Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Pathogens ; 13(6)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38921778

ABSTRACT

The sole known heme enzyme of the parasitic protist Giardia intestinalis is a flavohemoglobin (gFlHb) that acts as a nitric oxide dioxygenase (NOD) and protects the organism from the free radical nitric oxide. To learn more about the properties of this enzyme, we measured its nitric oxide dioxygenase, NADH oxidase, and cytochrome c reductase activities and compared these to the activities of the E. coli flavohemoglobin (Hmp). The turnover number for the NOD activity of gFlHb (23 s-1) is about two-thirds of that of Hmp (34 s-1) at pH 6.5 and 37 °C. The two enzymes differ in their sensitivity towards molecules that act as heme ligands. For both gFlHb and Hmp, inhibition with miconazole, a large imidazole ligand, is adequately described by simple competitive inhibition, with KI = 10 µM and 0.27 µM for gFlHb and Hmp, respectively. Inhibition plots with the small ligand imidazole were biphasic, which is consistent with previous experiments with carbon monoxide as a probe that show that the active site of flavohemoglobins exists in two conformations. Interestingly, the largest difference is observed with nitrite, which, like imidazole, also shows a biphasic inhibition plot; however, nitrite inhibits gFlHb at sub-millimolar concentrations while Hmp is not significantly affected. NADH oxidase activity measured under aerobic conditions in the absence of nitric oxide for Hmp was more than twice the activity of gFlHb. The addition of 1 mM hydrogen peroxide in these assays stimulated the NADH oxidase activity of gFlHb but not Hmp. Both enzymes had nearly identical cytochrome c reductase activities but the extent of the contribution of indirect reduction by flavohemoglobin-generated superoxide was much lower with gFlHb (4% SOD-inhibited) than with Hmp (17% SOD-inhibited). Although the active sites of the two enzymes share the same highly conserved residues that are important for catalysis, differences in the distal ligand binding site may account for these differences in activity and sensitivity towards NOD inhibitors. The differences observed in the NADH oxidase and cytochrome c reductase assays suggest that gFlHb may have evolved to protect the protist, which lacks both superoxide dismutase and catalase, from the damaging effects of superoxide by minimizing its production and from peroxide by actively reducing it.

2.
PLoS Pathog ; 18(9): e1010840, 2022 09.
Article in English | MEDLINE | ID: mdl-36166467

ABSTRACT

Giardia duodenalis causes giardiasis, a major diarrheal disease in humans worldwide whose treatment relies mainly on metronidazole (MTZ) and albendazole (ABZ). The emergence of ABZ resistance in this parasite has prompted studies to elucidate the molecular mechanisms underlying this phenomenon. G. duodenalis trophozoites convert ABZ into its sulfoxide (ABZSO) and sulfone (ABZSOO) forms, despite lacking canonical enzymes involved in these processes, such as cytochrome P450s (CYP450s) and flavin-containing monooxygenases (FMOs). This study aims to identify the enzyme responsible for ABZ metabolism and its role in ABZ resistance in G. duodenalis. We first determined that the iron-containing cofactor heme induces higher mRNA expression levels of flavohemoglobin (gFlHb) in Giardia trophozoites. Molecular docking analyses predict favorable interactions of gFlHb with ABZ, ABZSO and ABZSOO. Spectral analyses of recombinant gFlHb in the presence of ABZ, ABZSO and ABZSOO showed high affinities for each of these compounds with Kd values of 22.7, 19.1 and 23.8 nM respectively. ABZ and ABZSO enhanced gFlHb NADH oxidase activity (turnover number 14.5 min-1), whereas LC-MS/MS analyses of the reaction products showed that gFlHb slowly oxygenates ABZ into ABZSO at a much lower rate (turnover number 0.01 min-1). Further spectroscopic analyses showed that ABZ is indirectly oxidized to ABZSO by superoxide generated from the NADH oxidase activity of gFlHb. In a similar manner, the superoxide-generating enzyme xanthine oxidase was able to produce ABZSO in the presence of xanthine and ABZ. Interestingly, we find that gFlHb mRNA expression is lower in albendazole-resistant clones compared to those that are sensitive to this drug. Furthermore, all albendazole-resistant clones transfected to overexpress gFlHb displayed higher susceptibility to the drug than the parent clones. Collectively these findings indicate a role for gFlHb in ABZ conversion to its sulfoxide and that gFlHb down-regulation acts as a passive pharmacokinetic mechanism of resistance in this parasite.


