Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters










Publication year range
1.
PLoS Comput Biol ; 14(11): e1006626, 2018 11.
Article in English | MEDLINE | ID: mdl-30496278

ABSTRACT

The conformational dynamics of proteins is rarely used in methodologies used to predict the impact of genetic mutations due to the paucity of three-dimensional protein structures as compared to the vast number of available sequences. Until now a three-dimensional (3D) structure has been required to predict the conformational dynamics of a protein. We introduce an approach that estimates the conformational dynamics of a protein, without relying on structural information. This de novo approach utilizes coevolving residues identified from a multiple sequence alignment (MSA) using Potts models. These coevolving residues are used as contacts in a Gaussian network model (GNM) to obtain protein dynamics. B-factors calculated using sequence-based GNM (Seq-GNM) are in agreement with crystallographic B-factors as well as theoretical B-factors from the original GNM that utilizes the 3D structure. Moreover, we demonstrate the ability of the calculated B-factors from the Seq-GNM approach to discriminate genomic variants according to their phenotypes for a wide range of proteins. These results suggest that protein dynamics can be approximated based on sequence information alone, making it possible to assess the phenotypes of nSNVs in cases where a 3D structure is unknown. We hope this work will promote the use of dynamics information in genetic disease prediction at scale by circumventing the need for 3D structures.


Subject(s)
Acyl-CoA Dehydrogenase/chemistry , Computational Biology/methods , Disease Susceptibility , Neurons/metabolism , Protein Isoforms , Proteins/chemistry , Animals , Computer Simulation , Cytochrome Reductases/chemistry , Genomics , Humans , Imaging, Three-Dimensional , Molecular Conformation , Muramidase/chemistry , Normal Distribution , Phenotype , Protein Conformation , ROC Curve , Rats
2.
Arch Biochem Biophys ; 579: 1-7, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26014136

ABSTRACT

Mia40 participates in oxidative protein folding within the mitochondrial intermembrane space (IMS) by mediating the transfer of reducing equivalents from client proteins to FAD-linked oxidoreductases of the Erv1 family (lfALR in mammals). Here we investigate the specificity of the human Mia40/lfALR system towards non-cognate unfolded protein substrates to assess whether the efficient introduction of disulfides requires a particular amino acid sequence context or the presence of an IMS targeting signal. Reduced pancreatic ribonuclease A (rRNase), avian lysozyme, and riboflavin binding protein are all competent substrates of the Mia40/lfALR system, although they lack those sequence features previously thought to direct disulfide bond formation in cognate IMS substrates. The oxidation of rRNase by Mia40 does not limit overall turnover of unfolded substrate by the Mia40/lfALR system. Mia40 is an ineffective protein disulfide isomerase when its ability to restore enzymatic activity from scrambled RNase is compared to that of protein disulfide isomerase. Mia40's ability to bind amphipathic peptides is evident by avid binding to the isolated B-chain during the insulin reductase assay. In aggregate these data suggest that the Mia40/lfALR system has a broad sequence specificity and that potential substrates may be protected from adventitious oxidation by kinetic sequestration within the mitochondrial IMS.


Subject(s)
Cytochrome Reductases/chemistry , Cytochrome Reductases/ultrastructure , Isomerases/chemistry , Isomerases/ultrastructure , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/ultrastructure , Amino Acid Sequence , Binding Sites , Computer Simulation , Enzyme Activation , Humans , Mitochondrial Precursor Protein Import Complex Proteins , Models, Chemical , Models, Molecular , Molecular Sequence Data , Oxidants/chemistry , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors , Protein Binding , Protein Conformation , Protein Folding , Structure-Activity Relationship
3.
Biochem J ; 464(3): 449-59, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25269795

ABSTRACT

Erv1 (essential for respiration and viability 1) is an FAD-dependent thiol oxidase of the Erv/ALR (augmenter of liver regeneration) sub-family. It is an essential component of the mitochondrial import and assembly (MIA) pathway, playing an important role in the oxidative folding of the mitochondrial intermembrane space (IMS) proteins and linking the MIA pathway to the mitochondrial respiratory chain via cytochrome c (cyt c). The importance of the Erv/ALR enzymes was also demonstrated in a recent study where a single mutation in the human ALR (R194H) leads to autosomal recessive myopathy [Di Fonzo, Ronchi, Lodi, Fassone, Tigano, Lamperti, Corti, Bordoni, Fortunato, Nizzardo et al. (2009) Am. J. Hum. Genet. 84, 594-604]. However, the molecular mechanism of the disease is still unclear. In the present study, we use yeast Erv1 as a model to provide clear evidence for a progressive functional defect in the catalytic activity of the corresponding Erv1 R182H mutant. We show that the FAD cofactor was released from Erv1 R182H during its catalytic cycle, which led to the inactivation of the enzyme. We also characterized the effects of the mutation on the folding and stability of Erv1 and tested our in vitro findings in vivo using a yeast genetic approach. The results of the present study allow us to provide a model for the functional defect in Erv1 R182H, which could potentially be extended to human ALR R194H and provides insights into the molecular basis of autosomal recessive myopathy.


