Thomas L. Poulos

Find an error

Name: Poulos, Thomas
Organization: University of California , USA
Department: Departments of Molecular Biology and Biochemistry
Title: (PhD)

TOPICS

Co-reporter:Scott A. Hollingsworth, Brian D. Nguyen, Georges Chreifi, Anton P. Arce, and Thomas L. Poulos
Journal of Chemical Information and Modeling September 25, 2017 Volume 57(Issue 9) pp:2344-2344
Publication Date(Web):August 25, 2017
DOI:10.1021/acs.jcim.7b00421
Leishmania major peroxidase (LmP) is structurally and functionally similar to the well-studied yeast Cytochrome c peroxidase (CCP). A recent Brownian dynamics study showed that L. major Cytochrome c (LmCytc) associates with LmP by forming an initial complex with the N-terminal helix A of LmP, followed by a movement toward the electron transfer (ET) site observed in the LmP–LmCytc crystal structure. Critical to forming the active electron transfer complex is an intermolecular Arg-Asp ion pair at the center of the interface. If the dissociation reaction is effectively the reverse of the association reaction, then rupture of the Asp-Arg ion pair should be followed by movement of LmCytc back toward LmP helix A. To test this possibility, we have performed multiple molecular dynamics (MD) simulations of the LmP–LmCytc complex. In five separate simulations, LmCytc is observed to indeed move toward helix A, and in two of the simulations, the Asp–Arg ion pair breaks, which frees LmCytc to fully associate with the LmP helix A secondary binding site. These results support the “bind and crawl” or “velcro” mechanism of association, wherein LmCytc forms a nonspecific electrostatic complex with LmP helix A, followed by a “crawl” toward the ET-active site, where the Asp–Arg ion pair holds the LmCytc in position for rapid ET. These simulations also point to Tyr134LmP as being important in the association/dissociation reactions. Experimentally mutating Tyr134 to Phe was found to decrease Km by 3.6-fold, which is consistent with its predicted role in complex formation by MD simulations.
Co-reporter:Dipanwita Batabyal, Logan S. Richards, and Thomas L. Poulos
Journal of the American Chemical Society September 20, 2017 Volume 139(Issue 37) pp:13193-13193
Publication Date(Web):August 21, 2017
DOI:10.1021/jacs.7b07656
Previous crystal structures of cytochrome P450cam complexed with its redox partner, putidaredoxin (Pdx), shows that P450cam adopts the open conformation. It has been hypothesized that the Pdx-induced shift toward the open state frees the essential Asp251 from salt bridges with Arg186 and Lys178 so that Asp251 can participate in a proton relay network required for O2 activation. This in part explains why P450cam has such a strict requirement for Pdx. One problem with this view is that looser substrate–protein interactions in the open state may not be compatible with the observed regio- and stereoselective hydroxylation. In the present study, molecular dynamics simulations show that Pdx binding favors a conformation that stabilizes the active site and decreases camphor mobility yet retains a partially open conformation compatible with the required proton relay network. The R186A mutant which frees Asp251 in the absence of Pdx retains good enzyme activity, and the crystal structure shows that product, 5-exo-hydroxycamphor, is bound. This indicates that rupture of the Asp251–Arg186 relaxes selectivity with respect to source of electrons and enables X-ray generated reducing equivalents to support substrate hydroxylation. These combined computational and experimental results are consistent with the proposed role of Pdx in assisting the release of Asp251 from ion pairs so that it can participate in proton-coupled electron transfer.
Co-reporter:Maris A. Cinelli, Huiying Li, Georges Chreifi, Thomas L. Poulos, and Richard B. Silverman
Journal of Medicinal Chemistry May 11, 2017 Volume 60(Issue 9) pp:3958-3958
Publication Date(Web):April 19, 2017
DOI:10.1021/acs.jmedchem.7b00259
Neuronal nitric oxide synthase (nNOS) inhibition is a promising strategy to treat neurodegenerative disorders, but the development of nNOS inhibitors is often hindered by poor pharmacokinetics. We previously developed a class of membrane-permeable 2-aminoquinoline inhibitors and later rearranged the scaffold to decrease off-target binding. However, the resulting compounds had decreased permeability, low human nNOS activity, and low selectivity versus human eNOS. In this study, 5-substituted phenyl ether-linked aminoquinolines and derivatives were synthesized and assayed against purified NOS isoforms. 5-Cyano compounds are especially potent and selective rat and human nNOS inhibitors. Activity and selectivity are mediated by the binding of the cyano group to a new auxiliary pocket in nNOS. Potency was enhanced by methylation of the quinoline and by introduction of simple chiral moieties, resulting in a combination of hydrophobic and auxiliary pocket effects that yielded high (∼500-fold) n/e selectivity. Importantly, the Caco-2 assay also revealed improved membrane permeability over previous compounds.
Co-reporter:Anthony V. Pensa, Maris A. Cinelli, Huiying Li, Georges Chreifi, Paramita Mukherjee, Linda J. Roman, Pavel Martásek, Thomas L. Poulos, and Richard B. Silverman
Journal of Medicinal Chemistry August 24, 2017 Volume 60(Issue 16) pp:7146-7146
Publication Date(Web):August 4, 2017
DOI:10.1021/acs.jmedchem.7b00835
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.
Co-reporter:Georges Chreifi;Dillon Dejam
JBIC Journal of Biological Inorganic Chemistry 2017 Volume 22( Issue 6) pp:919-927
Publication Date(Web):05 June 2017
DOI:10.1007/s00775-017-1469-9
Leishmania major pseudoperoxidase (LmPP) is a recently discovered heme protein expressed by the human pathogen. Previous in vivo and in vitro studies suggest that LmPP is a crucial element of the pathogen’s defense mechanism against the reactive nitrogen species peroxynitrite produced during the host immune response. To shed light on the potential mechanism of peroxynitrite detoxification, we have determined the 1.76-Å X-ray crystal structure of LmPP, revealing a striking degree of homology with heme peroxidases. The most outstanding structural feature is a Cys/His heme coordination, which corroborates previous spectroscopic and mutagenesis studies. We also used a combination of stopped-flow and electron paramagnetic spectroscopies that together suggest that peroxynitrite is not a substrate for LmPP catalysis, leaving the function of LmPP an open question.
Co-reporter:Heng-Yen Wang; Yajuan Qin; Huiying Li; Linda J. Roman; Pavel Martásek; Thomas L. Poulos;Richard B. Silverman
Journal of Medicinal Chemistry 2016 Volume 59(Issue 10) pp:4913-4925
Publication Date(Web):April 6, 2016
DOI:10.1021/acs.jmedchem.6b00273
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.
Co-reporter:Jeffrey K. Holden, Matthew C. Lewis, Maris A. Cinelli, Ziad Abdullatif, Anthony V. Pensa, Richard B. Silverman, and Thomas L. Poulos
Biochemistry 2016 Volume 55(Issue 39) pp:5587
Publication Date(Web):September 8, 2016
DOI:10.1021/acs.biochem.6b00786
Nitric oxide is produced in Gram-positive pathogens Bacillus anthracis and Staphylococcus aureus by the bacterial isoform of nitric oxide synthase (NOS). Inhibition of bacterial nitric oxide synthase (bNOS) has been identified as a promising antibacterial strategy for targeting methicillin-resistant S. aureus [Holden, J. K., et al. (2015) Chem. Biol. 22, 785–779]. One class of NOS inhibitors that demonstrates antimicrobial efficacy utilizes an aminoquinoline scaffold. Here we report on a variety of aminoquinolines that target the bacterial NOS active site, in part, by binding to a hydrophobic patch that is unique to bNOS. Through mutagenesis and crystallographic studies, our findings demonstrate that aminoquinolines are an excellent scaffold for further aiding in the development of bNOS specific inhibitors.
Co-reporter:Huiying Li, Heng-Yen Wang, Soosung Kang, Richard B. Silverman, and Thomas L. Poulos
Biochemistry 2016 Volume 55(Issue 26) pp:3702-3707
Publication Date(Web):June 2, 2016
DOI:10.1021/acs.biochem.6b00261
Development of potent and isoform selective nitric oxide synthase (NOS) inhibitors is challenging because of the structural similarity in the heme active sites. One amino acid difference between NOS isoforms, Asp597 in rat neuronal NOS (nNOS) versus Asn368 in bovine endothelial NOS (eNOS), has been identified as the structural basis for why some dipeptide amide inhibitors bind more tightly to nNOS than to eNOS. We now have found that the same amino acid variation is responsible for substantially different binding modes and affinity for a new class of aminopyridine-based inhibitors.
Co-reporter:Dipanwita Batabyal, Ariel Lewis-Ballester, Syun-Ru Yeh, and Thomas L. Poulos
Biochemistry 2016 Volume 55(Issue 47) pp:
Publication Date(Web):November 3, 2016
DOI:10.1021/acs.biochem.6b00913
The camphor monooxygenase, cytochrome P450cam, exhibits a strict requirement for its own redox partner, putidaredoxin (Pdx), a two-iron–sulfur ferredoxin. The closest homologue to P450cam, CYP101D1, is structurally very similar, uses a similar redox partner, and exhibits nearly identical enzymatic properties in the monooxygenation of camphor to give the same single 5-exo-hydroxy camphor product. However, CYP101D1 does not strictly require its own ferredoxin (Arx) for activity because Pdx can support CYP101D1 catalysis but Arx cannot support P450cam catalysis. We have further examined the differences between these two P450s by determining the effect of spin equilibrium, redox properties, and stability of oxygen complexes. We find that Arx shifts the spin state equilibrium toward high-spin, which is the opposite of the effect of Pdx on P450cam. In both P450s, redox partner binding destabilizes the oxy–P450 complex but this effect is much weaker with CYP101D1. In addition, resonance Raman data show that structural perturbations observed in P450cam upon addition of Pdx are absent in CYP101D1. These data indicate that Arx does not play the same effector role in catalysis as Pdx does with P450cam. The most relevant structural difference between these two P450s centers on a catalytically important Asp residue required for proton-coupled electron transfer. We postulate that with P450cam larger Pdx-assisted motions are required to free this Asp for catalysis while the smaller number of restrictions in CYP101D1 precludes the need for redox partner-assisted structural changes.