Subject(s)
Anthelmintics , Giardia lamblia , Albendazole/chemistry , Albendazole/pharmacokinetics , Animals , Anthelmintics/pharmacology , Biotransformation , Chromatography, Liquid , Cytochromes/metabolism , Flavins/metabolism , Giardia lamblia/genetics , Giardia lamblia/metabolism , Heme/metabolism , Humans , Iron , Metronidazole/pharmacology , Mixed Function Oxygenases/metabolism , Molecular Docking Simulation , RNA, Messenger/metabolism , Sulfones , Sulfoxides/metabolism , Superoxides , Tandem Mass Spectrometry , Trophozoites/metabolism , Xanthine Oxidase/metabolism , Xanthines
3.
Polymers (Basel) ; 14(3)2022 Jan 19.
Article in English | MEDLINE | ID: mdl-35160373

ABSTRACT

A series of films containing chitosan (CS), eggshell membrane (ESM), soluble eggshell membrane (SEP), and plant extracts from Thymus vulgaris and Origanum valgare were prepared with varying concentrations and compositions. These novel films were characterized extensively with respect to film thickness and uniformity, solution absorption, degradation, microenvironmental pH, and antibacterial properties. All the films were flexible with appropriate mechanical stability. After 48 h of soaking in a lysozyme solution, all the films degraded 64 ± 4%, which would be expected to allow for the release of the plant extracts. The plant extracts on their own showed a pH of approximately 4, with the blended films having microenvironmental pHs from approximately 6.4-7.0, which would be expected to promote wound healing. A CS-ESM-SEP film with 5% of each plant extract inhibited almost all E. coli growth in liquid cultures and had no detriments to fluid absorption. Fluid absorption was approximately 100-150% by weight for all the films. The incorporation of SEP and plant extracts to a CS-ESM film provides a promising and novel method for the incorporation of SEP and antibacterial agents in a film with no detriment to wound fluid absorption or film degradation.

4.
Protein Expr Purif ; 157: 70-85, 2019 05.
Article in English | MEDLINE | ID: mdl-30708035

ABSTRACT

NMR is an important method in the structural and functional characterization of proteins, but such experiments typically require isotopic labelling because of the low natural abundance of the nuclei of interest. Isotope-labelled protein for NMR experiments is typically obtained from IPTG-inducible bacterial expression systems in a minimal media that contains labelled carbon or nitrogen sources. Optimization of expression conditions is crucial yet challenging; large amounts of labelled protein are desired, yet protein yields are lower in minimal media, while the labelled precursors are expensive. Faced with these challenges there is a growing body of literature that apply innovative methods of induction to optimize the yield of isotope-labelled protein. A promising technique is lactose-driven auto-induction as it mitigates user intervention and can lead to higher protein yields. This review assesses the current advances and limitations surrounding the ability of researchers to isotope label proteins using auto-induction, and it identifies key components for optimization.


Subject(s)
Isotope Labeling/methods , Nuclear Magnetic Resonance, Biomolecular/methods , Proteins/chemistry , Animals , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Lac Operon , Lactose/genetics , Lactose/metabolism , Proteins/genetics , Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
5.
J Biol Inorg Chem ; 22(7): 1099-1108, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28884403

