Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Dis Esophagus ; 29(1): 34-40, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25327568

ABSTRACT

This study aimed to study the factors that are associated with urgent esophagectomy for the treatment of esophageal perforations and the impact of this therapy. A retrospective review of all esophageal perforations treated at a tertiary care hospital from January 1984 to January 2012 was performed. Compiling demographics, cause and site of perforations, time to presentation, comorbidities, radiological tests, the length of perforation, the hemodynamic status of the patient, type of treatment required, and outcomes were performed. Univariate, multivariate, and Cox regression analyses were conducted. Of 127 cases of esophageal perforation, it was spontaneous in 44 (35%), iatrogenic in 53 (44%), foreign body ingestion in 22 (17%), and traumatic perforation in 7 (6%) cases. Overall, 85 of the 127 (67%) patients were managed operatively, 35 (27.6%) patients were treated conservatively, and 7 (6.3%) patients were treated by endoscopic stent placement. Of the 85 patients who were managed operatively, 21 (16.5%) required esophagectomies, 13 (15.3%) had esophagectomy with immediate reconstruction, 5 (5.9%) patients had esophagectomy followed by delayed reconstruction, and 3 (3.5%) patients failed primary repair and required an esophagectomy as a secondary definitive procedure. Multivariate analysis revealed that esophagectomy in esophageal perforations was associated with the presence of benign or malignant esophageal stricture (P = 0.001) and a perforation >5 cm (P = 0.001). Mortality was mainly associated with the presence of a benign or malignant esophageal stricture (P = 0.04). The presence of pre-existing benign or malignant stricture or large perforation (>5 cm) is associated with the need for an urgent esophagectomy with or without immediate reconstruction. Performing esophagectomy was not found to be a significant prognosticator for mortality.


Subject(s)
Esophageal Perforation , Esophagectomy , Adult , Aged , Aged, 80 and over , Esophageal Perforation/diagnosis , Esophageal Perforation/etiology , Esophageal Perforation/mortality , Esophageal Perforation/physiopathology , Esophageal Perforation/surgery , Esophageal Stenosis/complications , Esophageal Stenosis/diagnosis , Esophagectomy/adverse effects , Esophagectomy/instrumentation , Esophagectomy/methods , Esophagectomy/statistics & numerical data , Esophagus/diagnostic imaging , Esophagus/surgery , Female , Humans , Male , Middle Aged , Radiography , Retrospective Studies , Saudi Arabia/epidemiology , Severity of Illness Index , Stents , Time-to-Treatment/statistics & numerical data
2.
J Gastrointest Surg ; 13(7): 1245-50, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19296183

ABSTRACT

OBJECTIVE: The objective was to study the factors associated with mortality in mesenteric venous thrombosis (MVT). METHODS: We reviewed all cases of bowel ischemia at our institute from 1984 to 2004 and identified 31 cases of MVT and compiled data concerning their demographics, risk factors, investigations, management, surgical procedures, and outcomes. Survival was analyzed for both 30-day and 5-year periods. RESULTS: Analysis of factors associated with mortality in our 31 case series revealed that 30-day mortality was strongly associated with colonic involvement in ischemia (p = .008) as well as short bowel syndrome (p = .028) and possibly failure to anti-coagulate the patient (p = .07). While 5-year mortality was strongly associated with "short bowel syndrome" as defined by small bowel remaining less than 100 cm (p = .031). Further study using a multivariate Cox proportional hazard analysis showed that mortality within the 30-day period was mainly related to colon ischemia with p value of .014 and an odds ratio of 17.4, while short-bowel syndrome was the predominated factor in the 5-year mortality analysis with a p value of .029 and an odds ratio of 5. CONCLUSION: Thirty-day mortality for MVT is strongly associated with colonic involvement as well as "short-bowel" syndrome, while anticoagulation may be protective. Five-year survival was found to be strongly associated with "short-bowel" syndrome.


Subject(s)
Cause of Death , Hospital Mortality/trends , Mesenteric Vascular Occlusion/mortality , Mesenteric Vascular Occlusion/surgery , Mesenteric Veins , Adult , Age Distribution , Aged , Cohort Studies , Colon/blood supply , Female , Follow-Up Studies , Humans , Incidence , Intestine, Small/blood supply , Logistic Models , Male , Mesenteric Vascular Occlusion/diagnostic imaging , Middle Aged , Phlebography/methods , Probability , Proportional Hazards Models , Registries , Retrospective Studies , Risk Factors , Sex Distribution , Survival Rate , Thrombectomy/adverse effects , Thrombectomy/methods , Time Factors , Treatment Outcome , Venous Thrombosis/diagnostic imaging , Venous Thrombosis/mortality , Venous Thrombosis/surgery , Young Adult
3.
Biochem Soc Trans ; 31(Pt 3): 502-5, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12773144