Co-reporter:Scott A. Hollingsworth;Dipanwita Batabyal;Brian D. Nguyen;
Proceedings of the National Academy of Sciences 2016 113(31) pp:8723-8728
Publication Date(Web):July 20, 2016
DOI:10.1073/pnas.1606474113
Co-reporter:Georges Chreifi;Elizabeth L. Baxter;Tzanko Doukov;Aina E. Cohen;Jinhu Song;Yergalem T. Meharenna;Scott E. McPhillips;S. Michael Soltis
PNAS 2016 Volume 113 (Issue 5 ) pp:1226-1231
Publication Date(Web):2016-02-02
DOI:10.1073/pnas.1521664113
The reaction of peroxides with peroxidases oxidizes the heme iron from Fe(III) to Fe(IV)=O and a porphyrin or aromatic side chain to a cationic radical. X-ray–generated hydrated electrons rapidly reduce Fe(IV), thereby requiring very short exposures using many crystals, and, even then, some reduction cannot be avoided. The new generation of X-ray free electron lasers capable of generating intense X-rays on the tenths of femtosecond time scale enables structure determination with no reduction or X-ray damage. Here, we report the 1.5-Å crystal structure of cytochrome c peroxidase (CCP) compound I (CmpI) using data obtained with the Stanford Linear Coherent Light Source (LCLS). This structure is consistent with previous structures. Of particular importance is the active site water structure that can mediate the proton transfer reactions required for both CmpI formation and reduction of Fe(IV)=O to Fe(III)-OH. The structures indicate that a water molecule is ideally positioned to shuttle protons between an iron-linked oxygen and the active site catalytic His. We therefore have carried out both computational and kinetic studies to probe the reduction of Fe(IV)=O. Kinetic solvent isotope experiments show that the transfer of a single proton is critical in the peroxidase rate-limiting step, which is very likely the proton-coupled reduction of Fe(IV)=O to Fe(III)-OH. We also find that the pKa of the catalytic His substantially increases in CmpI, indicating that this active site His is the source of the proton required in the reduction of Fe(IV)=O to Fe(IV)-OH.
Co-reporter:Wei Tang; Huiying Li; Emma H. Doud; Yunqiu Chen; Stephanie Choing; Carla Plaza; Neil L. Kelleher; Thomas L. Poulos;Richard B. Silverman
Journal of the American Chemical Society 2015 Volume 137(Issue 18) pp:5980-5989
Publication Date(Web):April 15, 2015
DOI:10.1021/jacs.5b01202
Nitric oxide synthase (NOS) catalyzes the conversion of l-arginine to l-citrulline and the second messenger nitric oxide. Three mechanistic pathways are proposed for the inactivation of neuronal NOS (nNOS) by (S)-2-amino-5-(2-(methylthio)acetimidamido)pentanoic acid (1): sulfide oxidation, oxidative dethiolation, and oxidative demethylation. Four possible intermediates were synthesized. All compounds were assayed with nNOS, their IC50, KI, and kinact values were obtained, and their crystal structures were determined. The identification and characterization of the products formed during inactivation provide evidence for the details of the inactivation mechanism. On the basis of these studies, the most probable mechanism for the inactivation of nNOS involves oxidative demethylation with the resulting thiol coordinating to the cofactor heme iron. Although nNOS is a heme-containing enzyme, this is the first example of a NOS that catalyzes an S-demethylation reaction; the novel mechanism of inactivation described here could be applied to the design of inactivators of other heme-dependent enzymes.
Co-reporter:Paramita Mukherjee; Huiying Li; Irina Sevrioukova; Georges Chreifi; Pavel Martásek; Linda J. Roman; Thomas L. Poulos;Richard B. Silverman
Journal of Medicinal Chemistry 2015 Volume 58(Issue 3) pp:1067-1088
Publication Date(Web):December 9, 2014
DOI:10.1021/jm501719e
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.
Co-reporter:Jeffrey K. Holden, Soosung Kang, Federico C. Beasley, Maris A. Cinelli, Huiying Li, Saurabh G. Roy, Dillon Dejam, Aimee L. Edinger, Victor Nizet, Richard B. Silverman, Thomas L. Poulos
Chemistry & Biology 2015 Volume 22(Issue 6) pp:785-792
Publication Date(Web):18 June 2015
DOI:10.1016/j.chembiol.2015.05.013
•Inhibitors selective toward bacterial nitric oxide synthase have been identified•These inhibitors are antimicrobial against MRSA•Crystallography reveals the structural basis for selectivity•NOS inhibitor library rapidly screened to identify potent inhibitorsBacterial infections associated with methicillin-resistant Staphylococcus aureus (MRSA) are a major economic burden to hospitals, and confer high rates of morbidity and mortality among those infected. Exploitation of novel therapeutic targets is thus necessary to combat this dangerous pathogen. Here, we report on the identification and characterization, including crystal structures, of two nitric oxide synthase (NOS) inhibitors that function as antimicrobials against MRSA. These data provide the first evidence that bacterial NOS (bNOS) inhibitors can work synergistically with oxidative stress to enhance MRSA killing. Crystal structures show that each inhibitor contacts an active site Ile residue in bNOS that is Val in the mammalian NOS isoforms. Mutagenesis studies show that the additional nonpolar contacts provided by the Ile in bNOS contribute to tighter binding toward the bacterial enzyme.Figure optionsDownload full-size imageDownload high-quality image (156 K)Download as PowerPoint slide
Co-reporter:Jeffrey K. Holden; Soosung Kang; Scott A. Hollingsworth; Huiying Li; Nathan Lim; Steven Chen; He Huang; Fengtian Xue; Wei Tang; Richard B. Silverman
Journal of Medicinal Chemistry 2015 Volume 58(Issue 2) pp:994-1004
Publication Date(Web):December 18, 2014
DOI:10.1021/jm501723p
Inhibition of bacterial nitric oxide synthase (bNOS) has the potential to improve the efficacy of antimicrobials used to treat infections by Gram-positive pathogens Staphylococcus aureus and Bacillus anthracis. However, inhibitor specificity toward bNOS over the mammalian NOS (mNOS) isoforms remains a challenge because of the near identical NOS active sites. One key structural difference between the NOS isoforms is the amino acid composition of the pterin cofactor binding site that is adjacent to the NOS active site. Previously, we demonstrated that a NOS inhibitor targeting both the active and pterin sites was potent and functioned as an antimicrobial (Holden, , Proc. Natl. Acad. Sci. U.S.A. 2013, 110, 18127). Here we present additional crystal structures, binding analyses, and bacterial killing studies of inhibitors that target both the active and pterin sites of a bNOS and function as antimicrobials. Together, these data provide a framework for continued development of bNOS inhibitors, as each molecule represents an excellent chemical scaffold for the design of isoform selective bNOS inhibitors.
Co-reporter:Soosung Kang; Huiying Li; Wei Tang; Pavel Martásek; Linda J. Roman; Thomas L. Poulos;Richard B. Silverman
Journal of Medicinal Chemistry 2015 Volume 58(Issue 14) pp:5548-5560
Publication Date(Web):June 29, 2015
DOI:10.1021/acs.jmedchem.5b00573
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.
Co-reporter:Maris A. Cinelli; Huiying Li; Anthony V. Pensa; Soosung Kang; Linda J. Roman; Pavel Martásek; Thomas L. Poulos;Richard B. Silverman
Journal of Medicinal Chemistry 2015 Volume 58(Issue 21) pp:8694-8712
Publication Date(Web):October 15, 2015
DOI:10.1021/acs.jmedchem.5b01330
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.
Co-reporter:James B. Fields, Scott A. Hollingsworth, Georges Chreifi, Matthias Heyden, Anton P. Arce, Hugo I. Magaña-Garcia, Thomas L. Poulos, and Douglas J. Tobias
Biochemistry 2015 Volume 54(Issue 49) pp:7272-7282
Publication Date(Web):November 24, 2015
DOI:10.1021/acs.biochem.5b00569
Leishmania major, the parasitic causative agent of leishmaniasis, produces a heme peroxidase (LmP), which catalyzes the peroxidation of mitochondrial cytochrome c (LmCytc) for protection from reactive oxygen species produced by the host. The association of LmP and LmCytc, which is known from kinetics measurements to be very fast (∼108 M–1 s–1), does not involve major conformational changes and has been suggested to be dominated by electrostatic interactions. We used Brownian dynamics simulations to investigate the mechanism of formation of the LmP–LmCytc complex. Our simulations confirm the importance of electrostatic interactions involving the negatively charged D211 residue at the LmP active site, and reveal a previously unrecognized role in complex formation for negatively charged residues in helix A of LmP. The crystal structure of the D211N mutant of LmP reported herein is essentially identical to that of wild-type LmP, reinforcing the notion that it is the loss of charge at the active site, and not a change in structure, that reduces the association rate of the D211N variant of LmP. The Brownian dynamics simulations further show that complex formation occurs via a “bind and crawl” mechanism, in which LmCytc first docks to a location on helix A that is far from the active site, forming an initial encounter complex, and then moves along helix A to the active site. An atomistic molecular dynamics simulation confirms the helix A binding site, and steady state activity assays and stopped-flow kinetics measurements confirm the role of helix A charges in the association mechanism.
Co-reporter:Wei Tang, Huiying Li, Thomas L. Poulos, and Richard B. Silverman
Biochemistry 2015 Volume 54(Issue 15) pp:2530-2538
Publication Date(Web):March 26, 2015
DOI:10.1021/acs.biochem.5b00135
Nitric oxide synthase (NOS) catalyzes the conversion of l-arginine to l-citrulline and nitric oxide. N5-(1-Iminoethyl)-l-ornithine (l-NIO), an amidine-containing molecule, is a natural product known to be an inactivator of inducible NOS (iNOS). Because of the presence of the amidine methyl group in place of the guanidine amino group of substrate l-arginine, the active site heme peroxy intermediate sometimes cannot be protonated, thereby preventing its conversion to the heme oxo intermediate; instead, a heme oxygenase-type mechanism occurs, leading to conversion of the heme to biliverdin. This might be a new and general inactivation mechanism for heme-containing enzymes. In the studies described here, we attempted to provide support for amidines as substrates and inactivators of iNOS by the design and synthesis of amidine analogues of l-NIO having groups other than the amidine methyl group. No nitric oxide- or enzyme-catalyzed products could be detected by incubation of these amidines with iNOS. Although none of the l-NIO analogues acted as substrates, they all inhibited iNOS; increased inhibitory potency correlated with decreased substituent size. Computer modeling and molecular dynamics simulations were run on 10 and 11 to rationalize why these compounds do not act as substrates. Unlike the methyl amidine (l-NIO), the other alkyl groups block binding of O2 at the heme iron. Compounds 8, 9, and 11 were inactivators; however, no heme was lost, and no biliverdin was formed. No kinetic isotope effect on inactivation was observed with perdeuterated ethyl 8. A small amount of dimer disruption occurred with these inactivators, although the amount would not account for complete enzyme inactivation. The l-NIO analogues inactivate iNOS by a yet unknown mechanism; however, it is different from that of l-NIO, and the inactivation mechanism previously reported for l-NIO appears to be unique to methyl amidines.
Co-reporter:Jeffrey K. Holden, Dillon Dejam, Matthew C. Lewis, He Huang, Soosung Kang, Qing Jing, Fengtian Xue, Richard B. Silverman, and Thomas L. Poulos
Biochemistry 2015 Volume 54(Issue 26) pp:4075-4082
Publication Date(Web):June 11, 2015
DOI:10.1021/acs.biochem.5b00431
Nitric oxide generated by bacterial nitric oxide synthase (NOS) increases the susceptibility of Gram-positive pathogens Staphylococcus aureus and Bacillus anthracis to oxidative stress, including antibiotic-induced oxidative stress. Not surprisingly, NOS inhibitors also improve the effectiveness of antimicrobials. Development of potent and selective bacterial NOS inhibitors is complicated by the high active site sequence and structural conservation shared with the mammalian NOS isoforms. To exploit bacterial NOS for the development of new therapeutics, recognition of alternative NOS surfaces and pharmacophores suitable for drug binding is required. Here, we report on a wide number of inhibitor-bound bacterial NOS crystal structures to identify several compounds that interact with surfaces unique to the bacterial NOS. Although binding studies indicate that these inhibitors weakly interact with the NOS active site, many of the inhibitors reported here provide a revised structural framework for the development of new antimicrobials that target bacterial NOS. In addition, mutagenesis studies reveal several key residues that unlock access to bacterial NOS surfaces that could provide the selectivity required to develop potent bacterial NOS inhibitors.
