Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters










Publication year range
1.
Antioxidants (Basel) ; 9(2)2020 Jan 21.
Article in English | MEDLINE | ID: mdl-31972975

ABSTRACT

: A specific light trigger for activating endothelial Nitric Oxide-Synthase (eNOS) in real time would be of unique value to decipher cellular events associated with eNOS activation or to generate on demand cytotoxic levels of NO at specific sites for cancer research. We previously developed novel tools called nanotriggers (NT), which recognized constitutive NO-synthase, eNOS or neuronal NOS (nNOS), mainly via their 2' phosphate group which is also present in NADPH in its binding site. Laser excitation of NT1 bound to eNOS triggered recombinant NOS activity and released NO. We recently generated new NTs carrying a 2' or 3' carboxylate group or two 2' and 3' carboxylate moieties replacing the 2' phosphate group of NADPH. Among these new NT, only the 3' carboxylate derivative released NO from endothelial cells upon laser activation. Here, Molecular Dynamics (MD) simulations showed that the 3' carboxylate NT formed a folded structure with a hydrophobic hub, inducing a good stacking on FAD that likely drove efficient activation of nNOS. This NT also carried an additional small charged group which increased binding to e/nNOS; fluorescence measurements determined a 20-fold improved affinity upon binding to nNOS as compared to NT1 affinity. To gain in specificity for eNOS, we augmented a previous NT with a "hook" targeting variable residues in the NADPH site of eNOS. We discuss the potential of exploiting the chemical diversity within the NADPH site of eNOS for reversal of endothelial dysfunction in cells and for controlled generation of cytotoxic NO-derived species in cancer tissues.

2.
J Biol Inorg Chem ; 24(1): 1-9, 2019 02.
Article in English | MEDLINE | ID: mdl-30315355

ABSTRACT

Phosphorylation is an important pathway for the regulation of nitric oxide synthase (NOS) at the posttranslational level. However, the molecular underpinnings of NOS regulation by phosphorylations remain unclear to date, mainly because of the problems in making a good amount of active phospho-NOS proteins. Herein, we have established a system in which recombinant rat nNOS holoprotein can be produced with site-specific incorporation of phosphoserine (pSer) at residue 1412, using a specialized bacterial host strain for pSer incorporation. The pSer1412 nNOS protein demonstrates UV-Vis, far-UV CD and fluorescence spectral properties that are identical to those of nNOS overexpressed in other bacterial strains. The protein is also functional, possessing normal NO production and NADPH oxidation activities in the presence of abundant substrate L-Arg. Conversely, the rate of FMN-heme interdomain electron transfer (IET) in pSer1412 nNOS is considerably lower than that of wild-type (wt) nNOS, while the phosphomimetic S1142E mutant possesses similar electron transfer kinetics to that of wt. The successful incorporation and high yield of pSer1412 into rat nNOS and the significant change in the IET kinetics upon the phosphorylation demonstrate a highly useful method for incorporating native phosphorylation sites as a substantial improvement to commonly used phosphomimetics.


Subject(s)
Nitric Oxide Synthase Type I/genetics , Phosphoserine/metabolism , Protein Engineering , Serine/genetics , Animals , Holoenzymes/genetics , Holoenzymes/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/metabolism , Phosphorylation , Point Mutation , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/analogs & derivatives
3.
J Med Chem ; 60(16): 7146-7165, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28776992

ABSTRACT

Neuronal nitric oxide synthase (nNOS) is a target for development of antineurodegenerative agents. Most nNOS inhibitors mimic l-arginine and have poor bioavailability. 2-Aminoquinolines showed promise as bioavailable nNOS inhibitors but suffered from low human nNOS inhibition, low selectivity versus human eNOS, and significant binding to other CNS targets. We aimed to improve human nNOS potency and selectivity and reduce off-target binding by (a) truncating the original scaffold or (b) introducing a hydrophilic group to interrupt the lipophilic, promiscuous pharmacophore and promote interaction with human nNOS-specific His342. We synthesized both truncated and polar 2-aminoquinoline derivatives and assayed them against recombinant NOS enzymes. Although aniline and pyridine derivatives interact with His342, benzonitriles conferred the best rat and human nNOS inhibition. Both introduction of a hydrophobic substituent next to the cyano group and aminoquinoline methylation considerably improved isoform selectivity. Most importantly, these modifications preserved Caco-2 permeability and reduced off-target CNS binding.