ABSTRACT

Flavohemoglobins are microbial enzymes that counter nitrosative stress, but the details of their underlying enzymatic activities and structure-function relationships are not completely understood. These enzymes have been identified in Gram-negative bacteria, certain fungi, and the parasitic protist Giardia intestinalis (gFlHb) which, despite lacking the ability to make heme, encodes several hemeproteins. To gain knowledge about the biophysical properties of the active site of gFlHb, we used resonance Raman spectroscopy to probe the wild-type protein and variants at globin domain positions E11, E7, and B10 on the distal, ligand-binding side of the heme. The heme of gFlHb has a peroxidase-like environment resembling that of the well-characterized E. coli flavohemoglobin HMP. We provide evidence that gFlHb has two Fe-His stretching modes, a feature that also occurs in type I/II-peroxidases in which a proximal histidine with strong imidazolate character and a nearby carboxylic acid residue can exist as a tautomeric pair depending on the position of a shared proton. Characterization of the distal variants Tyr30Phe, Gln54Leu, and Leu59Ala shows that TyrB10 and GlnE7 but not LeuE11 are implicated in stabilisation of bound exogenous ligands such as CO and O2. Our work revealed that several biophysical properties of the heme active site of gFlHb are highly conserved compared to HMP and suggest that they are conserved across the flavohemoglobin family.


Subject(s)
Giardia lamblia/enzymology , Hemeproteins/chemistry , Peroxidases/chemistry , Carbon Monoxide/metabolism , Catalytic Domain , Giardia lamblia/chemistry , Giardia lamblia/metabolism , Giardiasis/parasitology , Hemeproteins/metabolism , Humans , Models, Molecular , Oxygen/metabolism , Peroxidases/metabolism , Spectrum Analysis, Raman
6.
J Inorg Biochem ; 158: 110-114, 2016 05.
Article in English | MEDLINE | ID: mdl-27048807

ABSTRACT

Although it lacks mitochondria and the ability to synthesize heme, the protozoan parasite Giardia intestinalis encodes several heme proteins. This includes four members of the cytochrome b5 family, three of which are of similar size to mammalian cytochromes b5 but with reduction potentials that are 140 to 180mV lower. While no structures have yet been determined for any of these proteins, homology modeling points to an increase in heme pocket polarity as a reason for their low potentials. To test this we measured the reduction potentials of four mutants of Giardia cytochrome b5 isotype-I (gCYTB5-I) in which polar residues at two candidate positions (C84, Y51) in the heme pocket were changed to nonpolar ones (C84A, C84F; Y51L, Y51F). All mutants were expressed with comparable levels of heme incorporation and had UV-visible spectra consistent with low spin bis-histidyl coordination. These mutations increased the reduction potential by 18 to 57mV and highlight the influence of C84, which is a residue unique to gCYTB5-I and whose mutation to alanine caused the largest increase. The influence of these two residues plus that of Y61 reported previously accounts for much of the reduction potential difference between gCYTB5-I and microsomal cytochrome b5. A complementary triple mutant of the latter with the hydrophilic residues found in gCYTB5-I bound heme less effectively but nonetheless had a reduction potential that was 135mV lower than wild type.


Subject(s)
Cytochromes b5/chemistry , Cytochromes b5/metabolism , Giardia lamblia/metabolism , Animals , Cattle , Heme/chemistry , Heme/metabolism , Hemeproteins/chemistry , Hemeproteins/metabolism , Hydrophobic and Hydrophilic Interactions , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Secondary , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism
7.
Exp Parasitol ; 157: 197-201, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26299244

ABSTRACT

Despite lacking mitochondria and a known pathway for heme biosynthesis the micro-aerotolerant anaerobic protozoan parasite Giardia intestinalis encodes four members of the cytochrome b5 family of electron transfer proteins, three of which are small, single-domain proteins. While these are similar in size and fold to their better-known mammalian counterparts the Giardia proteins have distinctly lower reduction potentials, ranging from -140 to -171 mV compared to +6 mV for the bovine microsomal protein. This difference is accounted for by a more polar heme environment in the Giardia proteins, as mutation of a conserved heme pocket tyrosine residue to phenylalanine in the Giardia cytochrome b5 isotype-I (gCYTb5-I Y61F) raises its reduction potential by nearly 100 mV. All three isotypes have UV-visible spectra consistent with axial coordination of the heme by a pair of histidine residues, but electron paramagnetic spectroscopy indicates that the planes of their imidazole rings are nearly perpendicular rather than coplanar as observed in mammalian cytochrome b5, which may be due to geometrical constraints imposed by a one-residue shorter spacing between the ligand pair in the Giardia proteins. Although no function has yet to be ascribed to any Giardia cytochrome b5, the presence of similar sequences in many other eukaryotes indicates that these represent an under-characterized class of low reduction potential family members.