ABSTRACT

The nitric oxide synthases are large, modular, dimeric enzymes composed of a reductase domain, which is related to cytochrome P450 reductase, and a structurally unique oxygenase domain containing a Cys-ligated haem. Both the neuronal and endothelial isoforms are activated by the reversible binding of calmodulin (CaM) at elevated intracellular Ca(2+) levels to produce NO as part of a number of cell signalling pathways. CaM binds to the linker region between the two domains and activates the enzyme by inducing intramolecular electron transfer. Protein-engineering experiments have shown that a series of unusual autoinhibitory inserts found only in the CaM-dependent NOS isoforms control both CaM binding and the structural rearrangement it induces. These lie in the reductase domain of the enzyme and include a 40-amino-acid autoinhibitory loop in the FMN-binding module, a 30-amino-acid extension to the C-terminus and the CaM-binding site itself. The substrate (NADPH) also plays an important role in defining the CaM-dependence of the reductase domain by inducing a tight conformational lock in the absence of CaM. Both the substrate and the conformational lock appear to be released on CaM binding; the resultant domain mobility leads to activation.


Subject(s)
Calmodulin/physiology , Nitric Oxide Synthase/metabolism , Animals , Homeostasis , Kinetics , Mammals , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type I
4.
J Biol Chem ; 276(43): 39864-71, 2001 Oct 26.
Article in English | MEDLINE | ID: mdl-11518705

ABSTRACT

Electron transfer within rat neuronal nitric-oxide synthase (nNOS) was investigated by pulse radiolysis. Radiolytically generated 1-methyl-3-carbamoyl pyridinium (MCP) radical was found to react predominantly with the heme of the enzyme with a second-order rate constant for heme reduction of 3 x 10(8) m(-1) s(-1). In the calmodulin (CaM)-bound enzyme a subsequent first-order phase was observed which had a rate constant of 1.2 x 10(3) s(-1). In the absence of CaM, this phase was absent. Kinetic difference spectra for nNOS reduction indicated that the second phase consisted of heme reoxidation accompanied by formation of a neutral flavin semiquinone, suggesting that it is heme to flavin electron transfer. Experiments with the heme proximal surface mutant, K423E, had no second phase, confirming that the mutation blocks interdomain electron transfer. With the autoinhibitory loop deletion mutant, Delta40, the slow phase was observed even in the absence of CaM consistent with the role of the loop in impeding interdomain electron transfer. The rate of heme to FMN electron transfer observed in the wild-type enzyme is approximately 1000 times faster than the FMN to heme electron transfer rate predicted during catalysis from kinetic modeling, suggesting that the catalytic process is slowed by kinetic gating.


Subject(s)
Calmodulin/metabolism , Niacinamide/analogs & derivatives , Nitric Oxide Synthase/metabolism , Electron Transport , Free Radicals/metabolism , Mutation , Niacinamide/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I , Peptide Fragments , Protein Structure, Tertiary , Pulse Radiolysis
5.
J Biol Chem ; 276(32): 30036-42, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11395516

ABSTRACT

In neuronal nitric-oxide synthase (nNOS), calmodulin (CaM) binding is thought to trigger electron transfer from the reductase domain to the heme domain, which is essential for O(2) activation and NO formation. To elucidate the electron-transfer mechanism, we characterized a series of heterodimers consisting of one full-length nNOS subunit and one oxygenase-domain subunit. The results support an inter-subunit electron-transfer mechanism for the wild type nNOS, in that electrons for catalysis transfer in a Ca(2+)/CaM-dependent way from the reductase domain of one subunit to the heme of the other subunit, as proposed for inducible NOS. This suggests that the two different isoforms form similar dimeric complexes. In a series of heterodimers containing a Ca(2+)/CaM-insensitive mutant (delta40), electrons transferred from the reductase domain to both hemes in a Ca(2+)/CaM-independent way. Thus, in the delta40 mutant electron transfer from the reductase domains to the heme domains can occur via both inter-subunit and intra-subunit mechanisms. However, NO formation activity was exclusively linked to inter-subunit electron transfer and was observed only in the presence of Ca(2+)/CaM. This suggests that the mechanism of activation of nNOS by CaM is not solely dependent on the activation of electron transfer to the nNOS hemes but may involve additional structural factors linked to the catalytic action of the heme domain.