Co-reporter:Georges Chreifi, Scott A. Hollingsworth, Huiying Li, Sarvind Tripathi, Anton P. Arce, Hugo I. Magaña-Garcia, and Thomas L. Poulos
Biochemistry 2015 Volume 54(Issue 21) pp:3328-3336
Publication Date(Web):May 5, 2015
DOI:10.1021/acs.biochem.5b00338
Leishmania major peroxidase (LmP) is very similar to the well-known yeast cytochrome c peroxidase (CcP). Both enzymes catalyze the peroxidation of cytochrome c. Like CcP, LmP reacts with H2O2 to form Compound I, which consists of a ferryl heme and a Trp radical, FeIV═O;Trp•+. Cytochrome c (Cytc) reduces the Trp radical to give Compound II, FeIV═O;Trp, which is followed by an intramolecular electron transfer to give FeIII–OH;Trp•+, and in the last step, Cytc reduces the Trp radical. In this study, we have used steady-state and single-turnover kinetics to improve our understanding of the overall mechanism of LmP catalysis. While the activity of CcP greatly increases with ionic strength, the kcat for LmP remains relatively constant at all ionic strengths tested. Therefore, unlike CcP, where dissociation of oxidized Cytc is limiting at low ionic strengths, association/dissociation reactions are not limiting at any ionic strength in LmP. We conclude that in LmP, the intramolecular electron transfer reaction, FeIV═O;Trp to FeIII–OH;Trp•+, is limiting at all ionic strengths. Unlike CcP, LmP depends on key intermolecular ion pairs to form the electron transfer competent complex. Mutating these sites causes the initial rate of association to decrease by 2 orders of magnitude and a substantial decrease in kcat. The drop in kcat is due to a switch in the rate-limiting step of the mutants from intramolecular electron transfer to the rate of association in forming the LmP–LmCytc complex. These studies show that while LmP and CcP form very similar complexes and exhibit similar activities, they substantially differ in how their activity changes as a function of ionic strength. This difference is primarily due to the heavy reliance of LmP on highly specific intermolecular ion pairs, while CcP relies mainly on nonpolar interactions.
Co-reporter:Thomas L. Poulos
Chemical Reviews 2014 Volume 114(Issue 7) pp:3919
Publication Date(Web):January 8, 2014
DOI:10.1021/cr400415k
Co-reporter:He Huang ; Huiying Li ; Sun Yang ; Georges Chreifi ; Pavel Martásek ; Linda J. Roman ; Frank L. Meyskens ; Thomas L. Poulos ;Richard B. Silverman
Journal of Medicinal Chemistry 2014 Volume 57(Issue 3) pp:686-700
Publication Date(Web):January 21, 2014
DOI:10.1021/jm401252e
Selective inhibitors of neuronal nitric oxide synthase (nNOS) are regarded as valuable and powerful agents with therapeutic potential for the treatment of chronic neurodegenerative pathologies and human melanoma. Here, we describe a novel hybrid strategy that combines the pharmacokinetically promising thiophene-2-carboximidamide fragment and structural features of our previously reported potent and selective aminopyridine inhibitors. Two inhibitors, 13 and 14, show low nanomolar inhibitory potency (Ki = 5 nM for nNOS) and good isoform selectivities (nNOS over eNOS [440- and 540-fold, respectively] and over iNOS [260- and 340-fold, respectively]). The crystal structures of these nNOS–inhibitor complexes reveal a new hot spot that explains the selectivity of 14 and why converting the secondary to tertiary amine leads to enhanced selectivity. More importantly, these compounds are the first highly potent and selective nNOS inhibitory agents that exhibit excellent in vitro efficacy in melanoma cell lines.
Co-reporter:Maris A. Cinelli ; Huiying Li ; Georges Chreifi ; Pavel Martásek ; Linda J. Roman ; Thomas L. Poulos ;Richard B. Silverman
Journal of Medicinal Chemistry 2014 Volume 57(Issue 4) pp:1513-1530
Publication Date(Web):January 28, 2014
DOI:10.1021/jm401838x
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.
Co-reporter:Soosung Kang ; Wei Tang ; Huiying Li ; Georges Chreifi ; Pavel Martásek ; Linda J. Roman ; Thomas L. Poulos ;Richard B. Silverman
Journal of Medicinal Chemistry 2014 Volume 57(Issue 10) pp:4382-4396
Publication Date(Web):April 23, 2014
DOI:10.1021/jm5004182
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.
Co-reporter:Yarrow Madrona, Scott A. Hollingsworth, Sarvind Tripathi, James B. Fields, Jean-Christophe N. Rwigema, Douglas J. Tobias, and Thomas L. Poulos
Biochemistry 2014 Volume 53(Issue 9) pp:
Publication Date(Web):February 17, 2014
DOI:10.1021/bi500010m
The crystal structure of the flavin mononucleotide (FMN)-containing redox partner to P450cin, cindoxin (Cdx), has been determined to 1.3 Å resolution. The overall structure is similar to that of the FMN domain of human cytochrome P450 reductase. A Brownian dynamics–molecular dynamics docking method was used to produce a model of Cdx with its redox partner, P450cin. This Cdx–P450cin model highlights the potential importance of Cdx Tyr96 in bridging the FMN and heme cofactors as well P450cin Arg102 and Arg346. Each of the single-site Ala mutants exhibits ∼10% of the wild-type activity, thus demonstrating the importance of these residues for binding and/or electron transfer. In the well-studied P450cam system, redox partner binding stabilizes the open low-spin conformation of P450cam and greatly decreases the stability of the oxy complex. In sharp contrast, Cdx does not shift P450cin to a low-spin state, although the stability of oxy-P450cin is decreased 10-fold in the presence of Cdx. This indicates that Cdx may have a modest effect on the open–closed equilibrium in P450cin compared to that in P450cam. It has been postulated that part of the effector role of Pdx on P450cam is to promote a significant structural change that makes available a proton relay network involving Asp251 required for O2 activation. The structure around the corresponding Asp in P450cin, Asp241, provides a possible structural reason for why P450cin is less dependent on its redox partner for functionally important structural changes.
Co-reporter:Georges Chreifi, Huiying Li, Craig R. McInnes, Colin L. Gibson, Colin J. Suckling, and Thomas L. Poulos
Biochemistry 2014 Volume 53(Issue 25) pp:4216-4223
Publication Date(Web):May 12, 2014
DOI:10.1021/bi5003986
The nitric oxide synthase (NOS) dimer is stabilized by a Zn2+ ion coordinated to four symmetry-related Cys residues exactly along the dimer 2-fold axis. Each of the two essential tetrahydrobiopterin (H4B) molecules in the dimer interacts directly with the heme, and each H4B molecule is ∼15 Å from the Zn2+. We have determined the crystal structures of the bovine endothelial NOS dimer oxygenase domain bound to three different pterin analogues, which reveal an intimate structural communication between the H4B and Zn2+ sites. The binding of one of these compounds, 6-acetyl-2-amino-7,7-dimethyl-7,8-dihydro-4(3H)-pteridinone (1), to the pterin site and Zn2+ binding are mutually exclusive. Compound 1 both directly and indirectly disrupts hydrogen bonding between key residues in the Zn2+ binding motif, resulting in destabilization of the dimer and a complete disruption of the Zn2+ site. Addition of excess Zn2+ stabilizes the Zn2+ site at the expense of weakened binding of 1. The unique structural features of 1 that disrupt the dimer interface are extra methyl groups that extend into the dimer interface and force a slight opening of the dimer, thus resulting in disruption of the Zn2+ site. These results illustrate a very delicate balance of forces and structure at the dimer interface that must be maintained to properly form the Zn2+, pterin, and substrate binding sites.
Co-reporter:Huiying Li, Joumana Jamal, Silvia Delker, Carla Plaza, Haitao Ji, Qing Jing, He Huang, Soosung Kang, Richard B. Silverman, and Thomas L. Poulos
Biochemistry 2014 Volume 53(Issue 32) pp:5272-5279
Publication Date(Web):August 4, 2014
DOI:10.1021/bi500561h
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.
Co-reporter:Qing Jing, Huiying Li, Linda J. Roman, Pavel Martásek, Thomas L. Poulos, and Richard B. Silverman
ACS Medicinal Chemistry Letters 2014 Volume 5(Issue 1) pp:56-60
Publication Date(Web):November 5, 2013
DOI:10.1021/ml400381s
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 headgroup 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.Keywords: enzyme inhibition; neurodegenerative diseases; Nitric oxide; nitric oxide synthase;
Co-reporter:Qing Jing, Huiying Li, Linda J. Roman, Pavel Martásek, Thomas L. Poulos, Richard B. Silverman
Bioorganic & Medicinal Chemistry Letters 2014 Volume 24(Issue 18) pp:4504-4510
Publication Date(Web):15 September 2014
DOI:10.1016/j.bmcl.2014.07.079
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.
Co-reporter:Sarvind Tripathi;Huiying Li
Science 2013 Vol 340(6137) pp:1227-1230
Publication Date(Web):07 Jun 2013
DOI:10.1126/science.1235797

Getting Active

The P450 cytochromes are a family of enzymes that contain a heme cofactor and catalyze the oxidation of organic substrates, including drugs. Protein partners are required to deliver electrons to P450. Insight into this mechanism has come from studies of bacterial P450cam; however, a lack of structures of redox complexes has hindered understanding. Tripathi et al. (p. 1227) describe the high resolution crystal structures of oxidized and reduced P450cam complexed with its redox partner putidaredoxin (Pdx). Pdx favors binding to the more open form of P450cam, which enables establishment of the water-mediated H-bonded network required for proton-coupled electron transfer and O2 activation.