Subject(s)
Aminoquinolines/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Aminoquinolines/chemical synthesis , Animals , Caco-2 Cells , Cattle , Cell Membrane Permeability/drug effects , Enzyme Assays , Histidine/chemistry , Humans , Mice , Nitric Oxide Synthase Type I/chemistry , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type III/antagonists & inhibitors , Rats
4.
Free Radic Biol Med ; 110: 261-269, 2017 09.
Article in English | MEDLINE | ID: mdl-28666850

ABSTRACT

Previously published studies strongly suggested that insulin- and exercise-induced skeletal muscle glucose uptake require nitric oxide (NO) production. However, the signal transduction mechanisms by which insulin and contraction regulated NO production and subsequent glucose transport are not known. In the present study, we utilized the myotube cell lines treated with insulin or hydrogen peroxide, the latter to mimic contraction-induced oxidative stress, to characterize these mechanisms. We found that insulin stimulation of neuronal nitric oxide synthase (nNOS) phosphorylation, NO production, and GLUT4 translocation were all significantly reduced by inhibition of either nNOS or Akt2. Hydrogen peroxide (H2O2) induced phosphorylation of nNOS at the same residue as did insulin, and also stimulated NO production and GLUT4 translocation. nNOS inhibition prevented H2O2-induced GLUT4 translocation. AMP activated protein kinase (AMPK) inhibition prevented H2O2 activation and phosphorylation of nNOS, leading to reduced NO production and significantly attenuated GLUT4 translocation. We conclude that nNOS phosphorylation and subsequently increased NO production are required for both insulin- and H2O2-stimulated glucose transport. Although the two stimuli result in phosphorylation of the same residue on nNOS, they do so through distinct protein kinases. Thus, insulin and H2O2-activated signaling pathways converge on nNOS, which is a common mediator of glucose uptake in both pathways. However, the fact that different kinases are utilized provides a basis for the use of exercise to activate glucose transport in the face of insulin resistance.


Subject(s)
Glucose/metabolism , Hydrogen Peroxide/pharmacology , Insulin/pharmacology , Muscle Fibers, Skeletal/drug effects , Nitric Oxide Synthase Type I/genetics , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Gene Expression Regulation , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Insulin Resistance , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type I/metabolism , Oxidative Stress , Phosphorylation , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
5.
J Biol Chem ; 291(39): 20487-502, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27496950

ABSTRACT

Human NADPH-cytochrome P450 oxidoreductase (POR) gene mutations are associated with severe skeletal deformities and disordered steroidogenesis. The human POR mutation A287P presents with disordered sexual development and skeletal malformations. Difficult recombinant expression and purification of this POR mutant suggested that the protein was less stable than WT. The activities of cytochrome P450 17A1, 19A1, and 21A2, critical in steroidogenesis, were similar using our purified, full-length, unmodified A287P or WT POR, as were those of several xenobiotic-metabolizing cytochromes P450, indicating that the A287P protein is functionally competent in vitro, despite its functionally deficient phenotypic behavior in vivo Differential scanning calorimetry and limited trypsinolysis studies revealed a relatively unstable A287P compared with WT protein, leading to the hypothesis that the syndrome observed in vivo results from altered POR protein stability. The crystal structures of the soluble domains of WT and A287P reveal only subtle differences between them, but these differences are consistent with the differential scanning calorimetry results as well as the differential susceptibility of A287P and WT observed with trypsinolysis. The relative in vivo stabilities of WT and A287P proteins were also examined in an osteoblast cell line by treatment with cycloheximide, a protein synthesis inhibitor, showing that the level of A287P protein post-inhibition is lower than WT and suggesting that A287P may be degraded at a higher rate. Current studies demonstrate that, unlike previously described mutations, A287P causes POR deficiency disorder due to conformational instability leading to proteolytic susceptibility in vivo, rather than through an inherent flavin-binding defect.