Subject(s)
Cytochromes b5/chemistry , Giardia lamblia/chemistry , Animals , Binding Sites , Cattle , Cytochromes b5/metabolism , Dielectric Spectroscopy , Heme/metabolism , Molecular Structure , Oxidation-Reduction , Protein Folding
8.
Exp Parasitol ; 159: 13-23, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26297679

ABSTRACT

Among the few organisms that cannot make the iron cofactor heme, some nonetheless possess heme proteins. This includes the protozoan parasite Giardia intestinalis, which encodes five known heme proteins: a flavohemoglobin and four members of the cytochrome b5 family. Giardia flavohemoglobin closely resembles those of the Enterobacteriaceae in structure and function, acting as a nitric oxide dioxygenase that is induced when trophozoites are exposed to reactive nitrogen species. The Giardia cytochromes b5 are soluble proteins having relatively low reduction potentials and lack several features that are expected to promote rapid electron transfer with redox partners. Only one potential electron donor, and no electron acceptors, have yet been identified in the Giardia genome, and the roles of these cytochromes are presently unknown. The answer may lie in the sequences that flank the heme-binding core of these proteins which could serve to localize them within the cell through reversible post-translational modifications and to promote specific protein-protein interactions.


Subject(s)
Giardia lamblia/chemistry , Hemeproteins/metabolism , Amino Acid Sequence , Animals , Escherichia coli/chemistry , Giardia lamblia/physiology , Heme/metabolism , Hemeproteins/chemistry , Humans , Models, Structural , Molecular Sequence Data , Sequence Alignment , Structural Homology, Protein , Yeasts/chemistry
9.
Metallomics ; 4(12): 1255-61, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23151674

ABSTRACT

The protozoan intestinal parasite Giardia lamblia lacks mitochondria and the ability to make haem yet encodes several putative haem-binding proteins, including three of the cytochrome b(5) family. We cloned one of these (gCYTb5-I) and expressed it within Escherichia coli as a soluble holoprotein. UV-visible and resonance Raman spectra of gCYTb5-I resemble those of microsomal cytochrome b(5), and homology modelling supports a structure in which a pair of invariant histidine residues act as axial ligands to the haem iron. The reduction potential of gCYTb5-I is -165 mV vs. SHE and is relatively low compared to most values (-110 to +80 mV) for this class of protein. The amino- and carboxy-terminal sequences that flank the central haem-binding core of the Giardia cytochromes are highly charged and differ from those of other family members. A core gCYTb5-I variant lacking these flanking sequences was also able to bind haem. The presence of one actual and two probable functional cytochromes b(5) in Giardia is evidence of uncharacterized cytochrome-mediated metabolic processes within this medically important protist.


Subject(s)
Cytochromes b5/metabolism , Giardia lamblia/metabolism , Protozoan Proteins/metabolism , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Cytochromes b5/chemistry , Cytochromes b5/genetics , DNA, Protozoan/genetics , Electrochemical Techniques , Genes, Protozoan , Giardia lamblia/genetics , Giardia lamblia/pathogenicity , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Spectrophotometry, Ultraviolet , Spectrum Analysis, Raman
10.
Arch Biochem Biophys ; 506(2): 165-72, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21147059