Co-reporter:Thomas L. Poulos and Huiying Li
Accounts of Chemical Research 2013 Volume 46(Issue 2) pp:390
Publication Date(Web):October 2, 2012
DOI:10.1021/ar300175n
Nitric oxide synthase (NOS) converts l-arginine into l-citrulline and releases the important signaling molecule nitric oxide (NO). In the cardiovascular system, NO produced by endothelial NOS (eNOS) relaxes smooth muscle which controls vascular tone and blood pressure. Neuronal NOS (nNOS) produces NO in the brain, where it influences a variety of neural functions such as neural transmitter release. NO can also support the immune system, serving as a cytotoxic agent during infections.Even with all of these important functions, NO is a free radical and, when overproduced, it can cause tissue damage. This mechanism can operate in many neurodegenerative diseases, and as a result the development of drugs targeting nNOS is a desirable therapeutic goal. However, the active sites of all three human isoforms are very similar, and designing inhibitors specific for nNOS is a challenging problem. It is critically important, for example, not to inhibit eNOS owing to its central role in controlling blood pressure.In this Account, we summarize our efforts in collaboration with Rick Silverman at Northwestern University to develop drug candidates that specifically target NOS using crystallography, computational chemistry, and organic synthesis. As a result, we have developed aminopyridine compounds that are 3800-fold more selective for nNOS than eNOS, some of which show excellent neuroprotective effects in animal models.Our group has solved approximately 130 NOS-inhibitor crystal structures which have provided the structural basis for our design efforts. Initial crystal structures of nNOS and eNOS bound to selective dipeptide inhibitors showed that a single amino acid difference (Asp in nNOS and Asn in eNOS) results in much tighter binding to nNOS. The NOS active site is open and rigid, which produces few large structural changes when inhibitors bind. However, we have found that relatively small changes in the active site and inhibitor chirality can account for large differences in isoform-selectivity. For example, we expected that the aminopyridine group on our inhibitors would form a hydrogen bond with a conserved Glu inside the NOS active site. Instead, in one group of inhibitors, the aminopyridine group extends outside of the active site where it interacts with a heme propionate. For this orientation to occur, a conserved Tyr side chain must swing out of the way. This unanticipated observation taught us about the importance of inhibitor chirality and active site dynamics.We also successfully used computational methods to gain insights into the contribution of the state of protonation of the inhibitors to their selectivity. Employing the lessons learned from the aminopyridine inhibitors, the Silverman lab designed and synthesized symmetric double-headed inhibitors with an aminopyridine at each end, taking advantage of their ability to make contacts both inside and outside of the active site.Crystal structures provided yet another unexpected surprise. Two of the double-headed inhibitor molecules bound to each enzyme subunit, and one molecule participated in the generation of a novel Zn2+ site that required some side chains to adopt alternate conformations. Therefore, in addition to achieving our specific goal, the development of nNOS selective compounds, we have learned how subtle differences in dynamics and structure can control protein–ligand interactions and often in unexpected ways.
Co-reporter:He Huang ; Huiying Li ; Pavel Martásek ; Linda J. Roman ; Thomas L. Poulos ;Richard B. Silverman
Journal of Medicinal Chemistry 2013 Volume 56(Issue 7) pp:3024-3032
Publication Date(Web):March 1, 2013
DOI:10.1021/jm4000984
Nitric oxide synthases (NOSs) comprise three closely related isoforms that catalyze the oxidation of l-arginine to l-citrulline and the important second messenger nitric oxide (NO). Pharmacological selective inhibition of neuronal NOS (nNOS) has the potential to be therapeutically beneficial in various neurodegenerative diseases. Here, we present a structure-guided, selective nNOS inhibitor design based on the crystal structure of lead compound 1 in nNOS. The best inhibitor, 7, exhibited low nanomolar inhibitory potency and good isoform selectivities (nNOS over eNOS and iNOS are 472-fold and 239-fold, respectively). Consistent with the good selectivity, 7 binds to nNOS and eNOS with different binding modes. The distinctly different binding modes of 7, driven by the critical residue Asp597 in nNOS, offers compelling insight to explain its isozyme selectivity, which should guide future drug design programs.
Co-reporter:Qing Jing, Huiying Li, Jianguo Fang, Linda J. Roman, Pavel Martásek, Thomas L. Poulos, Richard B. Silverman
Bioorganic & Medicinal Chemistry 2013 Volume 21(Issue 17) pp:5323-5331
Publication Date(Web):1 September 2013
DOI:10.1016/j.bmc.2013.06.014
In certain neurodegenerative diseases damaging levels of nitric oxide (NO) are produced by neuronal nitric oxide synthase (nNOS). It, therefore, is important to develop inhibitors selective for nNOS that do not interfere with other NOS isoforms, especially endothelial NOS (eNOS), which is critical for proper functioning of the cardiovascular system. While we have been successful in developing potent and isoform-selective inhibitors, such as lead compounds 1 and 2, the ease of synthesis and bioavailability have been problematic. Here we describe a new series of compounds including crystal structures of NOS-inhibitor complexes that integrate the advantages of easy synthesis and good biological properties compared to the lead compounds. These results provide the basis for additional structure–activity relationship (SAR) studies to guide further improvement of isozyme selective inhibitors.
Co-reporter:Sarvind Tripathi, Maura J. O'Neill, Angela Wilks, Thomas L. Poulos
Journal of Inorganic Biochemistry 2013 Volume 128() pp:131-136
Publication Date(Web):November 2013
DOI:10.1016/j.jinorgbio.2013.07.030
•1.98Å crystal structure of the P. aeruginosa cytoplasmic heme binding protein.•The structure is very similar to other heme binding proteins from other pathogenic bacteria.•Modeling studies suggest two binding modes for heme.•Multiple heme binding modes explains some of the biochemical/biophysical properties of PhuSIron is an essential element to all living organisms and is an important determinant of bacterial virulence. Bacteria have evolved specialized systems to sequester and transport iron from the environment or host. Pseudomonas aeruginosa, an opportunistic pathogen, uses two outer membrane receptor mediated systems (Phu and Has) to utilize host heme as a source of iron. PhuS is a 39 kDa soluble cytoplasmic heme binding protein which interacts and transports heme from the inner membrane heme transporter to the cytoplasm where it is degraded by heme oxygenase thus releasing iron. PhuS is unique among other cytoplasmic heme transporter proteins owing to the presence of three histidines in the heme binding pocket which can potentially serve as heme ligands. Out of the three histidine residues on the heme binding helix, His 209 is conserved among heme trafficking proteins while His 210 and His 212 are unique to PhuS. Here we report the crystal structure of PhuS at 1.98 Å resolution which shows a unique heme binding pocket and oligomeric structure compared to other known cytoplasmic heme transporter and accounts for some of the unusual biochemical properties of PhuS.PhuS structure showing the predicted location of the heme required for heme transfer to heme oxygenase.
Co-reporter:Kristin Jansen Labby, Huiying Li, Linda J. Roman, Pavel Martásek, Thomas L. Poulos, and Richard B. Silverman
Biochemistry 2013 Volume 52(Issue 18) pp:
Publication Date(Web):April 15, 2013
DOI:10.1021/bi301571v
Nitric oxide synthase (NOS) catalyzes the conversion of l-arginine to l-citrulline through the intermediate Nω-hydroxy-l-arginine (NHA), producing nitric oxide, an important mammalian signaling molecule. Several disease states are associated with improper regulation of nitric oxide production, making NOS a therapeutic target. The first step of the NOS reaction has been well-characterized and is presumed to proceed through a compound I heme species, analogous to the cytochrome P450 mechanism. The second step, however, is enzymatically unprecedented and is thought to occur via a ferric peroxo heme species. To gain insight into the details of this unique second step, we report here the synthesis of NHA analogues bearing guanidinium methyl or ethyl substitutions and their investigation as either inhibitors of or alternate substrates for NOS. Radiolabeling studies reveal that Nω-methoxy-l-arginine, an alternative NOS substrate, produces citrulline, nitric oxide, and methanol. On the basis of these results, we propose a mechanism for the second step of NOS catalysis in which a methylated nitric oxide species is released and is further metabolized by NOS. Crystal structures of our NHA analogues bound to nNOS have been determined, revealing the presence of an active site water molecule only in the presence of singly methylated analogues. Bulkier analogues displace this active site water molecule; a different mechanism is proposed in the absence of the water molecule. Our results provide new insights into the steric and stereochemical tolerance of the NOS active site and substrate capabilities of NOS.
Co-reporter:Yarrow Madrona, Scott A. Hollingsworth, Bushra Khan, and Thomas L. Poulos
Biochemistry 2013 Volume 52(Issue 30) pp:
Publication Date(Web):July 6, 2013
DOI:10.1021/bi4006946
In P450cin, Tyr81, Asp241, Asn242, two water molecules, and the substrate participate in a complex H-bonded network. The role of this H-bonded network in substrate binding and catalysis has been probed by crystallography, spectroscopy, kinetics, isothermal titration calorimetry (ITC), and molecular dynamics. For the Y81F mutant, the substrate binds about 20-fold more weakly and Vmax decreases by about 30% in comparison to WT. The enhanced susceptibility of the heme to H2O2-mediated destruction in Y81F suggests that this mutant favors the open, low-spin conformational state. Asn242 H-bonds directly with the substrate, and replacing this residue with Ala results in water taking the place of the missing Asn side chain. This mutant exhibits a 70% decrease in activity. Crystal structures and molecular dynamics simulations of substrate-bound complexes show that the solvent has more ready access to the active site, especially for the N242A mutant. This accounts for about a 64% uncoupling of electron transfer from substrate hydroxylation. These data indicate the importance of the interconnected water network on substrate binding and on the open/closed conformational equilibrium, which are both critically important for maintaining high-coupling efficiency.
Co-reporter:Huiying Li, Fengtian Xue, James M. Kraus II, Haitao Ji, Kristin Jansen Labby, Jan Mataka, Silvia L. Delker, Pavel Martásek, Linda J. Roman, Thomas L. Poulos, Richard B. Silverman
Bioorganic & Medicinal Chemistry 2013 Volume 21(Issue 5) pp:1333-1343
Publication Date(Web):1 March 2013
DOI:10.1016/j.bmc.2012.12.019
Inhibitors of neuronal nitric oxide synthase have been proposed as therapeutics for the treatment of different types of neurological disorders. On the basis of a cis-3,4-pyrrolidine scaffold, a series of trans-cyclopropyl- and methyl-containing nNOS inhibitors have been synthesized. The insertion of a rigid electron-withdrawing cyclopropyl ring decreases the basicity of the adjacent amino group, which resulted in decreased inhibitory activity of these inhibitors compared to the parent compound. Nonetheless, three of them exhibited double-digit nanomolar inhibition with high nNOS selectivity on the basis of in vitro enzyme assays. Crystal structures of nNOS and eNOS with these inhibitors bound provide a basis for detailed structure–activity relationship (SAR) studies. The conclusions from these studies will be used as a guide in the future development of selective NOS inhibitors.
Co-reporter:Dipanwita Batabyal and Thomas L. Poulos
Biochemistry 2013 Volume 52(Issue 49) pp:
Publication Date(Web):November 21, 2013
DOI:10.1021/bi401330c
Although CYP101D1 and P450cam catalyze the same reaction at similar rates and share strikingly similar active site architectures, there are significant functional differences. CYP101D1 thus provides an opportunity to probe what structural and functional features must be shared and what features can differ but maintain the high catalytic efficiency. Crystal structures of the cyanide complex of wild-type CYP101D1 and it active site mutants, D259N and T260A, have been determined. The conformational changes in CYP101D1 upon cyanide binding are very similar to those of P450cam, indicating a similar mechanism for proton delivery during oxygen activation using solvent-assisted proton transfer. The D259N–CN– complex shows a perturbed solvent structure compared to that of the wild type, which is similar to what was observed in the oxy complex of the corresonding D251N mutant in P450cam. As in P450cam, the T260A mutant is highly uncoupled while the D259N mutant gives barely detectable activity. Despite these similarities, CYP101D1 is able to use the P450cam redox partners while P450cam cannot use the CYP101D1 redox partners. Thus, the strict requirement of P450cam for its own redox partner is relaxed in CYP101D1. Differences in the local environment of the essential Asp (Asp259 in CYP101D1) provide a strucutral basis for understanding these functional differences.