Subject(s)
Antley-Bixler Syndrome Phenotype , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/genetics , Mutation, Missense , Amino Acid Substitution , Antley-Bixler Syndrome Phenotype/enzymology , Antley-Bixler Syndrome Phenotype/genetics , Crystallography, X-Ray , Cytochrome P-450 Enzyme System/metabolism , Enzyme Stability/genetics , Humans
6.
J Med Chem ; 59(10): 4913-25, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27050842

ABSTRACT

Neuronal nitric oxide synthase (nNOS) is an important therapeutic target for the treatment of various neurodegenerative disorders. A major challenge in the design of nNOS inhibitors focuses on potency in humans and selectivity over other NOS isoforms. Here we report potent and selective human nNOS inhibitors based on the 2-aminopyridine scaffold with a central pyridine linker. Compound 14j, the most promising inhibitor in this study, exhibits excellent potency for rat nNOS (Ki = 16 nM) with 828-fold n/e and 118-fold n/i selectivity with a Ki value of 13 nM against human nNOS with 1761-fold human n/e selectivity. Compound 14j also displayed good metabolic stability in human liver microsomes, low plasma protein binding, and minimal binding to cytochromes P450 (CYPs), although it had little to no Caco-2 permeability.


Subject(s)
Aminopyridines/chemistry , Aminopyridines/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Aminopyridines/chemical synthesis , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Humans , Molecular Structure , Nitric Oxide Synthase Type I/metabolism , Structure-Activity Relationship
8.
J Med Chem ; 58(21): 8694-712, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26469213

ABSTRACT

Excess nitric oxide (NO) produced by neuronal nitric oxide synthase (nNOS) is implicated in neurodegenerative disorders. As a result, inhibition of nNOS and reduction of NO levels is desirable therapeutically, but many nNOS inhibitors are poorly bioavailable. Promising members of our previously reported 2-aminoquinoline class of nNOS inhibitors, although orally bioavailable and brain-penetrant, suffer from unfavorable off-target binding to other CNS receptors, and they resemble known promiscuous binders. Rearranged phenyl ether- and aniline-linked 2-aminoquinoline derivatives were therefore designed to (a) disrupt the promiscuous binding pharmacophore and diminish off-target interactions and (b) preserve potency, isoform selectivity, and cell permeability. A series of these compounds was synthesized and tested against purified nNOS, endothelial NOS (eNOS), and inducible NOS (iNOS) enzymes. One compound, 20, displayed high potency, selectivity, and good human nNOS inhibition, and retained some permeability in a Caco-2 assay. Most promisingly, CNS receptor counterscreening revealed that this rearranged scaffold significantly reduces off-target binding.


Subject(s)
Aminoquinolines/chemistry , Aminoquinolines/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Phenyl Ethers/chemistry , Phenyl Ethers/pharmacology , Aminoquinolines/pharmacokinetics , Caco-2 Cells , Crystallography, X-Ray , Enzyme Inhibitors/pharmacokinetics , Humans , Models, Molecular , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Phenyl Ethers/pharmacokinetics , Structure-Activity Relationship
9.
Lab Invest ; 95(10): 1092-104, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26214584