ABSTRACT

Nitric oxide synthases (NOSs) share two invariant tryptophan residues within a conserved helical lariat that is part of the pterin-binding site and dimer interface. We mutated Staphylococcus aureus NOS Trp-314 (to alanine, phenylalanine, tyrosine and histidine) and Trp-316 (to alanine, phenylalanine and tyrosine) and characterized the effects of mutation on heme environment, quaternary structure, enzymatic activity, and substrate affinity. With arginine present, all saNOS variants bound heme with native thiolate ligation, formed high spin ferric complexes and were dimeric. All variants catalyze the peroxide-dependent oxidation of N-hydroxy-l-arginine, at rates from 10% to 55% of wild type activity. Arginine-free proteins are dimeric with the exception of W314A. Arginine affinity for all variants decreases with increasing temperature between 15 and 42 °C but is precipitous for position-314 variants. Previous structural and biophysical characterization of NOS oxygenase domains demonstrated that the protein can exist in either a tight or loose conformation, with the former corresponding to the active state of the protein. In the position-314 variants it is likely that the loose conformation is favoured, owing to the loss of a hydrogen bond between the indole side chain and the polypeptide backbone of the helical lariat.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/genetics , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Amino Acid Substitution , Arginine/metabolism , Bacterial Proteins/metabolism , Catalytic Domain , Conserved Sequence , Dimerization , Enzyme Activation , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Nitric Oxide Synthase/metabolism , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrophotometry , Substrate Specificity , Tryptophan/chemistry
11.
Environ Toxicol Chem ; 29(8): 1669-77, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20821618

ABSTRACT

Perfluorocarboxylic acids (PFCAs) of chain length greater than seven carbon atoms bioconcentrate in the livers of fish. However, a mechanistic cause for the empirically observed increase in the bioconcentration potential of PFCAs as a function of chain length has yet to be determined. To this end, recombinant rat liver fatty acid-binding protein (L-FABP) was purified, and its interaction with PFCAs was characterized in an aqueous system at pH 7.4. Relative binding affinities of L-FABP with PFCAs of carbon chain lengths of five to nine were established fluorimetrically. The energetics, mechanism, and stoichiometry of the interaction of perfluorooctanoic acid (PFOA) with L-FABP were examined further by isothermal titration calorimetry (ITC) and electrospray ionization combined with tandem mass spectrometry (ESI-MS/MS). Perfluorooctanoic acid was shown to bind to L-FABP with an affinity approximately an order of magnitude less than the natural ligand, oleic acid, and to have at least 3:1 PFOA:L-FABP stoichiometry. Two distinct modes of PFOA binding to L-FABP were observed by ESI-MS/MS analysis; in both cases, PFOA binds solely as the neutral species under typical physiological pH and aqueous concentrations of the anion. A comparison of their chemical and physical properties with other well-studied biologically relevant chemicals showed that accumulation of PFCAs in proteins as the neutral species is predictable. For example, the interaction of PFOA with L-FABP is almost identical to that of the acidic ionizing drugs ketolac, ibuprofen, and warfarin that show specificity to protein partitioning with a magnitude that is proportional to the K(OW) (octanol-water partitioning) of the neutral species. The experimental results suggest that routine pharmacochemical models may be applicable to predicting the protein-based bioaccumulation of long-chain PFCAs.


Subject(s)
Fluorocarbons/metabolism , Liver/metabolism , Animals , Caprylates/chemistry , Caprylates/metabolism , Carboxylic Acids , Fatty Acid-Binding Proteins/chemistry , Fatty Acid-Binding Proteins/metabolism , Fishes/metabolism , Fluorocarbons/chemistry , Liver/chemistry , Water Pollutants, Chemical/chemistry , Water Pollutants, Chemical/metabolism
12.
Biochem Biophys Res Commun ; 399(3): 347-51, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20655876

ABSTRACT

Giardia lamblia is a pathogenic protist that infects the small intestine of mammals. As a facultative anaerobe, Giardia obtains all of its energy by substrate-level phosphorylation, lacks a functioning respiratory chain, and is not thought to require heme. However, sequencing of the G. lamblia genome has identified several putative heme proteins, one of which shares high sequence similarity to flavohemoglobins found in bacteria and some single-celled eukaryotes. We have cloned and characterized the functional properties of the G. lamblia flavohemoglobin. The protein is monomeric, binds heme and flavin adenine dinucleotide, and exhibits similar behavior to known flavohemoglobins, including NADH and NADPH oxidase activity, which is stimulated by addition of the nitric oxide donor DEA/NO. Based on its structural and functional properties, the likely role of this protein is to protect Giardia against oxygen, nitric oxide, or both. The presence of a Giardia gene encoding a functional heme protein raises questions on how this organism acquires the heme cofactor, which hitherto have been unexplored.