Co-reporter:Dipanwita Batabyal, Huiying Li, and Thomas L. Poulos
Biochemistry 2013 Volume 52(Issue 32) pp:
Publication Date(Web):July 18, 2013
DOI:10.1021/bi400676d
A close orthologue to cytochrome P450cam (CYP101A1) that catalyzes the same hydroxylation of camphor to 5-exo-hydroxycamphor is CYP101D1. There are potentially important differences in and around the active site that could contribute to subtle functional differences. Adjacent to the heme iron ligand, Cys357, is Leu358 in P450cam, whereas this residue is Ala in CYP101D1. Leu358 plays a role in binding of the P450cam redox partner, putidaredoxin (Pdx). On the opposite side of the heme, about 15–20 Å away, Asp251 in P450cam plays a critical role in a proton relay network required for O2 activation but forms strong ion pairs with Arg186 and Lys178. In CYP101D1 Gly replaces Lys178. Thus, the local electrostatic environment and ion pairing are substantially different in CYP101D1. These sites have been systematically mutated in P450cam to the corresponding residues in CYP101D1 and the mutants analyzed by crystallography, kinetics, and UV–vis spectroscopy. Individually, the mutants have little effect on activity or structure, but in combination there is a major drop in enzyme activity. This loss in activity is due to the mutants being locked in the low-spin state, which prevents electron transfer from the P450cam redox partner, Pdx. These studies illustrate the strong synergistic effects on well-separated parts of the structure in controlling the equilibrium between the open (low-spin) and closed (high-spin) conformational states.
Co-reporter:Qing Jing, Huiying Li, Georges Chreifi, Linda J. Roman, Pavel Martásek, Thomas L. Poulos, Richard B. Silverman
Bioorganic & Medicinal Chemistry Letters 2013 Volume 23(Issue 20) pp:5674-5679
Publication Date(Web):15 October 2013
DOI:10.1016/j.bmcl.2013.08.034
To develop potent and selective nNOS inhibitors, new double-headed molecules with chiral linkers that derive from natural amino acids or their derivatives have been designed. The new structures contain two ether bonds, which greatly simplifies the synthesis and accelerates structure optimization. Inhibitor (R)-6b exhibits a potency of 32 nM against nNOS and is 475 and 244 more selective for nNOS over eNOS and iNOS, respectively. Crystal structures show that the additional binding between the aminomethyl moiety of 6b and the two heme propionates in nNOS, but not eNOS, is the structural basis for its high selectivity. This work demonstrates the importance of stereochemistry in this class of molecules, which significantly influences the potency and selectivity of the inhibitors. The structure–activity information gathered here provides a guide for future structure optimization.
Co-reporter:Qing Jing;Soosung Kang;Jeffrey K. Holden;Huiying Li;Richard B. Silverman;Jerry Richo
PNAS 2013 Volume 110 (Issue 45 ) pp:18127-18131
Publication Date(Web):2013-11-05
DOI:10.1073/pnas.1314080110
Nitric oxide (NO) produced by bacterial NOS functions as a cytoprotective agent against oxidative stress in Staphylococcus aureus, Bacillus anthracis, and Bacillus subtilis. The screening of several NOS-selective inhibitors uncovered two inhibitors with potential antimicrobial properties. These two compounds impede the growth of B. subtilis under oxidative stress, and crystal structures show that each compound exhibits a unique binding mode. Both compounds serve as excellent leads for the future development of antimicrobials against bacterial NOS-containing bacteria.
Co-reporter:He Huang, Haitao Ji, Huiying Li, Qing Jing, Kristin Jansen Labby, Pavel Martásek, Linda J. Roman, Thomas L. Poulos, and Richard B. Silverman
Journal of the American Chemical Society 2012 Volume 134(Issue 28) pp:11559-11572
Publication Date(Web):June 25, 2012
DOI:10.1021/ja302269r
The reduction of pathophysiologic levels of nitric oxide through inhibition of neuronal nitric oxide synthase (nNOS) has the potential to be therapeutically beneficial in various neurodegenerative diseases. We have developed a series of pyrrolidine-based nNOS inhibitors that exhibit excellent potencies and isoform selectivities (J. Am. Chem. Soc.2010, 132, 5437). However, there are still important challenges, such as how to decrease the multiple positive charges derived from basic amino groups, which contribute to poor bioavailability, without losing potency and/or selectivity. Here we present an interdisciplinary study combining molecular docking, crystallography, molecular dynamics simulations, synthesis, and enzymology to explore potential pharmacophoric features of nNOS inhibitors and to design potent and selective monocationic nNOS inhibitors. The simulation results indicate that different hydrogen bond patterns, electrostatic interactions, hydrophobic interactions, and a water molecule bridge are key factors for stabilizing ligands and controlling ligand orientation. We find that a heteroatom in the aromatic head or linker chain of the ligand provides additional stability and blocks the substrate binding pocket. Finally, the computational insights are experimentally validated with double-headed pyridine analogues. The compounds reported here are among the most potent and selective monocationic pyrrolidine-based nNOS inhibitors reported to date, and 10 shows improved membrane permeability.
Co-reporter:Yarrow Madrona, Sarvind Tripathi, Huiying Li, and Thomas L. Poulos
Biochemistry 2012 Volume 51(Issue 33) pp:
Publication Date(Web):July 9, 2012
DOI:10.1021/bi300666u
The crystal structure of the P450cin substrate-bound nitric oxide complex and the substrate-free form have been determined revealing a substrate-free structure that adopts an open conformation relative to the substrate-bound structure. The region of the I helix that forms part of the O2 binding pocket shifts from an α helix in the substrate-free form to a π helix in the substrate-bound form. Unique to P450cin is an active site residue, Asn242, in the I helix that H-bonds with the substrate. In most other P450s this residue is a Thr and plays an important role in O2 activation by participating in an H-bonding network required for O2 activation. The π/α I helix transition results in the carbonyl O atom of Gly238 moving in to form an H-bond with the water/hydroxide ligand in the substrate-free form. The corresponding residue, Gly248, in the substrate-free P450cam structure experiences a similar motion. Most significantly, in the oxy-P450cam complex Gly248 adopts a position midway between the substrate-free and -bound states. A comparison between these P450cam and the new P450cin structures provides insights into differences in how the two P450s activate O2. The structure of P450cin complexed with nitric oxide, a close mimic of the O2 complex, shows that Gly238 is likely to form tighter interactions with ligands than the corresponding Gly248 in P450cam. Having a close interaction between an H-bond acceptor, the Gly238 carbonyl O atom, and the distal oxygen atom of O2 will promote protonation and hence further reduction of the oxy complex to the hydroperoxy intermediate resulting in heterolytic cleavage of the peroxide O–O bond and formation of the active ferryl intermediate required for substrate hydroxylation.
Co-reporter:Victoria S. Jasion and Thomas L. Poulos
Biochemistry 2012 Volume 51(Issue 12) pp:
Publication Date(Web):February 29, 2012
DOI:10.1021/bi300169x
Leishmania major peroxidase (LmP) exhibits both ascorbate and cytochrome c peroxidase activities. Our previous results illustrated that LmP has a much higher activity against horse heart cytochrome c than ascorbate, suggesting that cytochrome c may be the biologically important substrate. To elucidate the biological function of LmP, we have recombinantly expressed, purified, and determined the 2.08 Å crystal structure of L. major cytochrome c (LmCytc). Like other types of cytochrome c, LmCytc has an electropositive surface surrounding the exposed heme edge that serves as the site of docking with redox partners. Kinetic assays performed with LmCytc and LmP show that LmCytc is a much better substrate for LmP than horse heart cytochrome c. Furthermore, unlike the well-studied yeast system, the reaction follows classic Michaelis–Menten kinetics and is sensitive to an increasing ionic strength. Using the yeast cocrystal as a control, protein–protein docking was performed using Rosetta to develop a model for the binding of LmP and LmCytc. These results suggest that the biological function of LmP is to act as a cytochrome c peroxidase.
Co-reporter:Kristin Jansen Labby, Fengtian Xue, James M. Kraus, Haitao Ji, Jan Mataka, Huiying Li, Pavel Martásek, Linda J. Roman, Thomas L. Poulos, Richard B. Silverman
Bioorganic & Medicinal Chemistry 2012 Volume 20(Issue 7) pp:2435-2443
Publication Date(Web):1 April 2012
DOI:10.1016/j.bmc.2012.01.037
Selective neuronal nitric oxide synthase (nNOS) inhibitors have therapeutic applications in the treatment of numerous neurodegenerative diseases. Here we report the synthesis and evaluation of a series of inhibitors designed to have increased cell membrane permeability via intramolecular hydrogen bonding. Their potencies were examined in both purified enzyme and cell-based assays; a comparison of these results demonstrates that two of the new inhibitors display significantly increased membrane permeability over previous analogs. NMR spectroscopy provides evidence of intramolecular hydrogen bonding under physiological conditions in two of the inhibitors. Crystal structures of the inhibitors in the nNOS active site confirm the predicted non-intramolecular hydrogen bonded binding mode. Intramolecular hydrogen bonding may be an effective approach for increasing cell membrane permeability without affecting target protein binding.
Co-reporter:Victoria S. Jasion;Tzanko Doukov;Huiying Li;Stephanie H. Pineda
PNAS 2012 Volume 109 (Issue 45 ) pp:18390-18394
Publication Date(Web):2012-11-06
DOI:10.1073/pnas.1213295109
The causative agent of leishmaniasis is the protozoan parasite Leishmania major. Part of the host protective mechanism is the production of reactive oxygen species including hydrogen peroxide. In response, L. major produces a peroxidase, L. major peroxidase (LmP), that helps to protect the parasite from oxidative stress. LmP is a heme peroxidase that catalyzes the peroxidation of mitochondrial cytochrome c. We have determined the crystal structure of LmP in a complex with its substrate, L. major cytochrome c (LmCytc) to 1.84 Å, and compared the structure to its close homolog, the yeast cytochrome c peroxidase–cytochrome c complex. The binding interface between LmP and LmCytc has one strong and one weak ionic interaction that the yeast system lacks. The differences between the steady-state kinetics correlate well with the Lm redox pair being more dependent on ionic interactions, whereas the yeast redox pair depends more on nonpolar interactions. Mutagenesis studies confirm that the ion pairs at the intermolecular interface are important to both kcat and KM. Despite these differences, the electron transfer path, with respect to the distance between hemes, along the polypeptide chain is exactly the same in both redox systems. A potentially important difference, however, is the side chains involved. LmP has more polar groups (Asp and His) along the pathway compared with the nonpolar groups (Leu and Ala) in the yeast system, and as a result, the electrostatic environment along the presumed electron transfer path is substantially different.
