1
|
Samanta S, Mondal P. A Comprehensive Computational Study on the Thermodynamics and Kinetics of Tetrahydrobiopterin Regeneration Process. Chemphyschem 2024; 25:e202400401. [PMID: 38861155 DOI: 10.1002/cphc.202400401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/12/2024]
Abstract
One of the most crucial enzymatic cofactors in the human body is tetrahydrobiopterin, which is acquired through biological synthesis and self-regeneration. During this regenerative process, it undergoes oxidation, deprotonation, further oxidation, and subsequent deprotonation, resulting in the formation of quinonoid-dihydrobiopterin, which then undergoes tautomerization to yield dihydrobiopterin. This study presents the thermodynamic and kinetic properties associated with each stage of the regeneration process using theoretical calculations. The redox potentials for oxidation steps and the pKa values for deprotonation steps are determined employing the Born-Haber cycle and the direct change of free energy in implicit solvent models. The redox metabolites are characterized and confirmed from their calculated absorption spectra using the time-dependent density functional theory method. For the tautomerization steps, an IRC calculation is executed, and rate constants are computed using Eyring's Transition State Theory (TST). The tunnelling probability of the H atom during the tautomerization process is incorporated using Wigner's tunnelling correction in the calculation of the rate constant. Notably, we identify the N3 atom as the most probable deprotonation site for H3B+ and predict its geometry based on our calculations. Furthermore, we elucidate the spectral properties of intermediates involved in the regeneration process, highlighting key electronic transitions responsible for their excitations. Our results indicate that each step of tautomerization occurs along vibrational bending modes. We have observed that these tautomerization processes have high activation energies by optimising transition states. Additionally, considering tunnelling correction can significantly affect the reaction rates associated with these processes. These results provide a comprehensive understanding of the thermodynamics and kinetics of the regeneration process of tetrahydrobiopterin, which will help in the modulation of its biological activity.
Collapse
Affiliation(s)
- Suvadip Samanta
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Tirupati, India
| | - Padmabati Mondal
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Tirupati, India
- Center for Atomic, Molecular and Optical Sciences and Technologies, Indian Institute of Science Education and Research (IISER), Tirupati, India
| |
Collapse
|
2
|
Maccallini C, Budriesi R, De Filippis B, Amoroso R. Advancements in the Research of New Modulators of Nitric Oxide Synthases Activity. Int J Mol Sci 2024; 25:8486. [PMID: 39126054 PMCID: PMC11313090 DOI: 10.3390/ijms25158486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Nitric oxide (NO) has been defined as the "miracle molecule" due to its essential pleiotropic role in living systems. Besides its implications in physiologic functions, it is also involved in the development of several disease states, and understanding this ambivalence is crucial for medicinal chemists to develop therapeutic strategies that regulate NO production without compromising its beneficial functions in cell physiology. Although nitric oxide synthase (NOS), i.e., the enzyme deputed to the NO biosynthesis, is a well-recognized druggable target to regulate NO bioavailability, some issues have emerged during the past decades, limiting the progress of NOS modulators in clinical trials. In the present review, we discuss the most promising advancements in the research of small molecules that are able to regulate NOS activity with improved pharmacodynamic and pharmacokinetic profiles, providing an updated framework of this research field that could be useful for the design and development of new NOS modulators.
Collapse
Affiliation(s)
- Cristina Maccallini
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (B.D.F.); (R.A.)
| | - Roberta Budriesi
- Department of Pharmacy and Biotechnology, Food Chemistry and Nutraceutical Lab, Alma Mater Studiorum-University of Bologna, 40126 Bologna, Italy;
| | - Barbara De Filippis
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (B.D.F.); (R.A.)
| | - Rosa Amoroso
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (B.D.F.); (R.A.)