Subject(s)
Calmodulin/pharmacology , Electrons , Neurons/enzymology , Nitric Oxide Synthase/chemistry , Animals , Calmodulin/metabolism , Catalysis/drug effects , Chromatography, Gel , DNA, Complementary/metabolism , Dimerization , Electron Transport , Gene Deletion , Heme/chemistry , Models, Biological , Mutagenesis, Site-Directed , Mutation , Nitric Oxide/metabolism , Oxygenases/chemistry , Plasmids/metabolism , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , Rats , Spectrophotometry
6.
Biochem Soc Trans ; 29(Pt 2): 147-52, 2001 May.
Article in English | MEDLINE | ID: mdl-11356143

ABSTRACT

The nitric oxide synthases (NOSs) are dimeric flavocytochromes consisting of an oxygenase domain with cytochrome P450-like Cys-ligated haem, coupled to a diflavin reductase domain, which is related to cytochrome P450 reductase. The NOSs catalyse the sequential mono-oxygenation of arginine to N-hydroxyarginine and then to citrulline and NO. The constitutive NOS isoforms (cNOSs) are regulated by calmodulin (CaM), which binds at elevated concentrations of free Ca(2+), whereas the inducible isoform binds CaM irreversibly. One of the main structural differences between the constitutive and inducible isoforms is an insert of 40-50 amino acids in the FMN-binding domain of the cNOSs. Deletion of the insert in rat neuronal NOS (nNOS) led to a mutant enzyme which binds CaM at lower Ca(2+) concentrations and which retains activity in the absence of CaM. In order to resolve the mechanism of action of CaM activation we determined reduction potentials for the FMN and FAD cofactors of rat nNOS in the presence and absence of CaM using a recombinant form of the reductase domain. The results indicate that CaM binding does not modulate the reduction potentials of the flavins, but appears to control electron transfer primarily via a large structural rearrangement. We also report the creation of chimaeric enzymes in which the reductase domains of nNOS and flavocytochrome P450 BM3 (Bacillus megaterium III) have been exchanged. Despite its very different flavin redox potentials, the BM3 reductase domain was able to support low levels of CaM-dependent NO synthesis, whereas the NOS reductase domain did not effectively substitute for that of cytochrome P450 BM3.


Subject(s)
Bacterial Proteins , Electron Transport , Neurons/enzymology , Nitric Oxide Synthase/metabolism , Amino Acid Substitution/genetics , Animals , Bacillus megaterium/enzymology , Bacillus megaterium/genetics , Calmodulin/metabolism , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Flavin Mononucleotide/metabolism , Flavin-Adenine Dinucleotide/metabolism , Kinetics , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , NADP/metabolism , NADPH-Ferrihemoprotein Reductase , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I , Oxidation-Reduction , Protein Binding , Protein Structure, Tertiary , Rabbits , Rats , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion/genetics , Structure-Activity Relationship
7.
Biochemistry ; 40(7): 1956-63, 2001 Feb 20.
Article in English | MEDLINE | ID: mdl-11329262

ABSTRACT

Midpoint reduction potentials for the flavin cofactors in human NADPH-cytochrome P450 oxidoreductase were determined by anaerobic redox titration of the diflavin (FAD and FMN) enzyme and by separate titrations of its isolated FAD/NADPH and FMN domains. Flavin reduction potentials are similar in the isolated domains (FAD domain E(1) [oxidized/semiquinone] = -286 +/- 6 mV, E(2) [semiquinone/reduced] = -371 +/- 7 mV; FMN domain E(1) = -43 +/- 7 mV, E(2) = -280 +/- 8 mV) and the soluble diflavin reductase (E(1) [FMN] = -66 +/- 8 mV, E(2) [FMN] = -269 +/- 10 mV; E(1) [FAD] = -283 +/- 5 mV, E(2) [FAD] = -382 +/- 8 mV). The lack of perturbation of the individual flavin potentials in the FAD and FMN domains indicates that the flavins are located in discrete environments and that these environments are not significantly disrupted by genetic dissection of the domains. Each flavin titrates through a blue semiquinone state, with the FMN semiquinone being most intense due to larger separation (approximately 200 mV) of its two couples. Both the FMN domain and the soluble reductase are purified in partially reduced, colored form from the Escherichia coli expression system, either as a green reductase or a gray-blue FMN domain. In both cases, large amounts of the higher potential FMN are in the semiquinone form. The redox properties of human cytochrome P450 reductase (CPR) are similar to those reported for rabbit CPR and the reductase domain of neuronal nitric oxide synthase. However, they differ markedly from those of yeast and bacterial CPRs, pointing to an important evolutionary difference in electronic regulation of these enzymes.