Co-reporter:Tzanko Doukov ; Huiying Li ; Ajay Sharma ; Jeffrey D. Martell ; S. Michael Soltis ; Richard B. Silverman
Journal of the American Chemical Society 2011 Volume 133(Issue 21) pp:8326-8334
Publication Date(Web):May 2, 2011
DOI:10.1021/ja201466v
A series of l-arginine analogue nitric oxide synthase inhibitors with a thioether tail have been shown to form an Fe–S thioether interaction as evidenced by continuous electron density between the Fe and S atoms. Even so, the Fe–S thioether interaction was found to be far less important for inhibitor binding than the hydrophobic interactions between the alkyl group in the thioether tail and surrounding protein (Martell et al. J. Am. Chem. Soc. 2010, 132, 798). However, among the few thioether inhibitors that showed Fe–S thioether interaction in crystal structures, variations in spin state (high-spin or low-spin) were observed dependent upon the heme iron oxidation state and temperature. Since modern synchrotron X-ray data collection is typically carried out at cryogenic temperatures, we reasoned that some of the discrepancies between cryo-crystal structures and room-temperature UV–visible spectroscopy could be the result of temperature-dependent spin-state changes. We, therefore, have characterized some of these neuronal nitric oxide synthase (nNOS)–thioether inhibitor complexes in both crystal and solution using EPR and UV–visible absorption spectrometry as a function of temperature and the heme iron redox state. We found that some thioether inhibitors switch from high to low spin at lower temperatures similar to the “spin crossover” phenomenon observed in many transition metal complexes.
Co-reporter:Fengtian Xue ; Jianguo Fang ; Silvia L. Delker ; Huiying Li ; Pavel Martásek ¥; Linda J. Roman ; Thomas L. Poulos ▲;Richard B. Silverman
Journal of Medicinal Chemistry 2011 Volume 54(Issue 7) pp:2039-2048
Publication Date(Web):March 16, 2011
DOI:10.1021/jm101071n
We report novel neuronal nitric oxide synthase (nNOS) inhibitors based on a symmetric double-headed aminopyridine scaffold. The inhibitors were designed from crystal structures of leads 1 and 2 (Delker, S. L.; Ji, H.; Li, H.; Jamal, J.; Fang, J.; Xue, F.; Silverman, R. B.; Poulos, T. L.Unexpected binding modes of nitric oxide synthase inhibitors effective in the prevention of cerebral palsy. J. Am. Chem. Soc. 2010, 132, 5437−5442) and synthesized using a highly efficient route. The best inhibitor, 3j, showed low nanomolar inhibitory potency and modest isoform selectivity. It also exhibited enhanced membrane permeability. Inhibitor 3j binds to both the substrate site and the pterin site in nNOS but only to the substrate site in eNOS. These compounds provide a basis for further development of novel, potent, isoform selective, and bioavailable inhibitors for nNOS.
Co-reporter:Fengtian Xue ; James M. Kraus ; Kristin Jansen Labby ; Haitao Ji ; Jan Mataka ; Guoyao Xia ; Huiying Li ; Silvia L. Delker ; Linda J. Roman ; Pavel Martásek ; Thomas L. Poulos ;Richard B. Silverman
Journal of Medicinal Chemistry 2011 Volume 54(Issue 18) pp:6399-6403
Publication Date(Web):August 2, 2011
DOI:10.1021/jm200411j
We report an efficient synthetic route to chiral pyrrolidine inhibitors of neuronal nitric oxide synthase (nNOS) and crystal structures of the inhibitors bound to nNOS and to endothelial NOS. The new route enables versatile structure–activity relationship studies on the pyrrolidine-based scaffold, which can be beneficial for further development of nNOS inhibitors. The X-ray crystal structures of five new fluorine-containing inhibitors bound to nNOS provide insights into the effect of the fluorine atoms on binding.
Co-reporter:Fengtian Xue ; Huiying Li ; Silvia L. Delker ; Jianguo Fang ; Pavel Martásek ; Linda J. Roman ; Thomas L. Poulos ;Richard B. Silverman
Journal of the American Chemical Society 2010 Volume 132(Issue 40) pp:14229-14238
Publication Date(Web):September 15, 2010
DOI:10.1021/ja106175q
In our efforts to discover neuronal isoform selective nitric oxide synthase (NOS) inhibitors, we have developed a series of compounds containing a pyrrolidine ring with two stereogenic centers. The enantiomerically pure compounds, (S,S) versus (R,R), exhibited two different binding orientations, with (R,R) inhibitors showing much better potency and selectivity. To improve the bioavailability of these inhibitors, we have introduced a CF2 moiety geminal to an amino group in the long tail of one of these inhibitors, which reduced its basicity, resulting in compounds with monocationic character under physiological pH conditions. Biological evaluations have led to a nNOS inhibitor with a Ki of 36 nM and high selectivity for nNOS over eNOS (3800-fold) and iNOS (1400-fold). MM-PBSA calculations indicated that the low pKa NH is, at least, partially protonated when bound to the active site. A comparison of rat oral bioavailability of the difluorinated compound to the parent molecule shows 22% for the difluorinated compound versus essentially no oral bioavailability for the parent compound. This indicates that the goal of this research to make compounds with only one protonated nitrogen atom at physiological pH to allow for membrane permeability, but which can become protonated when bound to NOS, has been accomplished.
Co-reporter:Silvia L Delker ; Haitao Ji ; Huiying Li ; Joumana Jamal ; Jianguo Fang ; Fengtian Xue ; Richard B. Silverman
Journal of the American Chemical Society 2010 Volume 132(Issue 15) pp:5437-5442
Publication Date(Web):March 25, 2010
DOI:10.1021/ja910228a
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 (∼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.
Co-reporter:Haitao Ji ; Silvia L. Delker ; Huiying Li ; Pavel Martásek ; Linda J. Roman ; Thomas L. Poulos ;Richard B. Silverman
Journal of Medicinal Chemistry 2010 Volume 53(Issue 21) pp:7804-7824
Publication Date(Web):October 19, 2010
DOI:10.1021/jm100947x
Neuronal nitric oxide synthase (nNOS) represents an important therapeutic target for the prevention of brain injury and the treatment of various neurodegenerative disorders. A series of trans-substituted amino pyrrolidinomethyl 2-aminopyridine derivatives (8−34) was designed and synthesized. A structure−activity relationship analysis led to the discovery of low nanomolar nNOS inhibitors ((±)-32 and (±)-34) with more than 1000-fold selectivity for nNOS over eNOS. Four enantiomerically pure isomers of 3′-[2′′-(3′′′-fluorophenethylamino)ethoxy]pyrrolidin-4′-yl}methyl}-4-methylpyridin-2-amine (4) also were synthesized. It was found that (3′R,4′R)-4 can induce enzyme elasticity to generate a new “hot spot” for ligand binding. The inhibitor adopts a unique binding mode, the same as that observed for (3′R,4′R)-3′-[2′′-(3′′′-fluorophenethylamino)ethylamino]pyrrolidin-4′-yl}methyl}-4-methylpyridin-2-amine ((3′R,4′R)-3) ( J. Am. Chem. Soc. 2010, 132 (15), 5437−5442). On the basis of structure−activity relationships of 8−34 and different binding conformations of the cis and trans isomers of 3 and 4, critical structural requirements of the NOS active site for ligand binding are revealed.
Co-reporter:Silvia L. Delker, Fengtian Xue, Huiying Li, Joumana Jamal, Richard B. Silverman, and Thomas L. Poulos
Biochemistry 2010 Volume 49(Issue 51) pp:
Publication Date(Web):October 30, 2010
DOI:10.1021/bi1013479
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 (H4B) cofactor and creation of a new Zn2+ site. These changes are due to binding of a second inhibitor molecule that results in the displacement of H4B and the placement of the inhibitor pyridine group in position to serve as a Zn2+ ligand together with Asp, His, and a chloride ion. Binding of the second inhibitor molecule and generation of the Zn2+ site do not occur in eNOS. Structural requirements for creation of the new Zn2+ site in nNOS were analyzed in detail. These observations open the way for the potential design of novel inhibitors selective for nNOS.
Co-reporter:Yergalem T. Meharenna and Thomas L. Poulos
Biochemistry 2010 Volume 49(Issue 31) pp:
Publication Date(Web):July 1, 2010
DOI:10.1021/bi100929x
High-temperature molecular dynamics (MD) has been used to assess if MD can be employed as a useful tool for probing the structural basis for enhanced stability in thermal stable cytochromes P450. CYP119, the most thermal stable P450 known, unfolds more slowly during 500 K MD simulations than P450s that melt at lower temperatures, P450cam and P450cin. A comparison of the 500 K MD trajectories shows that the Cys ligand loop, a critically important structural feature just under the heme, in both P450cin and P450cam completely unfolds while this region is quite stable in CYP119. In CYP119, this region is stabilized by tight nonpolar interactions involving Tyr26 and Leu308. The corresponding residues in P450cam are Gly and Thr, respectively. The in silico generated Y26A/L308A CYP119 double mutant is substantially less stable than wild-type CYP119, and the Cys ligand loop unfolds in a manner similar to that of P450cam. The MD thus has identified a potential “hot spot” important for stability. As an experimental test of the MD results, the Y26A/L308A double mutant was prepared, and thermal melting curves show that the double mutant exhibits a melting temperature (Tm) 16 °C lower than that of wild-type CYP119. Control mutations that were predicted by MD not to destabilize the protein were also generated, and the experimental melting temperature was not significantly different from that of the wild-type enzyme. Therefore, high-temperature MD is a useful tool in predicting the structural underpinnings of thermal stability in P450s.
Co-reporter:Fengtian Xue, Jinwen Huang, Haitao Ji, Jianguo Fang, Huiying Li, Pavel Martásek, Linda J. Roman, Thomas L. Poulos, Richard B. Silverman
Bioorganic & Medicinal Chemistry 2010 Volume 18(Issue 17) pp:6526-6537
Publication Date(Web):1 September 2010
DOI:10.1016/j.bmc.2010.06.074
Selective inhibitors of neuronal nitric oxide synthase (nNOS) have the potential to develop into new neurodegenerative therapeutics. Recently, we described the discovery of novel nNOS inhibitors (1a and 1b) based on a cis-pyrrolidine pharmacophore. These compounds and related ones were found to have poor blood–brain barrier permeability, presumably because of the basic nitrogens in the molecule. Here, a series of monocationic compounds was designed on the basis of docking experiments using the crystal structures of 1a,b bound to nNOS. These compounds were synthesized and evaluated for their ability to inhibit neuronal nitric oxide synthase. Despite the excellent overlap of these compounds with 1a,b bound to nNOS, they exhibited low potency. This is because they bound in the nNOS active site in the normal orientation rather than the expected flipped orientation used in the computer modeling. The biphenyl or phenoxyphenyl tail is disordered and does not form good protein–ligand interactions. These studies demonstrate the importance of the size and rigidity of the side chain tail and the second basic amino group for nNOS binding efficiency and the importance of the hydrophobic tail for conformational orientation in the active site of nNOS.