Subject(s)
Cytochrome Reductases/genetics , Cytochrome Reductases/metabolism , Muscular Diseases/genetics , Mutation, Missense , Amino Acid Sequence , Amino Acid Substitution , Catalysis , Catalytic Domain/genetics , Coenzymes/metabolism , Cytochrome Reductases/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Oxidoreductases Acting on Sulfur Group Donors , Protein Binding , Protein Structure, Tertiary/genetics , Sequence Homology, Amino Acid
4.
J Biol Chem ; 288(1): 401-11, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23172229

ABSTRACT

Here, we report for the first time in vitro reconstitution of the respiratory supercomplexes from individual complexes III and IV. Complexes III and IV were purified from Saccharomyces cerevisiae mitochondria. Complex III contained eight molecules of cardiolipin, and complex IV contained two molecules of cardiolipin, as determined by electrospray ionization-mass spectrometry. Complex IV also contained Rcf1p. No supercomplexes were formed upon mixing of the purified complexes, and low amounts of the supercomplex trimer III(2)IV(1) were formed after reconstitution into proteoliposomes containing only phosphatidylcholine and phosphatidylethanolamine. Further addition of cardiolipin to the proteoliposome reconstitution mixture resulted in distinct formation of both the III(2)IV(1) supercomplex trimer and III(2)IV(2) supercomplex tetramer. No other anionic phospholipid was as effective as cardiolipin in supporting tetramer formation. Phospholipase treatment of complex IV prevented trimer formation in the absence of cardiolipin. Both trimer and tetramer formations were restored by cardiolipin. Analysis of the reconstituted tetramer by single particle electron microscopy confirmed native organization of individual complexes within the supercomplex. In conclusion, although some trimer formation occurred dependent only on tightly bound cardiolipin, tetramer formation required additional cardiolipin. This is consistent with the high cardiolipin content in the native tetramer. The dependence on cardiolipin for supercomplex formation suggests that changes in cardiolipin levels resulting from changes in physiological conditions may control the equilibrium between individual respiratory complexes and supercomplexes in vivo.


Subject(s)
Cardiolipins/chemistry , Electron Transport Complex III/metabolism , Electron Transport Complex IV/metabolism , Saccharomyces cerevisiae/metabolism , Cytochrome Reductases/chemistry , Electron Transport Complex IV/chemistry , Lipids/chemistry , Microscopy, Electron/methods , Mitochondria/metabolism , Phospholipases/chemistry , Protein Binding , Proteolipids/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Ubiquinone/analogs & derivatives , Ubiquinone/chemistry
5.
J Biol Chem ; 288(4): 2676-88, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23233680

ABSTRACT

Mia40 and the sulfhydryl:cytochrome c oxidoreductase Erv1/ALR are essential for oxidative protein import into the mitochondrial intermembrane space in yeast and mammals. Although mitochondrial protein import is functionally conserved in the course of evolution, many organisms seem to lack Mia40. Moreover, except for in organello import studies and in silico analyses, nothing is known about the function and properties of protist Erv homologues. Here we compared Erv homologues from yeast, the kinetoplastid parasite Leishmania tarentolae, and the non-related malaria parasite Plasmodium falciparum. Both parasite proteins have altered cysteine motifs, formed intermolecular disulfide bonds in vitro and in vivo, and could not replace Erv1 from yeast despite successful mitochondrial protein import in vivo. To analyze its enzymatic activity, we established the expression and purification of recombinant full-length L. tarentolae Erv and compared the mechanism with related and non-related flavoproteins. Enzyme assays indeed confirmed an electron transferase activity with equine and yeast cytochrome c, suggesting a conservation of the enzymatic activity in different eukaryotic lineages. However, although Erv and non-related flavoproteins are intriguing examples of convergent molecular evolution resulting in similar enzyme properties, the mechanisms of Erv homologues from parasitic protists and opisthokonts differ significantly. In summary, the Erv-mediated reduction of cytochrome c might be highly conserved throughout evolution despite the apparent absence of Mia40 in many eukaryotes. Nevertheless, the knowledge on mitochondrial protein import in yeast and mammals cannot be generally transferred to all other eukaryotes, and the corresponding pathways, components, and mechanisms remain to be analyzed.