Subject(s)
NADPH-Ferrihemoprotein Reductase/metabolism , Electron Transport , Flavin Mononucleotide/analogs & derivatives , Flavin Mononucleotide/chemistry , Flavin Mononucleotide/metabolism , Flavin-Adenine Dinucleotide/chemistry , Flavin-Adenine Dinucleotide/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/isolation & purification , Isoenzymes/metabolism , NADPH-Ferrihemoprotein Reductase/chemistry , NADPH-Ferrihemoprotein Reductase/isolation & purification , Oxidation-Reduction , Potentiometry , Protein Structure, Tertiary , Reproducibility of Results , Solubility
8.
J Biol Chem ; 276(23): 20190-6, 2001 Jun 08.
Article in English | MEDLINE | ID: mdl-11285259

ABSTRACT

The midpoint reduction potentials of the FAD cofactor in wild-type Methylophilus methylotrophus (sp. W3A1) electron-transferring flavoprotein (ETF) and the alphaR237A mutant were determined by anaerobic redox titration. The FAD reduction potential of the oxidized-semiquinone couple in wild-type ETF (E'(1)) is +153 +/- 2 mV, indicating exceptional stabilization of the flavin anionic semiquinone species. Conversion to the dihydroquinone is incomplete (E'(2) < -250 mV), because of the presence of both kinetic and thermodynamic blocks on full reduction of the FAD. A structural model of ETF (Chohan, K. K., Scrutton, N. S., and Sutcliffe, M. J. (1998) Protein Pept. Lett. 5, 231-236) suggests that the guanidinium group of Arg-237, which is located over the si face of the flavin isoalloxazine ring, plays a key role in the exceptional stabilization of the anionic semiquinone in wild-type ETF. The major effect of exchanging alphaArg-237 for Ala in M. methylotrophus ETF is to engineer a remarkable approximately 200-mV destabilization of the flavin anionic semiquinone (E'(2) = -31 +/- 2 mV, and E'(1) = -43 +/- 2 mV). In addition, reduction to the FAD dihydroquinone in alphaR237A ETF is relatively facile, indicating that the kinetic block seen in wild-type ETF is substantially removed in the alphaR237A ETF. Thus, kinetic (as well as thermodynamic) considerations are important in populating the redox forms of the protein-bound flavin. Additionally, we show that electron transfer from trimethylamine dehydrogenase to alphaR237A ETF is severely compromised, because of impaired assembly of the electron transfer complex.


Subject(s)
Arginine/metabolism , Benzoquinones/metabolism , Flavoproteins/metabolism , Methylophilus methylotrophus/metabolism , Quinones/metabolism , Base Sequence , DNA Primers , Electron-Transferring Flavoproteins , Flavoproteins/chemistry , Flavoproteins/genetics , Flavoproteins/isolation & purification , Kinetics , Mutagenesis, Site-Directed , Oxidation-Reduction , Potentiometry
9.
Biochem Biophys Res Commun ; 282(5): 1092-7, 2001 Apr 20.
Article in English | MEDLINE | ID: mdl-11302726

ABSTRACT

Neuronal nitric-oxide synthase (nNOS) is composed of a heme oxygenase domain and a flavin-bound reductase domain. Ca(2+)/calmodulin (CaM) is essential for interdomain electron transfer during catalysis, whereas the role of the catalytically important cofactor, tetrahydrobiopterin (H4B) remains elusive. The product NO appears to bind to the heme and works as a feedback inhibitor. The present study shows that the Fe(3+)-NO complex is reduced to the Fe(2+)-NO complex by NADPH in the presence of both l-Arg and H4B even in the absence of Ca(2+)/CaM. The complex could not be fully reduced in the absence of H4B under any circumstances. However, dihydrobiopterin and N(G)-hydroxy-l-Arg could be substituted for H4B and l-Arg, respectively. No direct correlation could be found between redox potentials of the nNOS heme and the observed reduction of the Fe(3+)-NO complex. Thus, our data indicate the importance of the pterin binding to the active site structure during the reduction of the NO-heme complex by NADPH during catalytic turnover.


Subject(s)
Antioxidants/chemistry , Biopterins/analogs & derivatives , Biopterins/physiology , Nitric Oxide Synthase/metabolism , Nitric Oxide/chemistry , Anaerobiosis , Animals , Arginine/chemistry , Biopterins/chemistry , Calcium/metabolism , Calmodulin/metabolism , Catalysis/drug effects , Iron/chemistry , Macromolecular Substances , Models, Chemical , Mutagenesis, Site-Directed , NADP/chemistry , NADP/pharmacology , Nitric Oxide Donors/chemistry , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I , Oxidation-Reduction/drug effects , Rats , Spectrophotometry
10.
Biochem Biophys Res Commun ; 275(3): 752-8, 2000 Sep 07.
Article in English | MEDLINE | ID: mdl-10973794