Co-reporter:Yergalem T. Meharenna, Tzanko Doukov, Huiying Li, S. Michael Soltis and Thomas L. Poulos
Biochemistry 2010 Volume 49(Issue 14) pp:
Publication Date(Web):March 15, 2010
DOI:10.1021/bi100238r
The ferryl [Fe(IV)O] intermediate is important in many heme enzymes, and thus, the precise nature of the Fe(IV)−O bond is critical in understanding enzymatic mechanisms. The 1.40 Å crystal structure of cytochrome c peroxidase Compound I has been determined as a function of X-ray dose while the visible spectrum was being monitored. The Fe−O bond increases in length from 1.73 Å in the low-X-ray dose structure to 1.90 Å in the high-dose structure. The low-dose structure correlates well with an Fe(IV)═O bond, while we postulate that the high-dose structure is the cryo-trapped Fe(III)−OH species previously thought to be an Fe(IV)−OH species.
Co-reporter:Fengtian Xue, Huiying Li, Jianguo Fang, Linda J. Roman, Pavel Martásek, Thomas L. Poulos, Richard B. Silverman
Bioorganic & Medicinal Chemistry Letters 2010 Volume 20(Issue 21) pp:6258-6261
Publication Date(Web):1 November 2010
DOI:10.1016/j.bmcl.2010.08.096
Selective inhibition of the neuronal isoform of nitric oxide synthase (nNOS) over endothelial nitric oxide synthase (eNOS) and inducible nitric oxide synthase (iNOS) has become a promising strategy for the discovery of new therapeutic agents for neurodegenerative diseases. However, because of the high sequence homology of different isozymes in the substrate binding pocket, developing inhibitors with both potency and excellent isoform selectivity remains a challenging problem. Herein, we report the evaluation of a recently discovered peripheral hydrophobic pocket (Tyr706, Leu337, and Met336) that opens up upon inhibitor binding and its potential in designing potent and selective nNOS inhibitors using three compounds, 2a, 2b, and 3. Crystal structure results show that inhibitors 2a and 3 adopted the same binding mode as lead compound 1. We also found that hydrophobic interactions between the 4-methyl group of the aminopyridine ring of these compounds with the side chain of Met336, as well as the π–π stacking interaction between the pyridinyl motif and the side chain of Tyr706 are important for the high potency and selectivity of these nNOS inhibitors.
Co-reporter:Jeffrey D. Martell ; Huiying Li ; Tzanko Doukov ; Pavel Martásek ; Linda J. Roman ; Michael Soltis ; Thomas L. Poulos ;Richard B. Silverman
Journal of the American Chemical Society 2009 Volume 132(Issue 2) pp:798-806
Publication Date(Web):December 16, 2009
DOI:10.1021/ja908544f
The heme−thioether ligand interaction often occurs between heme iron and native methionine ligands, but thioether-based heme-coordinating (type II) inhibitors are uncommon due to the difficulty in stabilizing the Fe−S bond. Here, a thioether-based inhibitor (3) of neuronal nitric oxide synthase (nNOS) was designed, and its binding was characterized by spectrophotometry and crystallography. A crystal structure of inhibitor 3 coordinated to heme iron was obtained, representing, to our knowledge, the first crystal structure of a thioether inhibitor complexed to any heme enzyme. A series of related potential inhibitors (4−8) also were evaluated. Compounds 4−8 were all found to be type I (non-heme-coordinating) inhibitors of ferric nNOS, but 4 and 6−8 were found to switch to type II upon heme reduction to the ferrous state, reflecting the higher affinity of thioethers for ferrous heme than for ferric heme. Contrary to what has been widely thought, thioether−heme ligation was found not to increase inhibitor potency, illustrating the intrinsic weakness of the thioether−ferric heme linkage. Subtle changes in the alkyl groups attached to the thioether sulfur caused drastic changes in the binding conformation, indicating that hydrophobic contacts play a crucial role in stabilizing the thioether−heme coordination.
Co-reporter:Jotaro Igarashi, Huiying Li, Joumana Jamal, Haitao Ji, Jianguo Fang, Graham R. Lawton, Richard B. Silverman and Thomas L. Poulos
Journal of Medicinal Chemistry 2009 Volume 52(Issue 7) pp:2060-2066
Publication Date(Web):March 18, 2009
DOI:10.1021/jm900007a
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 α-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.
Co-reporter:Tzanko Doukov, Huiying Li, Michael Soltis and Thomas L. Poulos
Biochemistry 2009 Volume 48(Issue 43) pp:
Publication Date(Web):September 30, 2009
DOI:10.1021/bi9009743
The X-ray structures of neuronal nitric oxide synthase (nNOS) with Nω-hydroxy-l-arginine (l-NHA) and CO (or NO) bound have been determined at 1.91−2.2 Å resolution. Microspectrophotometric techniques confirmed reduced redox state and the status of diatomic ligand complexes during X-ray diffraction data collection. The structure of nNOS-NHA-NO, a close mimic to the dioxygen complex, provides a picture of the potential interactions between the heme-bound diatomic ligand, substrate l-NHA, and the surrounding protein and solvent structure environment. The OH group of l-NHA in the X-ray structures deviates from the plane of the guanidinium moiety substantially, indicating that the OH-bearing, protonated guanidine Nω nitrogen of l-NHA has substantial sp3 hybridization character. This nitrogen geometry, different from that of the guanidinium Nω nitrogen of l-arginine, allows a hydrogen bond to be donated to the proximal oxygen of the heme-bound dioxygen complex, thus preventing cleavage of the O−O bond. Instead, it favors the stabilization of the ferric-hydroperoxy intermediate, Fe3+-OOH−, which serves as the active oxidant in the conversion of l-NHA to NO and citrulline in the second reaction of the NOS.
Co-reporter:Yergalem T. Meharenna, Patricia Oertel, B. Bhaskar and Thomas L. Poulos
Biochemistry 2008 Volume 47(Issue 39) pp:
Publication Date(Web):September 5, 2008
DOI:10.1021/bi8007565
Cytochrome c peroxidase (CCP) and ascorbate peroxidase (APX) have very similar structures, and yet neither CCP nor APX exhibits each otherʼs activities with respect to reducing substrates. APX has a unique substrate binding site near the heme propionates where ascorbate H-bonds with a surface Arg and one heme propionate (Sharp et al. (2003) Nat. Struct. Biol. 10, 303−307). The corresponding region in CCP has a much longer surface loop, and the critical Arg residue that is required for ascorbate binding in APX is Asn in CCP. In order to convert CCP into an APX, the ascorbate-binding loop and critical arginine were engineered into CCP to give the CCP2APX mutant. The mutant crystal structure shows that the engineered site is nearly identical to that found in APX. While wild-type CCP shows no APX activity, CCP2APX catalyzes the peroxidation of ascorbate at a rate of ≈12 min−1, indicating that the engineered ascorbate-binding loop can bind ascorbate.
Co-reporter:Huiying Li, Thomas L. Poulos
Journal of Inorganic Biochemistry 2005 Volume 99(Issue 1) pp:293-305
Publication Date(Web):January 2005
DOI:10.1016/j.jinorgbio.2004.10.016
Nitric oxide synthase (NOS) catalyzes the oxidation of one l-arginine guanidinium N atom to nitric oxide (NO). NOS consists of a heme domain linked to a flavin mononucleotide (FMN)/flavin adenine dinucleotide (FAD) reductase that shuttles electrons from nicotinamide adenine dinucleotide phosphate (NADPH) to the heme. This review summarizes various aspects of NOS structure and function derived from crystal structures coupled with a wealth of biochemical and biophysical data. This includes the binding of diatomic ligands, especially the product, NO, whose binding to the heme iron blocks enzyme activity. An unusual feature of NOS catalysis is the strict requirement for the essential cofactor, tetrahydrobiopterin (H4B). It now is generally agreed that H4B serves as an electron donor to the heme–oxy complex. The reason NOS may have recruited H4B as an electron transfer cofactor is to provide rapid coupled proton/electron transfer required for O2 activation. NOS is a highly regulated enzyme which is controlled by calmodulin (CaM) at the level of electron transfer within the FMN/FAD reductase and between the reductase and heme domains. Recent crystal structures provide a basis for developing models on the structural underpinnings of NOS regulation. In addition to the complex and fascinating functional and regulatory features of NOS, NOS is an important therapeutic target. Crystal structures have revealed the structural basis of isoform-selective inhibition by a group of dipeptide inhibitors which opens the way for structure-based inhibitor design.
Co-reporter:Latesh Lad, Paul R. Ortiz de Montellano, Thomas L. Poulos
Journal of Inorganic Biochemistry 2004 Volume 98(Issue 11) pp:1686-1695
Publication Date(Web):November 2004
DOI:10.1016/j.jinorgbio.2004.07.004
Heme oxygenase oxidatively degrades heme to biliverdin resulting in the release of iron and CO through a process in which the heme participates both as a cofactor and substrate. One of the least understood steps in the heme degradation pathway is the conversion of verdoheme to biliverdin. In order to obtain a better understanding of this step we report the crystal structures of ferrous–verdoheme and, as a mimic for the oxy–verdoheme complex, ferrous–NO verdoheme in a complex with human HO-1 at 2.20 and 2.10 Å, respectively. In both structures the verdoheme occupies the same binding location as heme in heme–HO-1, but rather than being ruffled verdoheme in both sets of structures is flat. Both structures are similar to their heme counterparts except for the distal helix and heme pocket solvent structure. In the ferrous–verdoheme structure the distal helix moves closer to the verdoheme, thus tightening the active site. NO binds to verdoheme in a similar bent conformation to that found in heme–HO-1. The bend angle in the verodoheme–NO structure places the terminal NO oxygen 1 Å closer to the α-meso oxygen of verdoheme compared to the α-meso carbon on the heme–NO structure. A network of water molecules, which provide the required protons to activate the iron–oxy complex of heme–HO-1, is absent in both ferrous–verdoheme and the verdoheme–NO structure.
Co-reporter:B. Bhaskar;Huiying Li;Maolin Guo;Tiffany P. Barrows
PNAS 2004 Volume 101 (Issue 16 ) pp:5940-5945
Publication Date(Web):2004-04-20
DOI:10.1073/pnas.0306708101
A specific covalently cross-linked complex between redox partners yeast cytochrome c peroxidase (CCP) and cytochrome c (cyt. c) has been made by engineering cysteines into CCP and cyt. c that form an intermolecular disulfide bond in high yield. The crystal structure of the cross-linked complex has been solved to 1.88-Å resolution and closely resembles the structure of the noncovalent complex [Pellitier, H. & Kraut, J. (1992) Science 258, 1748–1755]. The higher resolution of the covalent complex has enabled the location of ordered water molecules at the peroxidase–cytochrome c interface that serve to bridge between the two proteins by hydrogen bonding. As in the noncovalent complex, direct electrostatic interactions between protein groups appear not to be critical in complex formation. UV–visible spectroscopic and stopped-flow studies indicate that CCP in the covalent complex reacts normally with H2O2 to give compound I. Stopped-flow kinetic studies also show that intramolecular electron transfer between the cross-linked ferrocytochrome c and the Trp-191 cation radical site in CCP compound I occurs fast and is nearly complete within the dead time (≈2 ms) of the instrument. These results indicate that the structure of the covalent complex closely mimics the physiological electron transfer complex. In addition, single-turnover and steady-state experiments reveal that CCP compound I in the covalent complex oxidizes exogenously added ferrocytochrome c at a slow rate (t 1/2 ≈ 2 min), indicating that CCP does not have a second independent site for physiologically relevant electron transfer.