Subject(s)
Cytochrome Reductases/chemistry , Cytochromes c/chemistry , Mitochondrial Proteins/physiology , Oxidoreductases Acting on Sulfur Group Donors/physiology , Oxidoreductases/chemistry , Saccharomyces cerevisiae Proteins/physiology , Amino Acid Sequence , Animals , Cell Lineage , Computational Biology/methods , Electrons , Genetic Complementation Test , Kinetics , Kinetoplastida/metabolism , Leishmania , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Molecular Conformation , Molecular Sequence Data , Mutagenesis, Site-Directed , Oxidoreductases Acting on Sulfur Group Donors/genetics , Plasmodium/metabolism , Protein Transport , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Amino Acid
6.
J Mol Biol ; 425(2): 222-31, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23159557

ABSTRACT

Sulfur, a key contributor to biological reactivity, is not amendable to investigations by biological NMR spectroscopy. To utilize selenium as a surrogate, we have developed a generally applicable (77)Se isotopic enrichment method for heterologous proteins expressed in Escherichia coli. We demonstrate (77)Se NMR spectroscopy of multiple selenocysteine and selenomethionine residues in the sulfhydryl oxidase augmenter of liver regeneration (ALR). The resonances of the active-site residues were assigned by comparing the NMR spectra of ALR bound to oxidized and reduced flavin adenine dinucleotide. An additional resonance appears only in the presence of the reducing agent and disappears readily upon exposure to air and subsequent reoxidation of the flavin. Hence, (77)Se NMR spectroscopy can be used to report the local electronic environment of reactive and structural sulfur sites, as well as changes taking place in those locations during catalysis.


Subject(s)
Cysteine/metabolism , Cytochrome Reductases/chemistry , Flavins/metabolism , Magnetic Resonance Spectroscopy , Selenocysteine/metabolism , Selenomethionine/metabolism , Catalysis , Crystallography, X-Ray , Cysteine/chemistry , Cysteine/genetics , Cytochrome Reductases/genetics , Cytochrome Reductases/metabolism , Humans , Mutation/genetics , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors , Protein Conformation , Protein Folding , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
7.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 9): 1128-33, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22948913

ABSTRACT

The crystal structure of the protein augmenter of liver regeneration containing a 14-residue hexahistidine purification tag (hsALR) has been determined to 2.4 Å resolution by Cd-SAD using a highly redundant data set collected on a rotating-anode home X-ray source and processed in 1998. The hsALR crystal structure is a tetramer composed of two homodimers bridged by a novel Cd(2)Cl(4)O(6) cluster via binding to the side-chain carboxylate groups of two solvent-exposed aspartic acid residues. A comparison with the native sALR tetramer shows that the cluster dramatically changes the hsALR dimer-dimer interface, which can now better accommodate the extra 14 N-terminal residues associated with the purification tag. The refined 2.4 Šresolution structure is in good agreement with both the X-ray data (R(cryst) of 0.165, R(free) of 0.211) and the expected stereochemistry (r.m.s. deviations from ideality for bond lengths and bond angles of 0.007 Å and 1.15°, respectively).


Subject(s)
Acids/chemistry , Cadmium Chloride/chemistry , Cadmium Compounds/chemistry , Cytochrome Reductases/chemistry , Proteins/chemistry , Crystallography, X-Ray , Dimerization , Humans , Models, Molecular , Oxidoreductases Acting on Sulfur Group Donors , Protein Structure, Quaternary , Protein Structure, Tertiary
8.
Curr Protein Pept Sci ; 13(3): 224-31, 2012 May.
Article in English | MEDLINE | ID: mdl-22612783

ABSTRACT

The process of oxidative folding in the intermembrane mitochondrial space (IMS) is an exciting field of research because folding is simultaneously coupled to protein translocation and functional regulation. Contrary to the endoplasmatic reticulum ER where several chaperones of the disulfide isomerase family exist, oxidative folding in the IMS is exclusively catalyzed by the oxoreductase Mia40 that recognizes a group of proteins with characteristic cysteine motifs organized in twin CX(3)C, twin CX(9)C or CX(2)C motifs. In this review, we discuss the structural and biochemical studies leading to our current understanding of the Mia40 pathway as well as the open questions on the field. In fact, despite significant advances, several key points on the Mia40 pathway remain to clarify namely on the molecular mechanism trough which substrate oxidative folding is catalyzed. This issue is receiving increasing attention since failures in the import, sorting and folding of mitochondrial proteins is related to an increasing number of debilitating human disorders.