ABSTRACT

The balance of nitric oxide (NO) versus superoxide generation has a major role in the initiation and progression of endothelial dysfunction. Under conditions of high glucose, endothelial nitric oxide synthase (eNOS) functions as a chief source of superoxide rather than NO. In order to improve NO bioavailability within the vessel wall in type-1 diabetes, we investigated treatment strategies that improve eNOS phosphorylation and NO-dependent vasorelaxation. We evaluated methods to increase the eNOS activity by (1) feeding Ins2(Akita) spontaneously diabetic (type-1) mice with l-arginine in the presence of sepiapterin, a precursor of tetrahydrobiopterin; (2) preventing eNOS/NO deregulation by the inclusion of inhibitor kappa B kinase beta (IKKß) inhibitor, salsalate, in the diet regimen in combination with l-arginine and sepiapterin; and (3) independently increasing eNOS expression to improve eNOS activity and associated NO production through generating Ins2(Akita) diabetic mice that overexpress human eNOS predominantly in vascular endothelial cells. Our results clearly demonstrated that diet supplementation with l-arginine, sepiapterin along with salsalate improved phosphorylation of eNOS and enhanced vasorelaxation of thoracic/abdominal aorta in type-1 diabetic mice. More interestingly, despite the overexpression of eNOS, the in-house generated transgenic eNOS-GFP (TgeNOS-GFP)-Ins2(Akita) cross mice showed an unanticipated effect of reduced eNOS phosphorylation and enhanced superoxide production. Our results demonstrate that enhancement of endogenous eNOS activity by nutritional modulation is more beneficial than increasing the endogenous expression of eNOS by gene therapy modalities.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Dietary Supplements , Endothelium, Vascular/metabolism , Hypoglycemic Agents/therapeutic use , Nitric Oxide Synthase Type III/metabolism , Protein Kinase Inhibitors/therapeutic use , Protein Processing, Post-Translational , Animals , Aorta/cytology , Aorta/metabolism , Aorta/physiopathology , Arginine/metabolism , Arginine/therapeutic use , Cattle , Cells, Cultured , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiopathology , Female , Heterozygote , Humans , Hypoglycemic Agents/metabolism , Insulin/genetics , Insulin/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide Synthase Type III/chemistry , Nitric Oxide Synthase Type III/genetics , Phosphorylation , Protein Kinase Inhibitors/metabolism , Pterins/metabolism , Pterins/therapeutic use , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Salicylates/metabolism , Salicylates/therapeutic use , Weaning
10.
J Med Chem ; 58(14): 5548-60, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26120733

ABSTRACT

We have analyzed a recently obtained crystal structure of human neuronal nitric oxide synthase (nNOS) and then designed and synthesized several 2-aminopyridine derivatives containing a truncated side chain to avoid the hydrophobic pocket that differentiates human and rat nNOS in an attempt to explore alternative binding poses along the substrate access channel of human nNOS. Introduction of an N-methylethane-1,2-diamine side chain and conformational constraints such as benzonitrile and pyridine as the middle aromatic linker were sufficient to increase human and rat nNOS binding affinity and inducible and endothelial NOS selectivity. We found that 14b is a potent inhibitor; the binding modes with human and rat nNOS are unexpected, inducing side chain rotamer changes in Gln478 (rat) at the top of the active site. Compound 19c exhibits Ki values of 24 and 55 nM for rat and human nNOS, respectively, with 153-fold iNOS and 1040-fold eNOS selectivity. 19c has 18% oral bioavailability.


Subject(s)
Aminopyridines/chemistry , Aminopyridines/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Aminopyridines/metabolism , Aminopyridines/pharmacokinetics , Animals , Biological Availability , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Humans , Male , Mice , Models, Molecular , Nitric Oxide Synthase Type I/chemistry , Nitric Oxide Synthase Type I/metabolism , Protein Conformation , Rats , Structure-Activity Relationship , Substrate Specificity
11.
J Phys Chem A ; 119(25): 6641-9, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26035438