| |
Collapse
|
3
|
Satpathy JK, Yadav R, Bagha UK, Kumar D, Sastri CV, de Visser SP. Enhanced Reactivity through Equatorial Sulfur Coordination in Nonheme Iron(IV)-Oxo Complexes: Insights from Experiment and Theory. Inorg Chem 2024; 63:6752-6766. [PMID: 38551622 DOI: 10.1021/acs.inorgchem.4c00070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Sulfur ligation in metalloenzymes often gives the active site unique properties, whether it is the axial cysteinate ligand in the cytochrome P450s or the equatorial sulfur/thiol ligation in nonheme iron enzymes. To understand sulfur ligation to iron complexes and how it affects the structural, spectroscopic, and intrinsic properties of the active species and the catalysis of substrates, we pursued a systematic study and compared sulfur with amine-ligated iron(IV)-oxo complexes. We synthesized and characterized a biomimetic N4S-ligated iron(IV)-oxo complex and compared the obtained results with an analogous N5-ligated iron(IV)-oxo complex. Our work shows that the amine for sulfur replacement in the equatorial ligand framework leads to a rate enhancement for oxygen atom and hydrogen atom transfer reactions. Moreover, the sulfur-ligated iron(IV)-oxo complex reacts through a different reaction mechanism as compared to the N5-ligated iron(IV)-oxo complex, where the former reacts through hydride transfer with the latter reacting via radical pathways. We show that the reactivity differences are caused by a dramatic change in redox potential between the two complexes. Our studies highlight the importance of implementing a sulfur ligand into the equatorial ligand framework of nonheme iron(IV)-oxo complexes and how it affects the physicochemical properties of the oxidant and its reactivity.
Collapse
Affiliation(s)
- Jagnyesh K Satpathy
- Department of Chemistry, Indian Institute of Technology, Guwahati 781039, Assam, India
| | - Rolly Yadav
- Department of Chemistry, Indian Institute of Technology, Guwahati 781039, Assam, India
| | - Umesh K Bagha
- Department of Chemistry, Indian Institute of Technology, Guwahati 781039, Assam, India
| | - Devesh Kumar
- Department of Applied Physics, Babasaheb Bhimrao Ambedkar University, School for Physical Sciences, Vidya Vihar, Rae Bareilly Road, Lucknow 226025, UP, India
| | - Chivukula V Sastri
- Department of Chemistry, Indian Institute of Technology, Guwahati 781039, Assam, India
| | - Sam P de Visser
- Department of Chemistry, Indian Institute of Technology, Guwahati 781039, Assam, India
- The Manchester Institute of Biotechnology and Department of Chemical Engineering, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| |
Collapse
|
4
|
Teng KX, Zhang D, Liu BK, Liu ZF, Niu LY, Yang QZ. Photo-Induced Disproportionation-Mediated Photodynamic Therapy: Simultaneous Oxidation of Tetrahydrobiopterin and Generation of Superoxide Radicals. Angew Chem Int Ed Engl 2024; 63:e202318783. [PMID: 38258371 DOI: 10.1002/anie.202318783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 01/24/2024]
Abstract
We herein present an approach of photo-induced disproportionation for preparation of Type-I photodynamic agents. As a proof of concept, BODIPY-based photosensitizers were rationally designed and prepared. The photo-induced intermolecular electron transfer between homotypic chromophores leads to the disproportionation reaction, resulting in the formation of charged intermediates, cationic and anionic radicals. The cationic radicals efficiently oxidize the cellularimportant coenzyme, tetrahydrobiopterin (BH4 ), and the anionic radicals transfer electrons to oxygen to produce superoxide radicals (O2 - ⋅). One of our Type-I photodynamic agents not only self-assembles in water but also effectively targets the endoplasmic reticulum. It displayed excellent photocytotoxicity even in highly hypoxic environments (2 % O2 ), with a half-maximal inhibitory concentration (IC50 ) of 0.96 μM, and demonstrated outstanding antitumor efficacy in murine models bearing HeLa tumors.
