Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 10): 2667-74, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25286850

ABSTRACT

Mammals produce three isoforms of nitric oxide synthase (NOS): neuronal NOS (nNOS), inducible NOS (iNOS) and endothelial NOS (eNOS). The overproduction of NO by nNOS is associated with a number of neurodegenerative disorders; therefore, a desirable therapeutic goal is the design of drugs that target nNOS but not the other isoforms. Crystallography, coupled with computational approaches and medicinal chemistry, has played a critical role in developing highly selective nNOS inhibitors that exhibit exceptional neuroprotective properties. For historic reasons, crystallography has focused on rat nNOS and bovine eNOS because these were available in high quality; thus, their structures have been used in structure-activity-relationship studies. Although these constitutive NOSs share more than 90% sequence identity across mammalian species for each NOS isoform, inhibitor-binding studies revealed that subtle differences near the heme active site in the same NOS isoform across species still impact enzyme-inhibitor interactions. Therefore, structures of the human constitutive NOSs are indispensible. Here, the first structure of human neuronal NOS at 2.03 Šresolution is reported and a different crystal form of human endothelial NOS is reported at 1.73 Šresolution.


Subject(s)
Nitric Oxide Synthase Type III/chemistry , Nitric Oxide Synthase Type I/chemistry , Aminopyridines/chemistry , Animals , Catalytic Domain , Cattle , Cloning, Molecular , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Heme/chemistry , Humans , Models, Molecular , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Protein Structure, Tertiary , Pyrrolidines/chemistry , Rats
2.
Biochemistry ; 53(32): 5272-9, 2014 Aug 19.
Article in English | MEDLINE | ID: mdl-25089924

ABSTRACT

Many pyrrolidine-based inhibitors highly selective for neuronal nitric oxide synthase (nNOS) over endothelial NOS (eNOS) exhibit dramatically different binding modes. In some cases, the inhibitor binds in a 180° flipped orientation in nNOS relative to eNOS. From the several crystal structures we have determined, we know that isoform selectivity correlates with the rotamer position of a conserved tyrosine residue that H-bonds with a heme propionate. In nNOS, this Tyr more readily adopts the out-rotamer conformation, while in eNOS, the Tyr tends to remain fixed in the original in-rotamer conformation. In the out-rotamer conformation, inhibitors are able to form better H-bonds with the protein and heme, thus increasing inhibitor potency. A segment of polypeptide that runs along the surface near the conserved Tyr has long been thought to be the reason for the difference in Tyr mobility. Although this segment is usually disordered in both eNOS and nNOS, sequence comparisons and modeling from a few structures show that this segment is structured quite differently in eNOS and nNOS. In this study, we have probed the importance of this surface segment near the Tyr by making a few mutants in the region followed by crystal structure determinations. In addition, because the segment near the conserved Tyr is highly ordered in iNOS, we also determined the structure of an iNOS-inhibitor complex. This new structure provides further insight into the critical role that mobility plays in isoform selectivity.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Tyrosine/chemistry , Animals , Binding Sites , Cattle , Conserved Sequence , Crystallization , Isoenzymes , Models, Molecular , Molecular Structure , Nitric Oxide Synthase/chemistry , Protein Conformation , Rats , Structure-Activity Relationship , X-Ray Diffraction
3.
J Inorg Biochem ; 124: 1-10, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23567464

ABSTRACT

Electron flow within the neuronal nitric oxide synthase reductase domain (nNOSrd) includes hydride transfer from NADPH to FAD followed by two one-electron transfer reactions from FAD to FMN. We have used stopped flow spectrometry to closely monitor these electron transfer steps for both the wild type and the ΔG810 mutant of nNOSrd using a protocol involving both global analyses of the photodiode array spectral scans and curve fittings of single wavelength kinetic traces. The charge transfer complex and interflavin electron transfer events recorded at 750nm and 600nm, respectively, show the kinetics in different time frames. All electron transfer events are slow enough at 4°C to enable measurements of rate constants even for the fast charge transfer event. To our knowledge this is the first time the rate constants for the charge transfer between NADP(+) and FADH2 have been determined for NOS. These procedures allow us to conclude that (1) binding of the second NADPH is necessary to drive the full reduction of FMN and; (2) charge transfer and the subsequent interflavin electron transfer have distinct spectral features that can be monitored separately with stopped flow spectroscopy. These studies also enable us to conclude that interflavin electron transfer reported at 600nm is not limiting in NOS catalysis.