ABSTRACT

Oxidation of L-arginine (L-Arg) to nitric oxide (NO) by NO synthase (NOS) takes place at the heme active site. It is of current interest to study structures of the heme species that activates O2 and transforms the substrate. The NOS ferrous-NO complex is a close mimic of the obligatory ferric (hydro)peroxo intermediate in NOS catalysis. In this work, pulsed electron-nuclear double resonance (ENDOR) spectroscopy was used to probe the hydrogen bonding of the NO ligand in the ferrous-NO heme center of neuronal NOS (nNOS) without a substrate and with L-Arg or N-hydroxy-L-arginine (NOHA) substrates. Unexpectedly, no H-bonding interaction connecting the NO ligand to the active site water molecule or the Arg substrate was detected, in contrast to the results obtained by X-ray crystallography for the Arg-bound nNOS heme domain [Li et al. J. Biol. Inorg. Chem. 2006, 11, 753-768]. The nearby exchangeable proton in both the no-substrate and Arg-containing nNOS samples is located outside the H-bonding range and, on the basis of the obtained structural constraints, can belong to the active site water (or OH). On the contrary, in the NOHA-bound sample, the nearby exchangeable hydrogen forms an H-bond with the NO ligand (on the basis of its distance from the NO ligand and a nonzero isotropic hfi constant), but it does not belong to the active site water molecule because the water oxygen atom (detected by (17)O ENDOR) is too far. This hydrogen should therefore come from the NOHA substrate, which is in agreement with the X-ray crystallography work [Li et al. Biochemistry 2009, 48, 10246-10254]. The nearby nonexchangeable hydrogen atom assigned as H(ε) of Phe584 was detected in all three samples. This hydrogen atom may have a stabilizing effect on the NO ligand and probably determines its position.


Subject(s)
Heme/chemistry , Nitric Oxide Synthase Type I/chemistry , Nitric Oxide/chemistry , Animals , Arginine/chemistry , Catalysis , Catalytic Domain , Electron Spin Resonance Spectroscopy , Hydrogen/chemistry , Hydrogen Bonding , Protons , Rats , Water/chemistry
12.
Biochem Biophys Res Commun ; 459(3): 393-7, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25732085

ABSTRACT

Neuronal nitric oxide synthase (nNOS) plays a critical role in regulating cardiomyocyte function. nNOS was reported to decrease superoxide production in the myocardium by inhibiting the function of xanthine oxidoreductase. However, the effect of oxidative stress on nNOS in cardiomyocytes has not been determined. We report here that brief exposure of HL-1 cardiomyocytes to hydrogen peroxide (H2O2) induces phosphorylation of nNOS at serine 1412. This increase in phosphorylation was concomitant with increased nitric oxide (NO) production. Prolonged exposure to the oxidant, however, resulted in decreased expression of the protein. H2O2 treatment for short periods also stimulated phosphorylation of AKT and AMPK. H2O2-induced phosphorylation of nNOS was reduced when AMPK activity was inhibited by compound C, suggesting that AMPK is a mediator of oxidative stress-induced phosphorylation of nNOS. However, inhibition of AKT activity by the pan AKT inhibitor, AKTi, had no effect on nNOS phosphorylation caused by H2O2. These data demonstrate the novel regulation of nNOS phosphorylation and expression by oxidative stress.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Nitric Oxide Synthase Type I/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Cell Line , Hydrogen Peroxide/pharmacology , Mice , Myocytes, Cardiac/drug effects , Nitric Oxide/metabolism , Oxidants/pharmacology , Oxidative Stress/drug effects , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors
13.
Am J Physiol Cell Physiol ; 308(8): C673-83, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25652452