Subject(s)
Mitochondrial Membranes/metabolism , Protein Folding , Animals , Carrier Proteins/metabolism , Carrier Proteins/physiology , Cytochrome Reductases/chemistry , Cytochrome Reductases/metabolism , Cytochrome Reductases/physiology , Humans , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/physiology , Mitochondrial Precursor Protein Import Complex Proteins , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Transport
9.
J Am Chem Soc ; 134(3): 1442-5, 2012 Jan 25.
Article in English | MEDLINE | ID: mdl-22224850

ABSTRACT

The oxidative folding mechanism in the intermembrane space of human mitochondria underpins a disulfide relay system consisting of the import receptor Mia40 and the homodimeric FAD-dependent thiol oxidase ALR. The flavoprotein ALR receives two electrons per subunit from Mia40, which are then donated through one-electron reactions to two cytochrome c molecules, thus mediating a switch from two-electron to one-electron transfer. We dissect here the mechanism of the electron flux within ALR, characterizing at the atomic level the ALR intermediates that allow electrons to rapidly flow to cytochrome c. The intermediate critical for the electron-transfer process implies the formation of a specific inter-subunit disulfide which exclusively allows electron flow from Mia40 to FAD. This finding allows us to present a complete model for the electron-transfer pathway in ALR.


Subject(s)
Cytochrome Reductases/metabolism , Cytochromes c/metabolism , Disulfides/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Circular Dichroism , Cytochrome Reductases/chemistry , Cytochromes c/chemistry , Disulfides/chemistry , Electron Transport , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Precursor Protein Import Complex Proteins , Models, Molecular , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors , Spectrophotometry, Ultraviolet
10.
Biochemistry ; 51(1): 265-72, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-22148553

ABSTRACT

Typically, simple flavoprotein oxidases couple the oxidation of their substrates with the formation of hydrogen peroxide without release of significant levels of the superoxide ion. However, two evolutionarily related single-domain sulfhydryl oxidases (Erv2p; a yeast endoplasmic reticulum resident protein and augmenter of liver regeneration, ALR, an enzyme predominantly found in the mitochondrial intermembrane) release up to ~30% of the oxygen they reduce as the superoxide ion. Both enzymes oxidize dithiol substrates via a redox-active disulfide adjacent to the flavin cofactor within the helix-rich Erv domain. Subsequent reduction of the flavin is followed by transfer of reducing equivalents to molecular oxygen. Superoxide release was initially detected using tris(3-hydroxypropyl)phosphine (THP) as an alternative reducing substrate to dithiothreitol (DTT). THP, and other phosphines, showed anomalously high turnover numbers with Erv2p and ALR in the oxygen electrode, but oxygen consumption was drastically suppressed upon the addition of superoxide dismutase. The superoxide ion initiates a radical chain reaction promoting the aerobic oxidation of phosphines with the formation of hydrogen peroxide. Use of a known flux of superoxide generated by the xanthine/xanthine oxidase system showed that one superoxide ion stimulates the reduction of 27 and 4.5 molecules of oxygen using THP and tris(2-carboxyethyl)phosphine (TCEP), respectively. This superoxide-dependent amplification of oxygen consumption by phosphines provides a new kinetic method for the detection of superoxide. Superoxide release was also observed by a standard chemiluminescence method using a luciferin analogue (MCLA) when 2 mM DTT was employed as a substrate of Erv2p and ALR. The percentage of superoxide released from Erv2p increased to ~65% when monomeric mutants of the normally homodimeric enzyme were used. In contrast, monomeric multidomain quiescin sulfhydryl oxidase enzymes that also contain an Erv FAD-binding fold release only 1-5% of their total reduced oxygen species as the superoxide ion. Aspects of the mechanism and possible physiological significance of superoxide release from these Erv-domain flavoproteins are discussed.


Subject(s)
Cation Transport Proteins/chemistry , Cytochrome Reductases/chemistry , Flavoproteins/chemistry , Oxidoreductases Acting on Sulfur Group Donors/chemistry , Oxidoreductases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Superoxides/chemistry , Animals , Aspergillus niger/enzymology , Catalysis , Cation Transport Proteins/genetics , Cattle , Humans , Milk/enzymology , Oxidation-Reduction , Oxidoreductases/genetics , Protein Multimerization , Protozoan Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Trypanosoma brucei brucei/enzymology
11.
Cell Cycle ; 10(14): 2263-8, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21636978