ABSTRACT

Nitric oxide synthase (NOS) is composed of an oxygenase domain and a reductase domain. The reductase domain has NADPH, FAD, and FMN binding sites. Wild-type nNOS reduced the azo bond of methyl red with a turnover number of approximately 130 min(-1) in the presence of Ca(2+)/calmodulin (CaM) and NADPH under anaerobic conditions. Diphenyleneiodonium chloride (DPI), a flavin/NADPH binding inhibitor, completely inhibited azo reduction. The omission of Ca(2+)/CaM from the reaction system decreased the activity to 5%. The rate of the azo reduction with an FMN-deficient mutant was also 5% that of the wild type. NADPH oxidation rates for the wild-type and mutant enzymes were well coupled with azo reduction. Thus, we suggest that electrons delivered from the FMN of the nNOS enzyme reduce the azo bond of methyl red and that this reductase activity is controlled by Ca(2+)/CaM.


Subject(s)
Azo Compounds/metabolism , Flavin Mononucleotide/metabolism , Nitric Oxide Synthase/metabolism , Animals , Azo Compounds/chemistry , Binding Sites , Calcium/metabolism , Calmodulin/metabolism , Catalysis/drug effects , Electron Spin Resonance Spectroscopy , Flavin Mononucleotide/antagonists & inhibitors , Gas Chromatography-Mass Spectrometry , Kinetics , Mutagenesis , NADP/antagonists & inhibitors , NADP/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I , Onium Compounds/pharmacology , Oxidation-Reduction/drug effects , Oxygen/metabolism , Protein Structure, Tertiary , Rats , Spectrophotometry , Spin Labels
11.
J Biol Chem ; 275(34): 26150-7, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-10846172

ABSTRACT

Nitric-oxide synthase (NOS) requires the cofactor, (6R)-5,6,7, 8-tetrahydrobiopterin (H4B), for catalytic activity. The crystal structures of NOSs indicate that H4B is surrounded by aromatic residues. We have mutated the conserved aromatic acids, Trp(676), Trp(678), Phe(691), His(692), and Tyr(706), together with the neighboring Arg(414) residue within the H4B binding region of full-length neuronal NOS. The W676L, W678L, and F691L mutants had no NO formation activity and had very low heme reduction rates (<0.02 min(-1)) with NADPH. Thus, it appears that Trp(676), Trp(678), and Phe(691) are important to retain the appropriate active site conformation for H4B/l-Arg binding and/or electron transfer to the heme from NADPH. The mutation of Tyr(706) to Leu and Phe decreased the activity down to 13 and 29%, respectively, of that of the wild type together with a dramatically increased EC(50) value for H4B (30-40-fold of wild type). The Tyr(706) phenol group interacts with the heme propionate and Arg(414) amine via hydrogen bonds. The mutation of Arg(414) to Leu and Glu resulted in the total loss of NO formation activity and of the heme reduction with NADPH. Thus, hydrogen bond networks consisting of the heme carboxylate, Tyr(706), and Arg(414) are crucial in stabilizing the appropriate conformation(s) of the heme active site for H4B/l-Arg binding and/or efficient electron transfer to occur.


Subject(s)
Arginine/metabolism , Biopterins/analogs & derivatives , Heme/metabolism , NADP/metabolism , Nitric Oxide Synthase/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Biopterins/metabolism , Catalysis , Dimerization , Drosophila , Glutamine/metabolism , Humans , Hydrogen Bonding , Leucine/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Nitric Oxide Synthase Type I , Oxidation-Reduction , Phenylalanine/metabolism , Rats , Spectrophotometry, Atomic , Structure-Activity Relationship , Tryptophan/metabolism
12.
J Inorg Biochem ; 82(1-4): 163-70, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11132623

