur/chemistry , Tumor Cells, Cultured
11.
J Biol Chem ; 269(1): 390-5, 1994 Jan 07.
Article in English | MEDLINE | ID: mdl-8276824

ABSTRACT

Ferrochelatase (EC 4.99.1.1) catalyzes the terminal step in the heme biosynthetic pathway, the insertion of ferrous iron into protoporphyrin IX. Herein we report the expression, purification, and characterization of the mature processed form of human and mouse ferrochelatase in Escherichia coli JM109. Metal analysis of the recombinant normal human ferrochelatase reveals that there are approximately 2 iron atoms/molecule of enzyme. This, along with the presence of spectral absorbance near 320 nm, is strongly suggestive that recombinant mammalian ferrochelatase as expressed in E. coli may contain an iron sulfur cluster. Two human protoporphyric ferrochelatases, F417S and M267I, were also expressed and characterized. The M267I mutant possesses the same Km and Vmax as the normal enzyme but exhibits increased thermolability when compared with normal human ferrochelatase. The F417S mutant has less than 2% of the normal activity. Since the Phe-->Ser substitution in this mutation is both chemically and structurally significant, three single amino acid substitutions (Lys, Tyr, and Trp) were engineered and characterized. None of these resulted in a protein with wild type activity. Additionally the carboxyl-terminal 10-amino acid segment, which contains Phe-417, from the yeast sequence was substituted, but this construct had no activity. Elimination of the carboxyl-terminal 30 amino acid residues (which include Phe-417) results in a protein the same length as the bacterial ferrochelatases, but it is an inactive enzyme.


Subject(s)
Ferrochelatase/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA Primers , Electrophoresis, Polyacrylamide Gel , Escherichia coli , Ferrochelatase/isolation & purification , Ferrochelatase/metabolism , Humans , Kinetics , Mice , Molecular Sequence Data , Mutation , Plasmids , Porphyrins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...