ABSTRACT

Endothelial nitric oxide (NO) synthase (eNOS) is the predominant isoform that generates NO in the blood vessels. Many different regulators, including heat shock protein 90 (Hsp90), govern eNOS function. Hsp90-dependent phosphorylation of eNOS is a critical event that determines eNOS activity. In our earlier study we demonstrated an inhibitor-κB kinase-ß (IKKß)-Hsp90 interaction in a high-glucose environment. In the present study we further define the putative binding domain of IKKß on Hsp90. Interestingly, IKKß binds to the middle domain of Hsp90, which has been shown to interact with eNOS to stimulate its activity. This new finding suggests a tighter regulation of eNOS activity than was previously assumed. Furthermore, addition of purified recombinant IKKß to the eNOS-Hsp90 complex reduces the eNOS-Hsp90 interaction and eNOS activity, indicating a competition for Hsp90 between eNOS and IKKß. The pathophysiological relevance of the IKKß-Hsp90 interaction has also been demonstrated using in vitro vascular endothelial growth factor-mediated signaling and an Ins2(Akita) in vivo model. Our study further defines the preferential involvement of α- vs. ß-isoforms of Hsp90 in the IKKß-eNOS-Hsp90 interaction, even though both Hsp90α and Hsp90ß stimulate NO production. These studies not only reinforce the significance of maintaining a homeostatic balance of eNOS and IKKß within the cell system that regulates NO production, but they also confirm that the IKKß-Hsp90 interaction is favored in a high-glucose environment, leading to impairment of the eNOS-Hsp90 interaction, which contributes to endothelial dysfunction and vascular complications in diabetes.


Subject(s)
Endothelial Cells/metabolism , HSP90 Heat-Shock Proteins/metabolism , I-kappa B Kinase/metabolism , Nitric Oxide Synthase Type III/metabolism , Animals , Aorta/cytology , Binding Sites , Cattle , Cells, Cultured , Diabetes Mellitus/pathology , Endothelial Cells/enzymology , Glucose/metabolism , HSP90 Heat-Shock Proteins/genetics , Humans , I-kappa B Kinase/genetics , Insulin/metabolism , Mice , Mice, Inbred NOD , Nitric Oxide/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering , Vascular Endothelial Growth Factor A/metabolism
14.
J Med Chem ; 58(3): 1067-88, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25489882

ABSTRACT

Selective inhibition of neuronal nitric oxide synthase (nNOS) is an important therapeutic approach to target neurodegenerative disorders. However, the majority of the nNOS inhibitors developed are arginine mimetics and, therefore, suffer from poor bioavailability. We designed a novel strategy to combine a more pharmacokinetically favorable 2-imidazolylpyrimidine head with promising structural components from previous inhibitors. In conjunction with extensive structure-activity studies, several highly potent and selective inhibitors of nNOS were discovered. X-ray crystallographic analysis reveals that these type II inhibitors utilize the same hydrophobic pocket to gain strong inhibitory potency (13), as well as high isoform selectivity. Interestingly, select compounds from this series (9) showed good permeability and low efflux in a Caco-2 assay, suggesting potential oral bioavailability, and exhibited minimal off-target binding to 50 central nervous system receptors. Furthermore, even with heme-coordinating groups in the molecule, modifying other pharmacophoric fragments minimized undesirable inhibition of cytochrome P450s from human liver microsomes.


Subject(s)
Cell Membrane Permeability/drug effects , Nitric Oxide Synthase Type I/antagonists & inhibitors , Pyrimidines/pharmacology , Animals , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Nitric Oxide Synthase Type I/metabolism , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Rats , Structure-Activity Relationship
15.
J Struct Biol ; 188(1): 46-54, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25175399

ABSTRACT

While the three-dimensional structures of heme- and flavin-binding domains of the NOS isoforms have been determined, the structures of the holoenzymes remained elusive. Application of electron cryo-microscopy and structural modeling of the bovine endothelial nitric oxide synthase (eNOS) holoenzyme produced detailed models of the intact holoenzyme in the presence and absence of Ca(2+)/calmodulin (CaM). These models accommodate the cross-electron transfer from the reductase in one monomer to the heme in the opposite monomer. The heme domain acts as the anchoring dimeric structure for the entire enzyme molecule, while the FMN domain is activated by CaM to move flexibly to bridge the distance between the reductase and oxygenase domains. Our results indicate that the key regulatory role of CaM involves the stabilization of structural intermediates and precise positioning of the pivot for the FMN domain tethered shuttling motion to accommodate efficient and rapid electron transfer in the homodimer of eNOS.