Subject(s)
Flavin-Adenine Dinucleotide/analogs & derivatives , NADP/chemistry , Nitric Oxide Synthase Type I/chemistry , Flavin-Adenine Dinucleotide/chemistry , Flavin-Adenine Dinucleotide/metabolism , Humans , Kinetics , Mutation , NADP/metabolism , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism
4.
Biochemistry ; 49(51): 10803-10, 2010 Dec 28.
Article in English | MEDLINE | ID: mdl-21138269

ABSTRACT

In previous studies [Delker, S. L., et al. (2010), J. Am. Chem. Soc. 132, 5437-5442], we determined the crystal structures of neuronal nitric oxide synthase (nNOS) in complex with nNOS-selective chiral pyrrolidine inhibitors, designed to have an aminopyridine group bound over the heme where it can electrostatically interact with the conserved active site Glu residue. However, in addition to the expected binding mode with the (S,S)-cis inhibitors, an unexpected "flipped" orientation was observed for the (R,R)-cis enantiomers. In the flipped mode, the aminopyridine extends out of the active site where it interacts with one heme propionate. This prompted us to design and synthesize symmetric "double-headed" inhibitors with an aminopyridine at each end of a bridging ring structure [Xue, F., Delker, S. L., Li, H., Fang, J., Jamal, J., Martásek, P., Roman, L. J., Poulos, T. L., and Silverman, R. B. Symmetric double-headed aminopyridines, a novel strategy for potent and membrane-permeable inhibitors of neuronal nitric oxide synthase. J. Med. Chem. (submitted for publication)]. One aminopyridine should interact with the active site Glu and the other with the heme propionate. Crystal structures of these double-headed aminopyridine inhibitors in complexes with nNOS show unexpected and significant protein and heme conformational changes induced by inhibitor binding that result in removal of the tetrahydrobiopterin (H(4)B) cofactor and creation of a new Zn(2+) site. These changes are due to binding of a second inhibitor molecule that results in the displacement of H(4)B and the placement of the inhibitor pyridine group in position to serve as a Zn(2+) ligand together with Asp, His, and a chloride ion. Binding of the second inhibitor molecule and generation of the Zn(2+) site do not occur in eNOS. Structural requirements for creation of the new Zn(2+) site in nNOS were analyzed in detail. These observations open the way for the potential design of novel inhibitors selective for nNOS.


Subject(s)
Aminopyridines/chemistry , Aminopyridines/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/metabolism , Zinc/metabolism , Animals , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Nitric Oxide Synthase Type I/chemistry , Protein Binding , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Rats , Zinc/chemistry
5.
J Am Chem Soc ; 132(15): 5437-42, 2010 Apr 21.
Article in English | MEDLINE | ID: mdl-20337441

ABSTRACT

Selective inhibition of the neuronal isoform of nitric oxide synthase NOS (nNOS) has been shown to prevent brain injury and is important for the treatment of various neurodegenerative disorders. However, given the high active site conservation among all three NOS isoforms, the design of selective inhibitors is an extremely challenging problem. Here we present the structural basis for why novel and potent nNOS inhibitors exhibit the highest level of selectivity over eNOS reported so far (approximately 3,800-fold). By using a combination of crystallography, computational methods, and site-directed mutagenesis, we found that inhibitor chirality and an unanticipated structural change of the target enzyme control both the orientation and selectivity of these novel nNOS inhibitors. A new hot spot generated as a result of enzyme elasticity provides important information for the future fragment-based design of selective NOS inhibitors.