ABSTRACT

Hematopoietic stem cells (HSC) are a relatively quiescent pool of cells that perform the arduous task of replacing the short-lived mature cells of the peripheral blood. While a rapid expansion of HSCs under periods of hematological stress is warranted, their enhanced proliferation during homeostasis leads to loss of function. We recently reported that in HSCs, the evolutionarily conserved growth factor erv1-like (Gfer) acts to counter jun activation domain-binding protein 1 (Jab1)-mediated nuclear export and destabilization of the cell cycle inhibitor, p27kip1, by directly binding to and sequestering the COP9 signalosome (CSN) subunit. Through this mechanism, Gfer promotes quiescence and maintains the functional integrity of HSCs. Here, we extend our study to demonstrate an association between Gfer and Ca2+/calmodulin-dependent protein kinase IV (CaMKIV) in the regulation of HSC proliferation. Highly proliferative and functionally deficient Camk4-/- HSCs possess significantly lower levels of Gfer and p27kip1. Ectopic expression of Gfer restores quiescence and elevates p27kip1 expression in Camk4-/- HSCs. These results further substantiate a critical role for Gfer in the restriction of unwarranted proliferation in HSCs through the inhibition of Jab1 and subsequent stabilization and nuclear retention of p27kip1. This Gfer-mediated pro-quiescence mechanism could be therapeutically exploited in the treatment of hematological malignancies associated with elevated Jab1 and reduced p27kip1.


Subject(s)
Cytochrome Reductases/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , COP9 Signalosome Complex , Calcium-Calmodulin-Dependent Protein Kinase Type 4/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cytochrome Reductases/chemistry , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Oxidoreductases Acting on Sulfur Group Donors , Peptide Hydrolases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/metabolism
12.
Proc Natl Acad Sci U S A ; 108(12): 4811-6, 2011 Mar 22.
Article in English | MEDLINE | ID: mdl-21383138

ABSTRACT

Oxidative protein folding in the mitochondrial intermembrane space requires the transfer of a disulfide bond from MIA40 to the substrate. During this process MIA40 is reduced and regenerated to a functional state through the interaction with the flavin-dependent sulfhydryl oxidase ALR. Here we present the mechanistic basis of ALR-MIA40 interaction at atomic resolution by biochemical and structural analyses of the mitochondrial ALR isoform and its covalent mixed disulfide intermediate with MIA40. This ALR isoform contains a folded FAD-binding domain at the C-terminus and an unstructured, flexible N-terminal domain, weakly and transiently interacting one with the other. A specific region of the N-terminal domain guides the interaction with the MIA40 substrate binding cleft (mimicking the interaction of the substrate itself), without being involved in the import of ALR. The hydrophobicity-driven binding of this region ensures precise protein-protein recognition needed for an efficient electron transfer process.


Subject(s)
Cytochrome Reductases/chemistry , Flavin-Adenine Dinucleotide/chemistry , Mitochondrial Membrane Transport Proteins/chemistry , Binding Sites , Cytochrome Reductases/metabolism , Electron Transport/physiology , Flavin-Adenine Dinucleotide/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Isoenzymes/chemistry , Isoenzymes/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Oxidoreductases Acting on Sulfur Group Donors , Protein Structure, Tertiary , Substrate Specificity/physiology
13.
Biochemistry ; 49(31): 6737-45, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20593814

ABSTRACT

The sulfhydryl oxidase augmenter of liver regeneration (ALR) binds FAD in a helix-rich domain that presents a CxxC disulfide proximal to the isoalloxazine ring of the flavin. Head-to-tail interchain disulfide bonds link subunits within the homodimer of both the short, cytokine-like, form of ALR (sfALR), and a longer form (lfALR) which resides in the mitochondrial intermembrane space (IMS). lfALR has an 80-residue N-terminal extension with an additional CxxC motif required for the reoxidation of reduced Mia40 during oxidative protein folding within the IMS. Recently, Di Fonzo et al. [Di Fonzo, A., Ronchi, D., Lodi, T., Fassone, E., Tigano, M., Lamperti, C., Corti, S., Bordoni, A., Fortunato, F., Nizzardo, M., Napoli, L., Donadoni, C., Salani, S., Saladino, F., Moggio, M., Bresolin, N., Ferrero, I., and Comi, G. P. (2009) Am. J. Hum. Genet. 84, 594-604] described an R194H mutation of human ALR that led to cataract, progressive muscle hypotonia, and hearing loss in three children. The current work presents a structural and enzymological characterization of the human R194H mutant in lf- and sfALR. A crystal structure of human sfALR was determined by molecular replacement using the rat sfALR structure. R194 is located at the subunit interface of sfALR, close to the intersubunit disulfide bridges. The R194 guanidino moiety participates in three H-bonds: two main-chain carbonyl oxygen atoms (from R194 itself and from C95 of the intersubunit disulfide of the other protomer) and with the 2'-OH of the FAD ribose. The R194H mutation has minimal effect on the enzyme activity using model and physiological substrates of short and long ALR forms. However, the mutation adversely affects the stability of both ALR forms: e.g., by decreasing the melting temperature by about 10 degrees C, by increasing the rate of dissociation of FAD from the holoenzyme by about 45-fold, and by strongly enhancing the susceptibility of sfALR to partial proteolysis and to reduction of its intersubunit disulfide bridges by glutathione. Finally, a comparison of the TROSY-HSQC 2D NMR spectra of wild-type sfALR and its R194H mutant reveals a significant increase in conformational flexibility in the mutant protein. In sum, these in vitro data document the major impact of the seemingly conservative R194H mutation on the stability of dimeric ALR and complement the in vivo observations of Di Fonzo et al.