Collapse
Affiliation(s)
- Kun-Xu Teng
- Institution Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, P. R. China
| | - Dongsheng Zhang
- Institution Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, P. R. China
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, P. R. China
| | - Bin-Kai Liu
- Institution Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, P. R. China
| | - Zheng-Fei Liu
- Institution Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, P. R. China
| | - Li-Ya Niu
- Institution Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, P. R. China
| | - Qing-Zheng Yang
- Institution Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, P. R. China
| |
Collapse
|
5
|
Kobayashi K, Ito YT, Kasu Y, Horitani M, Kozawa T. Intramolecular electron transfer from biopterin to Fe II-O 2 complex in nitric oxide synthases occurs at very different rates between bacterial and mammalian enzymes: Direct observation of a catalytically active intermediate. J Inorg Biochem 2023; 238:112035. [PMID: 36327499 DOI: 10.1016/j.jinorgbio.2022.112035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/05/2022] [Accepted: 10/17/2022] [Indexed: 11/21/2022]
Abstract
Nitric oxide synthase (NOS) is a cytochrome P450-type mono‑oxygenase that catalyzes the oxidation of L-arginine to nitric oxide. We previously observed that intramolecular electron transfer from biopterin to Fe2+-O2 in Deinococcus radiodurans NOS (DrNOS) using pulse radiolysis. However, the rate of electron transfer in DrNOS (2.2 × 103 s-1) contrasts with a reported corresponding rate (11 s-1) in a mammalian NOS determined using rapid freeze-quench (RFQ) EPR. We applied pulse radiolysis to Bacillus subtilis NOS (bsNOS) and to rat neural NOS oxygenase domain NOS (mNOS). Concurrently, RFQ EPR was used to trap a pterin radical during single-turnover enzyme reactions of the enzymes. By using the pulse radiolysis method, hydrated electrons (eaq-) reduced the heme iron of NOS enzymes. Subsequently, ferrous heme reacted with O2 to form a Fe2+-O2 intermediate. In the presence of pterin, the intermediate of bsNOS was found to convert to other intermediate in the time range of milliseconds. A similar process was determined to have occurred after pulse radiolysis of the pterin-bound mNOS, though the rate was much slower. The intermediates of all of the NOS enzymes further converted to the original ferric form in the time range of seconds. When using the RFQ method, pterin radicals were formed very rapidly in both DrNOS and bsNOS in the time range of milliseconds. In contrast, the pterin radical in mNOS was observed to form slowly, at a rate of ∼20 s-1.
Collapse
Affiliation(s)
- Kazuo Kobayashi
- The Institute of Scientific and Industrial Research, Osaka University, Mihogaoka 8-1, Ibaraki, Osaka 567-0047, Japan.
| | - Yuko Tsutsui Ito
- The Institute of Scientific and Industrial Research, Osaka University, Mihogaoka 8-1, Ibaraki, Osaka 567-0047, Japan
| | - Yuri Kasu
- Department of Applied Biochemistry and Food Science, Saga University, Honjo-machi Saga, 840-8502, Japan
| | - Masaki Horitani
- Department of Applied Biochemistry and Food Science, Saga University, Honjo-machi Saga, 840-8502, Japan; The United Graduate School of Agricultural Sciences, Kagoshima University, 1-21-24 Korimoto, Kagoshima, Kagoshima 890-0065, Japan
| | - Takahiro Kozawa
- The Institute of Scientific and Industrial Research, Osaka University, Mihogaoka 8-1, Ibaraki, Osaka 567-0047, Japan
| |
Collapse
|
6
|
Insights into Molecular Structure of Pterins Suitable for Biomedical Applications. Int J Mol Sci 2022; 23:ijms232315222. [PMID: 36499560 PMCID: PMC9737128 DOI: 10.3390/ijms232315222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/07/2022] Open
Abstract
Pterins are an inseparable part of living organisms. Pterins participate in metabolic reactions mostly as tetrahydropterins. Dihydropterins are usually intermediates of these reactions, whereas oxidized pterins can be biomarkers of diseases. In this review, we analyze the available data on the quantum chemistry of unconjugated pterins as well as their photonics. This gives a comprehensive overview about the electronic structure of pterins and offers some benefits for biomedicine applications: (1) one can affect the enzymatic reactions of aromatic amino acid hydroxylases, NO synthases, and alkylglycerol monooxygenase through UV irradiation of H4pterins since UV provokes electron donor reactions of H4pterins; (2) the emission properties of H2pterins and oxidized pterins can be used in fluorescence diagnostics; (3) two-photon absorption (TPA) should be used in such pterin-related infrared therapy because single-photon absorption in the UV range is inefficient and scatters in vivo; (4) one can affect pathogen organisms through TPA excitation of H4pterin cofactors, such as the molybdenum cofactor, leading to its detachment from proteins and subsequent oxidation; (5) metal nanostructures can be used for the UV-vis, fluorescence, and Raman spectroscopy detection of pterin biomarkers. Therefore, we investigated both the biochemistry and physical chemistry of pterins and suggested some potential prospects for pterin-related biomedicine.