Subject(s)
Enzyme Inhibitors/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type I/antagonists & inhibitors , Animals , Cerebral Palsy , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Mice , Models, Animal , Models, Molecular
6.
J Med Chem ; 52(7): 2060-6, 2009 Apr 09.
Article in English | MEDLINE | ID: mdl-19296678

ABSTRACT

New nitric oxide synthase (NOS) inhibitors were designed de novo with knowledge gathered from the studies on the nNOS-selective dipeptide inhibitors. Each of the new inhibitors consists of three fragments: an aminopyridine ring, a pyrrolidine, and a tail of various length and polarity. The in vitro inhibitory assays indicate good potency and isoform selectivity for some of the compounds. Crystal structures of these inhibitors bound to either wild type or mutant nNOS and eNOS have confirmed design expectations. The aminopyridine ring mimics the guanidinium group of L-arginine and functions as an anchor to place the compound in the NOS active site where it hydrogen bonds to a conserved Glu. The rigidity of the pyrrolidine ring places the pyrrolidine ring nitrogen between the same conserved Glu and the selective residue nNOS Asp597/eNOS Asn368, which results in similar interactions observed with the alpha-amino group of dipeptide inhibitors bound to nNOS. These structures provide additional information to help in the design of inhibitors with greater potency, physicochemical properties, and isoform selectivity.


Subject(s)
Aminopyridines/chemistry , Enzyme Inhibitors/chemistry , Nitric Oxide Synthase Type III/chemistry , Nitric Oxide Synthase Type I/chemistry , Pyrrolidines/chemistry , Crystallography, X-Ray , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Mutation , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/genetics , Protein Binding , Protein Conformation , Thermodynamics
7.
J Biol Chem ; 283(50): 34762-72, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-18852262

ABSTRACT

In nitric-oxide synthase (NOS) the FMN can exist as the fully oxidized (ox), the one-electron reduced semiquinone (sq), or the two-electron fully reduced hydroquinone (hq). In NOS and microsomal cytochrome P450 reductase the sq/hq redox potential is lower than that of the ox/sq couple, and hence it is the hq form of FMN that delivers electrons to the heme. Like NOS, cytochrome P450BM3 has the FAD/FMN reductase fused to the C-terminal end of the heme domain, but in P450BM3 the ox/sq and sq/hq redox couples are reversed, so it is the sq that transfers electrons to the heme. This difference is due to an extra Gly residue found in the FMN binding loop in NOS compared with P450BM3. We have deleted residue Gly-810 from the FMN binding loop in neuronal NOS (nNOS) to give Delta G810 so that the shorter binding loop mimics that in cytochrome P450BM3. As expected, the ox/sq redox potential now is lower than the sq/hq couple. Delta G810 exhibits lower NO synthase activity but normal levels of cytochrome c reductase activity. However, unlike the wild-type enzyme, the cytochrome c reductase activity of Delta G810 is insensitive to calmodulin binding. In addition, calmodulin binding to Delta G810 does not result in a large increase in FMN fluorescence as in wild-type nNOS. These results indicate that the FMN domain in Delta G810 is locked in a unique conformation that is no longer sensitive to calmodulin binding and resembles the "on" output state of the calmodulin-bound wild-type nNOS with respect to the cytochrome c reduction activity.