Subject(s)
Cytochrome Reductases/chemistry , Muscular Diseases/genetics , Mutation, Missense , Animals , Child , Cytochrome Reductases/genetics , Enzyme Stability , Humans , Mutation, Missense/physiology , Oxidoreductases Acting on Sulfur Group Donors , Pliability , Protein Conformation , Protein Multimerization , Rats
14.
J Biol Chem ; 285(25): 19450-9, 2010 Jun 18.
Article in English | MEDLINE | ID: mdl-20404342

ABSTRACT

Zinc is essential for function of mitochondria as a cofactor for several matrix zinc metalloproteins. We demonstrate that a labile cationic zinc component of low molecular mass exists in the yeast mitochondrial matrix. This zinc pool is homeostatically regulated in response to the cellular zinc status. This pool of zinc is functionally important because matrix targeting of a cytosolic zinc-binding protein reduces the level of labile zinc and interferes with mitochondrial respiratory function. We identified a series of proteins that modulate the matrix zinc pool, one of which is a novel conserved mitochondrial protein designated Mzm1. Mutant mzm1Delta cells have reduced total and labile mitochondrial zinc, and these cells are hypersensitive to perturbations of the labile pool. In addition, mzm1Delta cells have a destabilized cytochrome c reductase (Complex III) without any effects on Complexes IV or V. Thus, we have established that a link exists between Complex III integrity and the labile mitochondrial zinc pool.


Subject(s)
Candida albicans/metabolism , Metalloproteins/metabolism , Oxygen Consumption , Cations , Cytochrome Reductases/chemistry , Electron Transport , Electrophoresis, Polyacrylamide Gel , Gene Deletion , Gene Expression Regulation, Fungal , Genetic Vectors , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Phenotype , Subcellular Fractions/metabolism , Zinc/chemistry
15.
J Biol Chem ; 285(5): 3064-75, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-19948738

ABSTRACT

Neuronal nitric-oxide synthase (nNOS) contains a unique autoinhibitory insert (AI) in its FMN subdomain that represses nNOS reductase activities and controls the calcium sensitivity of calmodulin (CaM) binding to nNOS. How the AI does this is unclear. A conserved charged residue (Lys(842)) lies within a putative CaM binding helix in the middle of the AI. We investigated its role by substituting residues that neutralize (Ala) or reverse (Glu) the charge at Lys(842). Compared with wild type nNOS, the mutant enzymes had greater cytochrome c reductase and NADPH oxidase activities in the CaM-free state, were able to bind CaM at lower calcium concentration, and had lower rates of heme reduction and NO synthesis in one case (K842A). Moreover, stopped-flow spectrophotometric experiments with the nNOS reductase domain indicate that the CaM-free mutants had faster flavin reduction kinetics and had less shielding of their FMN subdomains compared with wild type and no longer increased their level of FMN shielding in response to NADPH binding. Thus, Lys(842) is critical for the known functions of the AI and also enables two additional functions of the AI as newly identified here: suppression of electron transfer to FMN and control of the conformational equilibrium of the nNOS reductase domain. Its effect on the conformational equilibrium probably explains suppression of catalysis by the AI.


Subject(s)
Calmodulin/chemistry , Flavins/chemistry , Lysine/chemistry , Neurons/enzymology , Nitric Oxide Synthase/metabolism , Amino Acid Sequence , Animals , Cattle , Cytochrome Reductases/chemistry , Humans , Kinetics , Molecular Sequence Data , Mutation , NADPH Oxidases/chemistry , Protein Binding , Protein Structure, Tertiary , Rats , Sequence Homology, Amino Acid
16.
Proc Natl Acad Sci U S A ; 106(38): 16169-73, 2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19805275