Subject(s)
Giardia lamblia/metabolism , Hemeproteins/chemistry , Amino Acid Sequence , Flavin-Adenine Dinucleotide/metabolism , Genetic Code , Giardia lamblia/genetics , Heme/metabolism , Hemeproteins/genetics , Molecular Sequence Data , Protein Structure, Tertiary
13.
J Inorg Biochem ; 103(7): 1102-12, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19539996

ABSTRACT

The proximal ligand of thiolate-coordinated heme proteins is crucial for the activation of the oxygen molecule and hydroxylation of substrates. In nitric oxide synthases (NOSs), the heme axial cysteine ligand forms a hydrogen bond to the side chain indole nitrogen of a tryptophan residue. Resonance Raman spectroscopy was used to probe W56F and W56Y variants of the NOS of Staphylococcus aureus (saNOS) and the analogous W180 variants of the endothelial NOS oxygenase domain (eNOSox). We show that the variants displayed lower nu(Fe-NO) and nu(Fe-CO) frequencies indicating that these mutations increased the electron density on the axial cysteine in their Fe(III)NO and Fe(II)CO complexes. We also show by UV-visible spectroscopy that the Fe(II)CO complexes of the variants displayed a red-shifted Soret optical transition in addition to the lower nu(Fe-CO) thus establishing that these properties are sensitive indicators of the modulation of the basicity of the axial cysteine. We infer, based on its spectroscopic properties, that ferrous eNOSox W180Y saturated with l-arginine and tetrahydrobiopterin forms a tyrosine-cysteine hydrogen bond when bound to CO. Evidence for such a hydrogen bond was not obtained for the Fe(III)NO protein nor for the analogous saNOS variant. These mutations reveal interesting differences in the response of NOS isotypes to analogous mutations at conserved residues and clearly show that the heme-Fe to cysteine sigma bond is modulated by the Cys-Trp hydrogen bond in NOSs. These studies serve as a basis to gain information on the role played by this hydrogen bond in oxygen activation in this class of enzymes.


Subject(s)
Cysteine/chemistry , Heme/chemistry , Nitric Oxide Synthase Type III/chemistry , Nitric Oxide/metabolism , Staphylococcus aureus/enzymology , Tryptophan/chemistry , Animals , Cloning, Molecular , Endothelium/enzymology , Endothelium/metabolism , Hydrogen Bonding , Mutation , Nitric Oxide/chemistry , Nitric Oxide Synthase Type III/genetics , Spectrum Analysis, Raman , Tryptophan/genetics
14.
Biochemistry ; 44(7): 2276-83, 2005 Feb 22.
Article in English | MEDLINE | ID: mdl-15709740

ABSTRACT

The inducible nitric oxide synthase core oxygen domain (iNOS(COD)) is a homodimeric protein complex of ca. 100 kDa. In this work, the subunit disassembly and unfolding of the protein following a pH jump from 7.5 to 2.8 were monitored by on-line rapid mixing in conjunction with electrospray (ESI) time-of-flight mass spectrometry. Various protein species become populated during the denaturation process. These can be distinguished by their ligand binding behavior, and by the different charge states that they produce during ESI. Detailed intensity-time profiles were obtained for all of these species, and the kinetics were subjected to a global analysis which allows a model of the denaturation process to be developed. The data are described well by three relaxation times (tau(1) = 0.36 s, tau(2) = 0.62 s, and tau(3) = 3.3 s), each of which has a characteristic amplitude spectrum. The initial step of the reaction is the disruption of the iNOS(COD) dimer, to generate heme-bound monomeric species in various degrees of unfolding. This first step is accompanied by the loss of two tetrahydrobiopterin cofactors. Subsequent heme loss generates monomeric apoproteins exhibiting various degrees of unfolding. In addition, the formation of proteins that are bound to two heme groups is observed. A subpopulation of holo monomers undergoes substantial unfolding while retaining contact with the heme cofactor. Together with previous studies, the results of this work suggest that the occurrence of complex reaction mechanisms involving several short-lived intermediates is a common feature for the denaturation of large multiprotein complexes.