Subject(s)
Flavin Mononucleotide/chemistry , Neurons/metabolism , Nitric Oxide Synthase Type I/metabolism , Oxidation-Reduction , Animals , Biochemistry/methods , Calmodulin/metabolism , Models, Chemical , Models, Molecular , Molecular Conformation , NADPH-Ferrihemoprotein Reductase/chemistry , Protein Binding , Rats , Spectrometry, Fluorescence/methods , Time Factors
8.
J Biol Inorg Chem ; 11(6): 753-68, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16804678

ABSTRACT

Crystal structures are reported for the endothelial nitric oxide synthase (eNOS)-arginine-CO ternary complex as well as the neuronal nitric oxide synthase (nNOS) heme domain complexed with L: -arginine and diatomic ligands, CO or NO, in the presence of the native cofactor, tetrahydrobiopterin, or its oxidized analogs, dihydrobiopterin and 4-aminobiopterin. The nature of the biopterin has no influence on the diatomic ligand binding. The binding geometries of diatomic ligands to nitric oxide synthase (NOS) follow the {MXY}(n) formalism developed from the inorganic diatomic-metal complexes. The structures reveal some subtle structural differences between eNOS and nNOS when CO is bound to the heme which correlate well with the differences in CO stretching frequencies observed by resonance Raman techniques. The detailed hydrogen-bonding geometries depicted in the active site of nNOS structures indicate that it is the ordered active-site water molecule rather than the substrate itself that would most likely serve as a direct proton donor to the diatomic ligands (CO, NO, as well as O(2)) bound to the heme. This has important implications for the oxygen activation mechanism critical to NOS catalysis.


Subject(s)
Nitric Oxide Synthase/chemistry , Biopterins/metabolism , Crystallization , Crystallography, X-Ray , Ligands , Models, Molecular , Nitric Oxide/chemistry , Nitric Oxide Synthase/metabolism , Protein Conformation
9.
Biochemistry ; 44(46): 15222-9, 2005 Nov 22.
Article in English | MEDLINE | ID: mdl-16285725

ABSTRACT

A series of L-nitroarginine-based dipeptide inhibitors are highly selective for neuronal nitric oxide synthase (nNOS) over the endothelial isoform (eNOS). Crystal structures of these dipeptides bound to both isoforms revealed two different conformations, curled in nNOS and extended in eNOS, corresponding to higher and lower binding affinity to the two isoforms, respectively. In previous studies we found that the primary reason for selectivity is that Asp597 in nNOS, which is Asn368 in eNOS, provides greater electrostatic stabilization in the inhibitor complex. While this is the case for smaller dipeptide inhibitors, electrostatic stabilization may no longer be the sole determinant for isoform selectivity with bulkier dipeptide inhibitors. Another residue farther away from the active site, Met336 in nNOS (Val106 in eNOS), is in contact with bulkier dipeptide inhibitors. Double mutants were made to exchange the D597/M336 pair in nNOS with N368/V106 in eNOS. Here we report crystal structures and inhibition constants for bulkier dipeptide inhibitors bound to nNOS and eNOS that illustrate the important role played by residues near the entry to the active site in isoform selective inhibition.


Subject(s)
Guanidines/chemistry , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitro Compounds/chemistry , Protein Conformation , Animals , Cattle , Crystallization , Mannitol/chemistry , Molecular Structure , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type III/genetics , Nitroarginine/chemistry , Point Mutation , Protein Binding , Rats , X-Ray Diffraction
10.
Biochemistry ; 43(18): 5181-7, 2004 May 11.
Article in English | MEDLINE | ID: mdl-15122883

ABSTRACT

In a continuing effort to unravel the structural basis for isoform-selective inhibition of nitric oxide synthase (NOS) by various inhibitors, we have determined the crystal structures of the nNOS and eNOS heme domain bound with two D-nitroarginine-containing dipeptide inhibitors, D-Lys-D-Arg(NO)2-NH(2) and D-Phe-D-Arg(NO)2-NH(2). These two dipeptide inhibitors exhibit similar binding modes in the two constitutive NOS isozymes, which is consistent with the similar binding affinities for the two isoforms as determined by K(i) measurements. The D-nitroarginine-containing dipeptide inhibitors are not distinguished by the amino acid difference between nNOS and eNOS (Asp 597 and Asn 368, respectively) which is key in controlling isoform selection for nNOS over eNOS observed for the L-nitroarginine-containing dipeptide inhibitors reported previously [Flinspach, M., et al. (2004) Nat. Struct. Mol. Biol. 11, 54-59]. The lack of a free alpha-amino group on the D-nitroarginine moiety makes the dipeptide inhibitor steer away from the amino acid binding pocket near the active site. This allows the inhibitor to extend into the solvent-accessible channel farther away from the active site, which enables the inhibitors to explore new isoform-specific enzyme-inhibitor interactions. This might be the structural basis for why these D-nitroarginine-containing inhibitors are selective for nNOS (or eNOS) over iNOS.