ABSTRACT

Nitric oxide synthase (NOS) has an oxygenase domain with a thiol-coordinated heme active side similar to cytochrome P450. In contrast to cytochrome P450, however, conserved aromatic amino acids are situated in the heme proximal side of NOS. For example, in endothelial NOS (eNOS), the indole-ring nitrogen of Trp180 hydrogen-binds to the thiol of Cys186, the internal axial ligand to the heme. And, the aromatic side chain of Trp192 forms a bridge between this residue and the protein. Trp180 and Trp192 of eNOS correspond to Trp409 and Trp421 of neuronal NOS (nNOS), respectively. In order to understand the roles of the aromatic amino acids in catalysis, we generated Trp409His, Trp409Leu, Trp421His and Trp421Leu mutants of nNOS and determined their catalytic parameters. The Trp409Leu mutant was very poorly expressed in E. coli and was easily denatured during purification procedures. The NO formation activities of the Trp409His and Trp421Leu mutants were 11 and 25 micromol/min per micromol heme, respectively, and are lower than that (44 micromol/min per micromol heme) of the wild type. The activity (46 micromol/min per micromol heme) of the Trp421His mutant was comparable to that of the wild-type enzyme. However, NADPH oxidation rates of Trp421His (230 micromol/min per micromol heme) and Trp421Leu (104 micromol/min per microol heme) in the presence of L-Arg were much larger than those observed for the wild type (65 micromol/min per micromol heme) and the Trp409His mutant (43 micromol/min per micromol heme). The cytochrome c reduction rate of the Trp421His mutant was 6-fold larger than that of the wild type. The heme reduction rate with NADPH for the Trp421His mutant (0.09 min(-1)) was much lower than that (1.0 min(-1)) of the wild type. Taken together, it appears that Trp421 may be involved in inter-domain/inter-subunit electron transfer reactions.


Subject(s)
Electron Transport , Heme/chemistry , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase/metabolism , Tryptophan/metabolism , Amino Acid Sequence , Animals , Binding Sites , Heme/metabolism , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Nerve Tissue Proteins/chemistry , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase Type I , Rabbits , Sequence Alignment , Tryptophan/chemistry
13.
Biochemistry ; 38(50): 16413-8, 1999 Dec 14.
Article in English | MEDLINE | ID: mdl-10600101

ABSTRACT

Midpoint reduction potentials for the flavin cofactors in the reductase domain of rat neuronal nitric oxide synthase (nNOS) in calmodulin (CaM)-free and -bound forms have been determined by direct anaerobic titration. In the CaM-free form, the FMN potentials are -49 +/- 5 mV (oxidized/semiquinone) -274 +/- 5 mV (semiquinone/reduced). The corresponding FAD potentials are -232 +/- 7, and -280 +/- 6 mV. The data indicate that each flavin can exist as a blue (neutral) semiquinone. The accumulation of blue semiquinone on the FMN is considerably higher than seen on the FAD due to the much larger separation (225 mV) of its two potentials (cf. 48 mV for FAD). For the CaM-bound form of the protein, the midpoint potentials are essentially identical: there is a small alteration in the FMN oxidized/semiquinone potential (-30 +/- 4 mV); the other three potentials are unaffected. The heme midpoint potentials for nNOS [-239 mV, L-Arg-free; -220 mV, L-Arg-bound; Presta, A., Weber-Main, A. M., Stankovich, M. T., and Stuehr, D. J. (1998) J. Am. Chem. Soc. 120, 9460-9465] are poised such that electron transfer from flavin domain is thermodynamically feasible. Clearly, CaM binding is necessary in eliciting conformational changes that enhance flavin to flavin and flavin to heme electron transfers rather than causing a change in the driving force.


Subject(s)
Flavin Mononucleotide/chemistry , Flavin-Adenine Dinucleotide/chemistry , Flavins/chemistry , Nerve Tissue Proteins/chemistry , Nitric Oxide Synthase/chemistry , Animals , Binding Sites , Calmodulin/chemistry , Calmodulin/metabolism , Flavin Mononucleotide/metabolism , Flavin-Adenine Dinucleotide/metabolism , Flavins/metabolism , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Oxidation-Reduction , Potentiometry/methods , Rabbits , Rats
14.
J Biol Chem ; 274(43): 30589-95, 1999 Oct 22.
Article in English | MEDLINE | ID: mdl-10521442

ABSTRACT

The neuronal and endothelial nitric-oxide synthases (nNOS and eNOS) differ from inducible NOS in their dependence on the intracellular Ca(2+) concentration. Both nNOS and eNOS are activated by the reversible binding of calmodulin (CaM) in the presence of Ca(2+), whereas inducible NOS binds CaM irreversibly. One major divergence in the close sequence similarity between the NOS isoforms is a 40-50-amino acid insert in the middle of the FMN-binding domains of nNOS and eNOS. It has previously been proposed that this insert forms an autoinhibitory domain designed to destabilize CaM binding and increase its Ca(2+) dependence. To examine the importance of the insert we constructed two deletion mutants designed to remove the bulk of it from nNOS. Both mutants (Delta40 and Delta42) retained maximal NO synthesis activity at lower concentrations of free Ca(2+) than the wild type enzyme. They were also found to retain 30% of their activity in the absence of Ca(2+)/CaM, indicating that the insert plays an important role in disabling the enzyme when the physiological Ca(2+) concentration is low. Reduction of nNOS heme by NADPH under rigorous anaerobic conditions was found to occur in the wild type enzyme only in the presence of Ca(2+)/CaM. However, reduction of heme in the Delta40 mutant occurred spontaneously on addition of NADPH in the absence of Ca(2+)/CaM. This suggests that the insert regulates activity by inhibiting electron transfer from FMN to heme in the absence of Ca(2+)/CaM and by destabilizing CaM binding at low Ca(2+) concentrations, consistent with its role as an autoinhibitory domain.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , DNA Transposable Elements , Flavin Mononucleotide/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Egtazic Acid/pharmacology , Electron Transport , Flavin-Adenine Dinucleotide/metabolism , Humans , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , NADH Dehydrogenase/chemistry , NADH, NADPH Oxidoreductases/metabolism , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Rats , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Spectrophotometry
15.
J Biol Chem ; 274(38): 26956-61, 1999 Sep 17.
Article in English | MEDLINE | ID: mdl-10480907