Co-reporter:
Nature Structural and Molecular Biology 2004 11(1) pp:54-59
Publication Date(Web):29 December 2003
DOI:10.1038/nsmb704
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 -nitroarginine-containing dipeptide inhibitors.
Co-reporter:Thomas L. Poulos;
Proceedings of the National Academy of Sciences 2003 100(23) pp:13121-13122
Publication Date(Web):November 3, 2003
DOI:10.1073/pnas.2336095100
Co-reporter:Chad E. Immoos, B. Bhaskar, Michael S. Cohen, Tiffany P. Barrows, Patrick J. Farmer, T.L. Poulos
Journal of Inorganic Biochemistry 2002 Volume 91(Issue 4) pp:635-643
Publication Date(Web):20 September 2002
DOI:10.1016/S0162-0134(02)00447-6
The effect of heme ring oxygenation on enzyme structure and function has been examined in a reconstituted cytochrome c peroxidase. Oxochlorin derivatives were formed by OsO4 treatment of mesoporphyrin followed by acid-catalyzed pinacol rearrangement. The northern oxochlorin isomers were isolated by chromatography, and the regio-isomers assignments determined by 2D COSY and NOE 1H NMR. The major isomer, 4-mesoporphyrinone (Mp), was metallated with FeCl2 and reconstituted into cytochrome c peroxidase (CcP) forming a hybrid green protein, MpCcP. The heme-altered enzyme has 99% wild-type peroxidase activity with cytochrome c. EPR spectroscopy of MpCcP intermediate compound I verifies the formation of the Trp191 radical similar to wild-type CcP in the reaction cycle. Peroxidase activity with small molecules is varied: guaiacol turnover increases approximately five-fold while that with ferrocyanide is ∼85% of native. The electron-withdrawing oxo-substitutents on the cofactor cause a ∼60-mV increase in FeIII/FeII reduction potential. The present investigation represents the first structural characterization of an oxochlorin protein with X-ray intensity data collected to 1.70 Å. Although a mixture of R- and S-mesopone isomers of the FeMP cofactor was used during heme incorporation into the apo-protein, only the S-isomer is found in the crystallized protein.
Co-reporter:Huiying Li, C.S. Raman, Pavel Martásek, Vladimir Král, Bettie Sue S. Masters, Thomas L. Poulos
Journal of Inorganic Biochemistry 2000 Volume 81(Issue 3) pp:133-139
Publication Date(Web):31 August 2000
DOI:10.1016/S0162-0134(00)00099-4
Analyzing the active site topology and plasticity of nitric oxide synthase (NOS) and understanding enzyme-drug interactions are crucial for the development of potent, isoform-selective NOS inhibitors. A small hydrophobic pocket in the active site is identified in the bovine eNOS heme domain structures complexed with potent isothiourea inhibitors: seleno analogue of S-ethyl-isothiourea, S-isopropyl-isothiourea, and 2-aminothiazoline, respectively. These structures reveal the importance of nonpolar van der Waals contacts in addition to the well-known hydrogen bonding interactions between inhibitor and enzyme. The scaffold of a potent NOS inhibitor should be capable of donating hydrogen bonds to as well as making nonpolar contacts with amino acids in the NOS active site.
Co-reporter:Chad E. Immoos;Irina F. Sevrioukova;Patric Farmer;Irina F. Sevrioukova;Patric Farmer;Chad E. Immoos
Israel Journal of Chemistry 2000 Volume 40(Issue 1) pp:47-53
Publication Date(Web):8 MAR 2010
DOI:10.1560/VRGJ-DE54-XDNM-5PH3

Two mutants of the heme domain of Bacillus megaterium cytochrome P450BM-3 (BMP) have been modified by covalent attachment of a photoactive Ru(bpy)3 complex at the surface-exposed cysteine residues, 62 and 387. The laser-flash/quench technique was used to study Ru1+ Fe3+ electron transfer (ET) within the Ru-BMP complexes. There was no reduction of the ferric heme by Ru1+ in Ru-62-BMP. In Ru-387-BMP, ET from Ru1+ to Fe3+ occurred with the rate constant of 4.6 × 105 s−1 and 2.5 × 106 s−1 in the absence and presence of substrate, respectively. The study demonstrates the importance of through-bond pathways for electron flow to the heme iron of P450 and that the Gln387–Cys400 peptide is a potential ET root in P450BM-3.

Co-reporter:
Nature Structural and Molecular Biology 2000 7(10) pp:876-880
Publication Date(Web):
DOI:10.1038/82820
CooA is a homodimeric transcription factor that belongs to the catabolite activator protein (CAP) family. Binding of CO to the heme groups of CooA leads to the transcription of genes involved in CO oxidation in Rhodospirillum rubrum. The 2.6 Å structure of reduced (Fe2+) CooA reveals that His 77 in both subunits provides one heme ligand while the N-terminal nitrogen of Pro 2 from the opposite subunit provides the other ligand. A structural comparison of CooA in the absence of effector and DNA (off state) with that of CAP in the effector and DNA bound state (on state) leads to a plausible model for the mechanism of allosteric control in this class of proteins as well as the CO dependent activation of CooA.
Co-reporter:
Nature Structural and Molecular Biology 1999 6(9) pp:860 - 867
Publication Date(Web):
DOI:10.1038/12319
Co-reporter:Sarvind Tripathi, Maura J. O'Neill, Angela Wilks, Thomas L. Poulos
Journal of Inorganic Biochemistry (November 2013) Volume 128() pp:131-136
Publication Date(Web):1 November 2013
DOI:10.1016/j.jinorgbio.2013.07.030
•1.98Å crystal structure of the P. aeruginosa cytoplasmic heme binding protein.•The structure is very similar to other heme binding proteins from other pathogenic bacteria.•Modeling studies suggest two binding modes for heme.•Multiple heme binding modes explains some of the biochemical/biophysical properties of PhuSIron is an essential element to all living organisms and is an important determinant of bacterial virulence. Bacteria have evolved specialized systems to sequester and transport iron from the environment or host. Pseudomonas aeruginosa, an opportunistic pathogen, uses two outer membrane receptor mediated systems (Phu and Has) to utilize host heme as a source of iron. PhuS is a 39 kDa soluble cytoplasmic heme binding protein which interacts and transports heme from the inner membrane heme transporter to the cytoplasm where it is degraded by heme oxygenase thus releasing iron. PhuS is unique among other cytoplasmic heme transporter proteins owing to the presence of three histidines in the heme binding pocket which can potentially serve as heme ligands. Out of the three histidine residues on the heme binding helix, His 209 is conserved among heme trafficking proteins while His 210 and His 212 are unique to PhuS. Here we report the crystal structure of PhuS at 1.98 Å resolution which shows a unique heme binding pocket and oligomeric structure compared to other known cytoplasmic heme transporter and accounts for some of the unusual biochemical properties of PhuS.PhuS structure showing the predicted location of the heme required for heme transfer to heme oxygenase.Download full-size image
Co-reporter:Thomas L. Poulos, Huiying Li
Nitric Oxide (28 February 2017) Volume 63() pp:68-77
Publication Date(Web):28 February 2017
DOI:10.1016/j.niox.2016.11.004
•Nitric oxide synthase.•Structure based drug design.•Isoform selectivity.•Neurodegeneration.•Melanoma.Once it was discovered that the enzyme nitric oxide synthase (NOS) is responsible for the biosynthesis of NO, NOS became a drug target. Particularly important is the over production of NO by neuronal NOS (nNOS) in various neurodegenerative disorders. After the various NOS isoforms were identified, inhibitor development proceeded rapidly. It soon became evident, however, that isoform selectivity presents a major challenge. All 3 human NOS isoforms, nNOS, eNOS (endothelial NOS), and iNOS (inducible NOS) have nearly identical active site structures thus making selective inhibitor design especially difficult. Of particular importance is the avoidance of inhibiting eNOS owing to its vital role in the cardiovascular system. This review summarizes some of the history of NOS inhibitor development and more recent advances in developing isoform selective inhibitors using primarily structure-based approaches.
Co-reporter:Irina F. Sevrioukova, Thomas L. Poulos
Archives of Biochemistry and Biophysics (1 March 2011) Volume 507(Issue 1) pp:
Publication Date(Web):1 March 2011
DOI:10.1016/j.abb.2010.08.022
The P450cam monooxygenase system consists of three separate proteins: the FAD-containing, NADH-dependent oxidoreductase (putidaredoxin reductase or Pdr), cytochrome P450cam and the 2Fe2S ferredoxin (putidaredoxin or Pdx), which transfers electrons from Pdr to P450cam. Over the past few years our lab has focused on the interaction between these redox components. It has been known for some time that Pdx can serve as an effector in addition to its electron shuttle role. The binding of Pdx to P450cam is thought to induce structural changes in the P450cam active site that couple electron transfer to substrate hydroxylation. The nature of these structural changes has remained unclear until a particular mutant of P450cam (Leu358Pro) was found to exhibit spectral perturbations similar to those observed in wild type P450cam bound to Pdx. The crystal structure of the L358P variant has provided some important insights on what might be happening when Pdx docks. In addition to these studies, many Pdx mutants have been analyzed to identify regions important for electron transfer. Somewhat surprisingly, we found that Pdx residues predicted to be at the P450cam–Pdx interface play different roles in the reduction of ferric P450cam and the ferrous P450–O2 complex. More recently we have succeeded in obtaining the structure of a chemically cross-linked Pdr–Pdx complex. This fusion protein represents a valid model for the noncovalent Pdr–Pdx complex as it retains the redox activities of native Pdr and Pdx and supports monooxygenase reactions catalyzed by P450cam. The insights gained from these studies will be summarized in this review.Research highlights► P450cam monooxygenase system. ► Crystal structure of the putidaredoxin (Pdx)–putidaredoxin reductase (Pdr) complex. ► The structural basis for the effector role of Pdx on P450cam camphor hydroxylation. ► Mutagenesis and kinetics to test the validity of structural models.
Acetamide, N-[7-(bromomethyl)-2-quinolinyl]-
2-BROMO-6-(2,5-DIMETHYLPYRROL-1-YL)-4-METHYLPYRIDINE
2H-1-Benzopyran-2-one, 7-(phenylmethoxy)-4-(trifluoromethyl)-
6-ethynyl-1H-Pyrrolo[2,3-b]pyridine
3,5-bis(bromomethyl)benzonitrile
Midazolam
L-Ornithine,N5-[(hydroxyamino)iminomethyl]-
(5-BROMO-PYRIDIN-3-YL)-ACETONITRILE
L-Ornithine,N5-(1-iminoethyl)-
4-Methylquinolin-2-amine