Subject(s)
Dipeptides/chemistry , Enzyme Inhibitors/chemistry , Heme/chemistry , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/chemistry , Nitroarginine/chemistry , Amino Acid Substitution , Animals , Cattle , Dipeptides/metabolism , Enzyme Inhibitors/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type III , Nitroarginine/metabolism , Protein Binding , Protein Structure, Tertiary , Rats
11.
Nat Struct Mol Biol ; 11(1): 54-9, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14718923

ABSTRACT

Three nitric oxide synthase (NOS) isoforms, eNOS, nNOS and iNOS, generate nitric oxide (NO) crucial to the cardiovascular, nervous and host defense systems, respectively. Development of isoform-selective NOS inhibitors is of considerable therapeutic importance. Crystal structures of nNOS-selective dipeptide inhibitors in complex with both nNOS and eNOS were solved and the inhibitors were found to adopt a curled conformation in nNOS but an extended conformation in eNOS. We hypothesized that a single-residue difference in the active site, Asp597 (nNOS) versus Asn368 (eNOS), is responsible for the favored binding in nNOS. In the D597N nNOS mutant crystal structure, a bound inhibitor switches to the extended conformation and its inhibition of nNOS decreases >200-fold. Therefore, a single-residue difference is responsible for more than two orders of magnitude selectivity in inhibition of nNOS over eNOS by L-N(omega)-nitroarginine-containing dipeptide inhibitors.


Subject(s)
Dipeptides/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Amides/chemistry , Amides/pharmacology , Animals , Catalytic Domain , Cattle , Crystallography, X-Ray , Dipeptides/chemistry , In Vitro Techniques , Models, Molecular , Mutagenesis, Site-Directed , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type III , Protein Isoforms/chemistry , Protein Isoforms/pharmacology , Protein Structure, Tertiary , Rats , Static Electricity
12.
Biochemistry ; 41(47): 13868-75, 2002 Nov 26.
Article in English | MEDLINE | ID: mdl-12437343

ABSTRACT

A series of N-alkyl-N'-hydroxyguanidine compounds have recently been characterized as non-amino acid substrates for all three nitric oxide synthase (NOS) isoforms which mimic NO formation from N(omega)-hydroxy-L-arginine. Crystal structures of the nNOS heme domain complexed with either N-isopropyl-N'-hydroxyguanidine or N-butyl-N'-hydroxyguanidine reveal two different binding modes in the substrate binding pocket. The binding mode of the latter is consistent with that observed for the substrate N(omega)-hydroxy-L-arginine bound in the nNOS active site. However, the former binds to nNOS in an unexpected fashion, thus providing new insights into the mechanism on how the hydroxyguanidine moiety leads to NO formation. Structural features of substrate binding support the view that the OH-substituted guanidine nitrogen, instead of the hydroxyl oxygen, is the source of hydrogen supplied to the active ferric-superoxy species for the second step of the NOS catalytic reaction.


Subject(s)
Guanidines/chemistry , Nitric Oxide Synthase/chemistry , Nitric Oxide/biosynthesis , Alkylation , Animals , Binding Sites , Cloning, Molecular , Guanidines/pharmacokinetics , Hydroxylamines , Isoenzymes/chemistry , Isoenzymes/metabolism , Models, Molecular , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Protein Conformation , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...