Collapse
|
7
|
Colston KJ, Basu P. Synthesis, Redox and Spectroscopic Properties of Pterin of Molybdenum Cofactors. Molecules 2022; 27:3324. [PMID: 35630801 PMCID: PMC9146068 DOI: 10.3390/molecules27103324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/10/2022] Open
Abstract
Pterins are bicyclic heterocycles that are found widely across Nature and are involved in a variety of biological functions. Notably, pterins are found at the core of molybdenum cofactor (Moco) containing enzymes in the molybdopterin (MPT) ligand that coordinates molybdenum and facilitates cofactor activity. Pterins are diverse and can be widely functionalized to tune their properties. Herein, the general methods of synthesis, redox and spectroscopic properties of pterin are discussed to provide more insight into pterin chemistry and their importance to biological systems.
Collapse
Affiliation(s)
| | - Partha Basu
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA;
| |
Collapse
|
8
|
Dent MR, DeMartino AW, Tejero J, Gladwin MT. Endogenous Hemoprotein-Dependent Signaling Pathways of Nitric Oxide and Nitrite. Inorg Chem 2021; 60:15918-15940. [PMID: 34313417 PMCID: PMC9167621 DOI: 10.1021/acs.inorgchem.1c01048] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Interdisciplinary research at the interface of chemistry, physiology, and biomedicine have uncovered pivotal roles of nitric oxide (NO) as a signaling molecule that regulates vascular tone, platelet aggregation, and other pathways relevant to human health and disease. Heme is central to physiological NO signaling, serving as the active site for canonical NO biosynthesis in nitric oxide synthase (NOS) enzymes and as the highly selective NO binding site in the soluble guanylyl cyclase receptor. Outside of the primary NOS-dependent biosynthetic pathway, other hemoproteins, including hemoglobin and myoglobin, generate NO via the reduction of nitrite. This auxiliary hemoprotein reaction unlocks a "second axis" of NO signaling in which nitrite serves as a stable NO reservoir. In this Forum Article, we highlight these NO-dependent physiological pathways and examine complex chemical and biochemical reactions that govern NO and nitrite signaling in vivo. We focus on hemoprotein-dependent reaction pathways that generate and consume NO in the presence of nitrite and consider intermediate nitrogen oxides, including NO2, N2O3, and S-nitrosothiols, that may facilitate nitrite-based signaling in blood vessels and tissues. We also discuss emergent therapeutic strategies that leverage our understanding of these key reaction pathways to target NO signaling and treat a wide range of diseases.
Collapse
Affiliation(s)
- Matthew R Dent
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Anthony W DeMartino
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Jesús Tejero
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Mark T Gladwin
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
9
|
Visser SP. Second‐Coordination Sphere Effects on Selectivity and Specificity of Heme and Nonheme Iron Enzymes. Chemistry 2020; 26:5308-5327. [DOI: 10.1002/chem.201905119] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/04/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Sam P. Visser
- The Manchester Institute of Biotechnology and Department of Chemical Engineering and Analytical ScienceThe University of Manchester 131 Princess Street Manchester M1 7DN UK
| |
Collapse
|
10
|
Li J, Zheng H, Feng C. Effect of Macromolecular Crowding on the FMN-Heme Intraprotein Electron Transfer in Inducible NO Synthase. Biochemistry 2019; 58:3087-3096. [PMID: 31251033 DOI: 10.1021/acs.biochem.9b00193] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Previous biochemical studies of nitric oxide synthase enzymes (NOSs) were conducted in diluted solutions. However, the intracellular milieu where the proteins perform their biological functions is crowded with macromolecules. The effect of crowding on the electron transfer kinetics of multidomain proteins is much less understood. Herein, we investigated the effect of macromolecular crowding on the FMN-heme intraprotein interdomain electron transfer (IET), an obligatory step in NOS catalysis. A noticeable increase in the IET rate in the bidomain oxygenase/FMN (oxyFMN) and the holoprotein of human