Subject(s)
Calmodulin/metabolism , Flavin Mononucleotide/chemistry , Holoenzymes/chemistry , Nitric Oxide Synthase Type III/chemistry , Allosteric Regulation , Animals , Calcium/chemistry , Calcium/metabolism , Calmodulin/chemistry , Cattle , Electron Transport , Heme/chemistry , Kinetics , Nitric Oxide Synthase Type III/metabolism , Oxidation-Reduction , Protein Structure, Tertiary
16.
Bioorg Med Chem Lett ; 24(18): 4504-4510, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25149509

ABSTRACT

To develop potent and selective nNOS inhibitors, a new series of double-headed molecules with chiral linkers that derive from natural amino acid derivatives have been designed and synthesized. The new structures integrate a thiophenecarboximidamide head with two types of chiral linkers, presenting easy synthesis and good inhibitory properties. Inhibitor (S)-9b exhibits a potency of 14.7 nM against nNOS and is 1134 and 322-fold more selective for nNOS over eNOS and iNOS, respectively. Crystal structures show that the additional binding between the aminomethyl moiety of 9b and propionate A on the heme and tetrahydrobiopterin (H4B) in nNOS, but not eNOS, contributes to its high selectivity. This work demonstrates the advantage of integrating known structures into structure optimization, and it should be possible to more readily develop compounds that incorporate bioavailability with these advanced features. Moreover, this integrative strategy is a general approach in new drug discovery.


Subject(s)
Enzyme Inhibitors/pharmacology , Imides/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Thiophenes/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Imides/chemical synthesis , Imides/chemistry , Models, Molecular , Molecular Structure , Nitric Oxide Synthase Type I/metabolism , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry
17.
J Med Chem ; 57(10): 4382-96, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24758147

ABSTRACT

Overproduction of NO by nNOS is implicated in the pathogenesis of diverse neuronal disorders. Since NO signaling is involved in diverse physiological functions, selective inhibition of nNOS over other isoforms is essential to minimize side effects. A series of α-amino functionalized aminopyridine derivatives (3-8) were designed to probe the structure-activity relationship between ligand, heme propionate, and H4B. Compound 8R was identified as the most potent and selective molecule of this study, exhibiting a Ki of 24 nM for nNOS, with 273-fold and 2822-fold selectivity against iNOS and eNOS, respectively. Although crystal structures of 8R complexed with nNOS and eNOS revealed a similar binding mode, the selectivity stems from the distinct electrostatic environments in two isoforms that result in much lower inhibitor binding free energy in nNOS than in eNOS. These findings provide a basis for further development of simple, but even more selective and potent, nNOS inhibitors.


Subject(s)
Biopterins/analogs & derivatives , Enzyme Inhibitors/chemical synthesis , Heme/analogs & derivatives , Nitric Oxide Synthase/antagonists & inhibitors , Biopterins/chemistry , Enzyme Inhibitors/pharmacology , Heme/chemistry , Humans , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type III/antagonists & inhibitors , Structure-Activity Relationship
18.
J Biol Chem ; 289(19): 13273-83, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24648521

ABSTRACT

Caveolin-1 (Cav-1) gene inactivation interferes with caveolae formation and causes a range of cardiovascular and pulmonary complications in vivo. Recent evidence suggests that blunted Cav-1/endothelial nitric-oxide synthase (eNOS) interaction, which occurs specifically in vascular endothelial cells, is responsible for the multiple phenotypes observed in Cav-1-null animals. Under basal conditions, Cav-1 binds eNOS and inhibits nitric oxide (NO) production via the Cav-1 scaffolding domain (CAV; amino acids 82-101). Although we have recently shown that CAV residue Phe-92 is responsible for eNOS inhibition, the "inactive" F92A Cav-1 mutant unexpectedly retains its eNOS binding ability and can increase NO release, indicating the presence of a distinct eNOS binding domain within CAV. Herein, we identified and characterized a small 10-amino acid CAV subsequence (90-99) that accounted for the majority of eNOS association with Cav-1 (Kd = 49 nM), and computer modeling of CAV(90-99) docking to eNOS provides a rationale for the mechanism of eNOS inhibition by Phe-92. Finally, using gene silencing and reconstituted cell systems, we show that intracellular delivery of a F92A CAV(90-99) peptide can promote NO bioavailability in eNOS- and Cav-1-dependent fashions. To our knowledge, these data provide the first detailed analysis of Cav-1 binding to one of its most significant client proteins, eNOS.