ABSTRACT

Nitric-oxide synthase (NOS) is composed of an oxygenase domain having cytochrome P450-type heme active site and a reductase domain having FAD- and FMN-binding sites. To investigate the route of electron transfer from the reductase domain to the heme, we generated mutants at Lys(423) in the heme proximal site of neuronal NOS and examined the catalytic activities, electron transfer rates, and NADPH oxidation rates. A K423E mutant showed no NO formation activity (<0.1 nmol/min/nmol heme), in contrast with that (72 nmol/min/nmol heme) of the wild type enzyme. The electron transfer rate (0.01 min(-1)) of the K423E on addition of excess NADPH was much slower than that (>10 min(-1)) of the wild type enzyme. From the crystal structure of the oxygenase domain of endothelial NOS, Lys(423) of neuronal NOS is likely to interact with Trp(409) which lies in contact with the heme plane and with Cys(415), the axial ligand. It is also exposed to solvent and lies in the region where the heme is closest to the protein surface. Thus, it seems likely that ionic interactions between Lys(423) and the reductase domain may help to form a flavin to heme electron transfer pathway.


Subject(s)
Lysine/physiology , Nitric Oxide Synthase/physiology , Amino Acid Sequence , Animals , Binding Sites , Catalysis , Cattle , Crystallography, X-Ray , Electron Transport , Heme/metabolism , Kinetics , Models, Molecular , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase Type I , Potassium Chloride/pharmacology , Protein Conformation , Spectrophotometry, Atomic
17.
Eur J Biochem ; 257(3): 577-85, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9839946

ABSTRACT

The genes encoding the Escherichia coli flavodoxin NADP+ oxidoreductase (FLDR) and flavodoxin (FLD) have been overexpressed in E. coli as the major cell proteins (at least 13.5% and 11.4% of total soluble protein, respectively) and the gene products purified to homogeneity. The FLDR reduces potassium ferricyanide with a kcat of 1610.3 min(-1) and a Km of 23.6 microM, and cytochrome c with a kcat of 141.3 min(-1) and a Km of 17.6 microM. The cytochrome c reductase rate is increased sixfold by addition of FLD and an apparent Km of 6.84 microM was measured for the affinity of the two flavoproteins. The molecular masses of FLDR and FLD apoproteins were determined as 27648 Da and 19606 Da and the isoelectric points as 4.8 and 3.5, respectively. The mass of the FLDR is precisely that predicted from the atomic structure and indicates that residue 126 is arginine, not glutamine as predicted from the gene sequence. FLDR and FLD were covalently crosslinked using 1-ethyl-3(dimethylamino-propyl) carbodiimide to generate a catalytically active heterodimer. The midpoint reduction potentials of the oxidised/semiquinone and semiquinone/hydroquinone couples of both FLDR (-308 mV and -268 mV, respectively) and FLD (-254 mV and -433 mV, respectively) were measured using redox potentiometry. This confirms the electron-transfer route as NADPH-->FLDR-->FLD. Binding of 2' adenosine monophosphate increases the midpoint reduction potentials for both FLDR couples. These data highlight the strong stabilisation of the flavodoxin semiquinone (absorption coefficient calculated as 4933 M(-1) cm(-1) at 583 nm) with respect to the hydroquinone state and indicate that FLD must act as a single electron shuttle from the semiquinone form in its support of cellular functions, and to facilitate catalytic activity of microsomal cytochromes P-450 heterologously expressed in E. coli. Kinetic studies of electron transfer from FLDR/FLD to the fatty acid oxidase P-450 BM3 support this conclusion, indicating a ping-pong mechanism. This is the first report of the potentiometric analysis of the full E. coli NAD(P)H/FLDR/FLD electron-transfer chain; a complex critical to the function of a large number of E. coli redox systems.