Subject(s)
Nitric Oxide Synthase/chemistry , Oxygenases/chemistry , Protein Folding , Animals , Dimerization , Heme Oxygenase (Decyclizing)/chemistry , Heme Oxygenase (Decyclizing)/metabolism , Hydrogen-Ion Concentration , Kinetics , Mice , Models, Chemical , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Oxygenases/metabolism , Protein Conformation , Protein Denaturation , Protein Structure, Tertiary , Spectrometry, Mass, Electrospray Ionization/methods , Time Factors
15.
J Am Soc Mass Spectrom ; 15(5): 629-38, 2004 May.
Article in English | MEDLINE | ID: mdl-15121191

ABSTRACT

The investigation of protein quaternary structure, protein-cofactor, and protein-ligand interactions by mass spectrometry is often limited by the fragility of such interactions under experimental conditions. To develop more gentle conditions of perhaps general use, we used as a model for study the oxygenase domain of murine inducible nitric oxide synthase (iNOS), which is homodimeric, binds heme and tetrahydrobiopterin H(4)B cofactors, and the substrate L-arginine. The energetics of the collisions in q2 and in the lens region of the mass spectrometer were manipulated for varying the degree of solvation around the non-covalently bound ions. Furthermore, the number of low-energy collisions in the collision cell of the instrument was varied, focusing and dampening the ion beam. Under gentle source collision conditions, and using multiple low-energy collisions in the collision cell of the mass spectrometer, dimers of the iNOS oxygenase domain containing heme, H(4)B, and arginine were observed intact after electrospraying at pH values near neutrality; a mutant of this protein (Trp188 --> Phe) was monomeric and did not bind cofactors. The pH dependence of the iNOS oxygenase domain under acidic conditions was also studied; while heme remained bound to the protein between pH 2.5 and 4.0, the dimeric structure was disrupted. Our findings confirm that non-covalently bound macromolecular complexes are retained and observable using electrospray mass spectrometry under the appropriate experimental conditions.


Subject(s)
Nitric Oxide Synthase/chemistry , Oxygenases/chemistry , Cold Temperature , Dimerization , Hydrogen-Ion Concentration , Mutation , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Oxygenases/genetics , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Solutions/chemistry
16.
Protein Expr Purif ; 27(1): 115-27, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12509993

ABSTRACT

A limitation of site-directed mutagenesis of homodimeric proteins is that both subunits will carry the same mutation. We have devised a way to prepare mixed dimers, in which only one chain bears a desired mutation, or each chain can bear a different mutation. Using the inducible nitric oxide oxygenase domain as a model, our strategy focused on the co-expression of two differentially tagged versions of the oxygenase domain, with isolation of the desired mixed dimer in two chromatography steps. We evaluated expression vectors encoding polyhistidine (His(6)), cellulose binding domain, glutathione-S-transferase, and polyglutamate (Glu(7))-tagged versions of the oxygenase domain for satisfactory levels of soluble protein expression and for their ability to form mixed dimers. The combination of His(6)- and Glu(7)-tagged subunits was successful in both respects, and the mixed dimers could be separated from either form of homodimer by sequential immobilized metal affinity chromatography and anion exchange chromatography. The UV-Vis spectrum, substrate binding properties, and enzymatic activity were not altered in the mixed dimer wild-type (His(6)/Glu(7)) compared to the two homodimers (His(6)/His(6) and Glu(7)/Glu(7)). We then characterized a mixed dimer variant in which one chain contained an E371A substitution (which prevents binding of the substrate L-arginine) while the other subunit was left unaltered. This species binds L-arginine and has about one-half the activity of the wild-type homodimer. Mutants known to destabilize the iNOS dimer (E411A, D454A, and W188F) were also investigated; in these cases co-expression with the wild-type subunit did not lead to the formation of stable mixed dimers.


Subject(s)
Catalytic Domain , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/metabolism , Oxygenases/chemistry , Oxygenases/metabolism , Dimerization , Models, Molecular , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/isolation & purification , Nitric Oxide Synthase Type II , Oxygenases/genetics , Oxygenases/isolation & purification , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrum Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...