Subject(s)
Caveolin 1 , Computer Simulation , Endothelial Cells/metabolism , Models, Molecular , Nitric Oxide Synthase Type III , Amino Acid Substitution , Animals , Cattle , Caveolin 1/chemistry , Caveolin 1/genetics , Caveolin 1/metabolism , Cells, Cultured , Endothelial Cells/cytology , Humans , Mutation, Missense , Nitric Oxide/chemistry , Nitric Oxide/genetics , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/chemistry , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Protein Binding
19.
ACS Med Chem Lett ; 5(1): 56-60, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24660051

ABSTRACT

The three important mammalian isozymes of nitric oxide synthase (NOS) are neuronal NOS (nNOS), endothelial NOS (eNOS), and inducible NOS (iNOS). Inhibitors of nNOS show promise as treatments for neurodegenerative diseases. Eight easily-synthesized compounds containing either one (20a,b) or two (9a-d; 15a,b) 2-amino-4-methylpyridine groups with a chiral pyrrolidine linker were designed as selective nNOS inhibitors. Inhibitor 9c is the best of these compounds, having a potency of 9.7 nM and dual selectivity of 693 and 295 against eNOS and iNOS, respectively. Crystal structures of nNOS complexed with either 9a or 9c show a double-headed binding mode, where each 2-aminopyridine head group interacts with either a nNOS active site Glu residue or a heme propionate. In addition, the pyrrolidine nitrogen of 9c contributes additional hydrogen bonds to the heme propionate, resulting in a unique binding orientation. In contrast, the lack of hydrogen bonds from the pyrrolidine of 9a to the heme propionate allows the inhibitor to adopt two different binding orientations. Both 9a and 9c bind to eNOS in a single-headed mode, which is the structural basis for the isozyme selectivity.

20.
J Med Chem ; 57(4): 1513-30, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24472039

ABSTRACT

Since high levels of nitric oxide (NO) are implicated in neurodegenerative disorders, inhibition of the neuronal isoform of nitric oxide synthase (nNOS) and reduction of NO levels are therapeutically desirable. Nonetheless, many nNOS inhibitors mimic l-arginine and are poorly bioavailable. 2-Aminoquinoline-based scaffolds were designed with the hope that they could (a) mimic aminopyridines as potent, isoform-selective arginine isosteres and (b) possess chemical properties more conducive to oral bioavailability and CNS penetration. A series of these compounds was synthesized and assayed against purified nNOS enzymes, endothelial NOS (eNOS), and inducible NOS (iNOS). Several compounds built on a 7-substituted 2-aminoquinoline core are potent and isoform-selective; X-ray crystallography indicates that aminoquinolines exert inhibitory effects by mimicking substrate interactions with the conserved active site glutamate residue. The most potent and selective compounds, 7 and 15, were tested in a Caco-2 assay and showed good permeability and low efflux, suggesting high potential for oral bioavailability.


Subject(s)
Aminoquinolines/chemistry , Enzyme Inhibitors/chemistry , Nitric Oxide Synthase Type I/antagonists & inhibitors , Aminoquinolines/pharmacology , Catalytic Domain , Enzyme Inhibitors/pharmacology , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Molecular
SELECTION OF CITATIONS
SEARCH DETAIL
...