ABSTRACT

The heme-copper oxygen reductases are redox-driven proton pumps that generate a proton motive force in both prokaryotes and mitochondria. These enzymes have been divided into 3 evolutionarily related groups: the A-, B- and C-families. Most experimental work on proton-pumping mechanisms has been performed with members of the A-family. These enzymes require 2 proton input pathways (D- and K-channels) to transfer protons used for oxygen reduction chemistry and for proton pumping, with the D-channel transporting all pumped protons. In this work we use site-directed mutagenesis to demonstrate that the ba(3) oxygen reductase from Thermus thermophilus, a representative of the B-family, does not contain a D-channel. Rather, it utilizes only 1 proton input channel, analogous to that of the A-family K-channel, and it delivers protons to the active site for both O2 chemistry and proton pumping. Comparison of available subunit I sequences reveals that the only structural elements conserved within the oxygen reductase families that could perform these functions are active-site components, namely the covalently linked histidine-tyrosine, the Cu(B) and its ligands, and the active-site heme and its ligands. Therefore, our data suggest that all oxygen reductases perform the same chemical reactions for oxygen reduction and comprise the essential elements of the proton-pumping mechanism (e.g., the proton-loading and kinetic-gating sites). These sites, however, cannot be located within the D-channel. These results along with structural considerations point to the A-propionate region of the active-site heme and surrounding water molecules as the proton-loading site.


Subject(s)
Bacterial Proteins/metabolism , Cytochrome Reductases/metabolism , Protons , Thermus thermophilus/enzymology , Amino Acid Sequence , Bacterial Proteins/genetics , Catalytic Domain , Crystallography, X-Ray , Cytochrome Reductases/chemistry , Cytochrome Reductases/genetics , Cytochrome b Group/metabolism , Electron Transport Complex IV/metabolism , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Oxidation-Reduction , Oxygen/metabolism , Protein Structure, Tertiary , Proton Pumps/genetics , Proton Pumps/metabolism , Sequence Homology, Amino Acid , Thermus thermophilus/genetics
17.
Biochemistry ; 48(22): 4828-37, 2009 Jun 09.
Article in English | MEDLINE | ID: mdl-19397338

ABSTRACT

Augmenter of liver regeneration (ALR) is both a growth factor and a sulfhydryl oxidase that binds FAD in an unusual helix-rich domain containing a redox-active CxxC disulfide proximal to the flavin ring. In addition to the cytokine form of ALR (sfALR) that circulates in serum, a longer form, lfALR, is believed to participate in oxidative trapping of reduced proteins entering the mitochondrial intermembrane space (IMS). This longer form has an 80-residue N-terminal extension containing an additional, distal, CxxC motif. This work presents the first enzymological characterization of human lfALR. The N-terminal region conveys no catalytic advantage toward the oxidation of the model substrate dithiothreitol (DTT). In addition, a C71A or C74A mutation of the distal disulfide does not increase the turnover number toward DTT. Unlike Erv1p, the yeast homologue of lfALR, static spectrophotometric experiments with the human oxidase provide no evidence of communication between distal and proximal disulfides. An N-terminal His-tagged version of human Mia40, a resident oxidoreductase of the IMS and a putative physiological reductant of lfALR, was subcloned and expressed in Escherichia coli BL21 DE3 cells. Mia40, as isolated, shows a visible spectrum characteristic of an Fe-S center and contains 0.56 +/- 0.02 atom of iron per subunit. Treatment of Mia40 with guanidine hydrochloride and triscarboxyethylphosphine hydrochloride during purification removed this chromophore. The resulting protein, with a reduced CxC motif, was a good substrate of lfALR. However, neither sfALR nor lfALR mutants lacking the distal disulfide could oxidize reduced Mia40 efficiently. Thus, catalysis involves a flow of reducing equivalents from the reduced CxC motif of Mia40 to distal and then proximal CxxC motifs of lfALR to the flavin ring and, finally, to cytochrome c or molecular oxygen.


Subject(s)
Cytochrome Reductases/chemistry , Cytochrome Reductases/metabolism , Flavins/chemistry , Mitochondrial Membranes/metabolism , Oxidoreductases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Cytochrome Reductases/genetics , Cytochromes c/metabolism , Humans , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/enzymology , Mitochondrial Precursor Protein Import Complex Proteins , Molecular Sequence Data , Mutagenesis, Site-Directed , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sequence Homology, Amino Acid , Substrate Specificity/genetics
18.
J Biol Chem ; 284(18): 11892-9, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19251696

ABSTRACT

We have investigated the possible biochemical basis for enhancements in NO production in endothelial cells that have been correlated with agonist- or shear stress-evoked phosphorylation at Ser-1179. We have found that a phosphomimetic substitution at Ser-1179 doubles maximal synthase activity, partially disinhibits cytochrome c reductase activity, and lowers the EC(50)(Ca(2+)) values for calmodulin binding and enzyme activation from the control values of 182 +/- 2 and 422 +/- 22 nm to 116 +/- 2 and 300 +/- 10 nm. These are similar to the effects of a phosphomimetic substitution at Ser-617 (Tran, Q. K., Leonard, J., Black, D. J., and Persechini, A. (2008) Biochemistry 47, 7557-7566). Although combining substitutions at Ser-617 and Ser-1179 has no additional effect on maximal synthase activity, cooperativity between the two substitutions completely disinhibits reductase activity and further reduces the EC(50)(Ca(2+)) values for calmodulin binding and enzyme activation to 77 +/- 2 and 130 +/- 5 nm. We have confirmed that specific Akt-catalyzed phosphorylation of Ser-617 and Ser-1179 and phosphomimetic substitutions at these positions have similar functional effects. Changes in the biochemical properties of eNOS produced by combined phosphorylation at Ser-617 and Ser-1179 are predicted to substantially increase synthase activity in cells at a typical basal free Ca(2+) concentration of 50-100 nm.