Subject(s)
Escherichia coli/metabolism , Flavodoxin/metabolism , NADH, NADPH Oxidoreductases/metabolism , Base Sequence , Cross-Linking Reagents/chemistry , Cytochrome P-450 Enzyme System/metabolism , DNA Primers , Electron Transport , Escherichia coli/enzymology , Flavodoxin/chemistry , NADH, NADPH Oxidoreductases/chemistry , Oxidation-Reduction , Potentiometry
18.
Biochem Biophys Res Commun ; 249(3): 678-82, 1998 Aug 28.
Article in English | MEDLINE | ID: mdl-9731196

ABSTRACT

Mutation of the conserved Thr319 residue to Ala of cytochrome P4501A2 (CYP1A2) increased the value of Vmax 9-fold for reductive dehalogenation of hexachloroethane in the reconstituted system under anaerobic conditions. The Thr319Ala mutation also increased the elimination over substitution product ratio by 5-fold. The addition of aliphatic alcohols increased by 22-fold the activity obtained with the wild type and varied the elimination over substitution product ratio. Increasing pH increased the ratio of elimination over substitution by primarily affecting the rate of elimination.


Subject(s)
Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Ethane/analogs & derivatives , Hydrocarbons, Chlorinated/metabolism , Point Mutation , Alcohols/pharmacology , Anaerobiosis , Binding Sites/genetics , Cytochrome P-450 CYP1A2/chemistry , Ethane/metabolism , Hydrogen-Ion Concentration , Kinetics , Mutagenesis, Site-Directed , NADP/metabolism , Oxidation-Reduction , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
19.
FEBS Lett ; 430(3): 377-80, 1998 Jul 03.
Article in English | MEDLINE | ID: mdl-9688574

ABSTRACT

The dissociation constant (Kd) for CO from neuronal nitric oxide synthase heme in the absence of the substrate and cofactor was less than 10(-3) microM. In the presence of L-Arg, it dramatically increased up to 1 microM. In the presence of inhibitors such as N(G)-nitro-L-arginine methyl ester and 7-nitroindazole (NI), the Kd value further increased up to more than 100 microM. Addition of the cofactor, 5,6,7,8-tetrahydrobiopterin (H4B), increased the Kd value by 10-fold in the presence of L-Arg, whereas it decreased the value to less than one 250th in the presence of NI. Addition of H4B increased the recombination rate constant (k(on)) for CO by more than two-fold in the presence of L-Arg or N6-(1-iminoethyl)-L-lysine, whereas it decreased the k(on) value by three-fold in the presence of L-thiocitrulline. Thus, the binding fashion of some of inhibitors, such as NI, may be different from that of L-Arg with respect to the H4B effect.


Subject(s)
Biopterins/analogs & derivatives , Carbon Monoxide/metabolism , Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase/metabolism , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Biopterins/pharmacology , Kinetics , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase Type I , Photolysis , Rats , Spectrophotometry
20.
Biochem Biophys Res Commun ; 248(3): 767-72, 1998 Jul 30.
Article in English | MEDLINE | ID: mdl-9704002

ABSTRACT

The effects of substrate, L-Arg and cofactors, (6R)-L-erythro-5,6,7,8-tetrahydrobiopterin (H4B) and calmodulin (CaM), on chiral discrimination by rat neuronal nitric oxide synthase (nNOS) for binding the enantiomers of 1-(1-naphthyl)ethylamine (ligand I), 1-cyclohexylethylamine (ligand II), and 1-(4-pyridyl)ethanol (ligand III) were studied under anaerobic conditions by optical absorption spectroscopy. The ratio of the dissociation constant (Kd) values for the S- and R-enantiomers of ligand I (S/R) was 30, while the S/R ratio for ligand II and the R/S ratio for ligand III were 1.8 and < 0.14, respectively, in the presence of 0.15 microM H4B. However, in the presence of 1 mM L-Arg, the S/R ratio of the Kd values for ligand I was decreased down to 5.9. In the presence of both 1 mM L-Arg and 0.1 mM H4B, the S/R ratios for ligands I and II and the R/S ratio for ligand III were enormously increased up to 29, > 80, and 60, respectively. These and other spectral observations strongly suggest that strict chiral recognition at the active site of nNOS during catalysis is exhibited only in the presence of the active effector.


Subject(s)
Antioxidants/pharmacology , Arginine/pharmacology , Biopterins/analogs & derivatives , Heme/metabolism , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/metabolism , Anaerobiosis , Animals , Binding Sites , Biopterins/pharmacology , Calmodulin/pharmacology , Cloning, Molecular , Kinetics , Nitric Oxide Synthase Type I , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Saccharomyces cerevisiae , Spectrophotometry , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...