Subject(s)
Calcium/chemistry , Calmodulin/chemistry , Nitric Oxide Synthase Type III/chemistry , Proto-Oncogene Proteins c-akt/chemistry , Amino Acid Substitution , Animals , Calcium/metabolism , Calmodulin/genetics , Calmodulin/metabolism , Cattle , Cytochrome Reductases/chemistry , Cytochrome Reductases/genetics , Cytochrome Reductases/metabolism , Humans , Mutation, Missense , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/physiology , Protein Binding/physiology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Stress, Physiological/physiology
19.
J Biol Chem ; 282(16): 12240-8, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17322303

ABSTRACT

Mitochondrial respiratory chain complexes are arranged in supercomplexes within the inner membrane. Interaction of cytochrome c reductase (complex III) and cytochrome c oxidase (complex IV) was investigated in Saccharomyces cerevisiae. Projection maps at 15 A resolution of supercomplexes III(2) + IV(1) and III(2) + IV(2) were obtained by electron microscopy. Based on a comparison of our maps with atomic x-ray structures for complexes III and IV we present a pseudo-atomic model of their precise interaction. Two complex IV monomers are specifically attached to dimeric complex III with their convex sides. The opposite sides, which represent the complex IV dimer interface in the x-ray structure, are open for complex IV-complex IV interactions. This could lead to oligomerization of III(2) + IV(2) supercomplexes, but this was not detected. Instead, binding of cytochrome c to the supercomplexes was revealed. It was calculated that cytochrome c has to move less than 40 A at the surface of the supercomplex for electron transport between complex III(2) and complex IV. Hence, the prime function of the supercomplex III(2) + IV(2) is proposed to be a scaffold for effective electron transport between complexes III and IV.


Subject(s)
Cytochrome Reductases/chemistry , Electron Transport Complex IV/chemistry , Saccharomyces cerevisiae/enzymology , Animals , Cattle , Cytochromes c/chemistry , Dimerization , Electron Transport , Electron Transport Complex IV/metabolism , Microscopy, Electron , Mitochondria/metabolism , Molecular Conformation , Oxygen Consumption , Protein Binding , Protein Conformation , Sucrose/pharmacology
20.
Biochemistry ; 45(51): 15405-10, 2006 Dec 26.
Article in English | MEDLINE | ID: mdl-17176062

ABSTRACT

In the respiratory chains of aerobic organisms, oxygen reductase members of the heme-copper superfamily couple the reduction of O2 to proton pumping, generating an electrochemical gradient. There are three distinct families of heme-copper oxygen reductases: A, B, and C types. The A- and B-type oxygen reductases have an active-site tyrosine that forms a unique cross-linked histidine-tyrosine cofactor. In the C-type oxygen reductases (also called cbb3 oxidases), an analogous active-site tyrosine has recently been predicted by molecular modeling to be located within a different transmembrane helix in comparison to the A- and B-type oxygen reductases. In this work, Fourier-transform mass spectrometry is used to show that the predicted tyrosine forms a histidine-tyrosine cross-linked cofactor in the active site of the C-type oxygen reductases. This is the first known example of the evolutionary migration of a post-translationally modified active-site residue. It also verifies the presence of a unique cofactor in all three families of proton-pumping respiratory oxidases, demonstrating that these enzymes likely share a common reaction mechanism and that the histidine-tyrosine cofactor may be a required component for proton pumping.


Subject(s)
Copper/chemistry , Electron Transport Complex IV/chemistry , Electron Transport Complex IV/metabolism , Evolution, Molecular , Heme/chemistry , Protein Processing, Post-Translational , Proton Pumps/chemistry , Proton Pumps/metabolism , Amino Acid Sequence , Binding Sites/genetics , Cytochrome Reductases/chemistry , Cytochrome Reductases/metabolism , Electron Transport Complex IV/genetics , Molecular Sequence Data , Protein Processing, Post-Translational/genetics , Protein Transport/genetics , Proton Pumps/genetics , Rhodobacter sphaeroides/enzymology , Vibrio cholerae/enzymology , Vibrio cholerae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...