inducible NOS (iNOS) was observed upon addition of Ficoll 70 in a nonsaturable manner. Additionally, the magnitude of IET enhancement for the holoenzyme is much higher than that that of the oxyFMN construct. The crowding effect is also evident at different ionic strengths. Importantly, the enhancing extent is similar for the iNOS oxyFMN protein with added Ficoll 70 and Dextran 70 that give the same solution viscosity, showing that specific interactions do not exist between the NOS protein and the crowder. Moreover, the population of the docked FMN-heme state is significantly increased upon addition of Ficoll 70 and the fluorescence lifetime values do not correspond to those in the absence of Ficoll 70. The steady-state cytochrome c reduction by the holoenzyme is noticeably enhanced by the crowder, while the ferricyanide reduction is unchanged. The NO production activity of the iNOS holoenzyme is stimulated by Ficoll 70. The effect of macromolecular crowding on the kinetics can be rationalized on the basis of the excluded volume effect, with an entropic origin. The intraprotein electron transfer kinetics, fluorescence lifetime, and steady-state enzymatic activity results indicate that macromolecular crowding modulates the NOS electron transfer through multiple pathways. Such a mechanism should be applicable to electron transfer in other multidomain redox proteins.
Collapse
Affiliation(s)
- Jinghui Li
- College of Pharmacy , University of New Mexico , Albuquerque , New Mexico 87131 , United States
| | - Huayu Zheng
- College of Pharmacy , University of New Mexico , Albuquerque , New Mexico 87131 , United States.,Department of Chemistry and Chemical Biology , University of New Mexico , Albuquerque , New Mexico 87131 , United States
| | - Changjian Feng
- College of Pharmacy , University of New Mexico , Albuquerque , New Mexico 87131 , United States.,Department of Chemistry and Chemical Biology , University of New Mexico , Albuquerque , New Mexico 87131 , United States
| |
Collapse
|
11
|
Tejero J, Shiva S, Gladwin MT. Sources of Vascular Nitric Oxide and Reactive Oxygen Species and Their Regulation. Physiol Rev 2019; 99:311-379. [PMID: 30379623 DOI: 10.1152/physrev.00036.2017] [Citation(s) in RCA: 290] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a small free radical with critical signaling roles in physiology and pathophysiology. The generation of sufficient NO levels to regulate the resistance of the blood vessels and hence the maintenance of adequate blood flow is critical to the healthy performance of the vasculature. A novel paradigm indicates that classical NO synthesis by dedicated NO synthases is supplemented by nitrite reduction pathways under hypoxia. At the same time, reactive oxygen species (ROS), which include superoxide and hydrogen peroxide, are produced in the vascular system for signaling purposes, as effectors of the immune response, or as byproducts of cellular metabolism. NO and ROS can be generated by distinct enzymes or by the same enzyme through alternate reduction and oxidation processes. The latter oxidoreductase systems include NO synthases, molybdopterin enzymes, and hemoglobins, which can form superoxide by reduction of molecular oxygen or NO by reduction of inorganic nitrite. Enzymatic uncoupling, changes in oxygen tension, and the concentration of coenzymes and reductants can modulate the NO/ROS production from these oxidoreductases and determine the redox balance in health and disease. The dysregulation of the mechanisms involved in the generation of NO and ROS is an important cause of cardiovascular disease and target for therapy. In this review we will present the biology of NO and ROS in the cardiovascular system, with special emphasis on their routes of formation and regulation, as well as the therapeutic challenges and opportunities for the management of NO and ROS in cardiovascular disease.
Collapse
Affiliation(s)
- Jesús Tejero
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Sruti Shiva
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
12
|
Shamovsky I, Belfield G, Lewis R, Narjes F, Ripa L, Tyrchan C, Öberg L, Sjö P. Theoretical studies of the second step of the nitric oxide synthase reaction: Electron tunneling prevents uncoupling. J Inorg Biochem 2018; 181:28-40. [PMID: 29407906 DOI: 10.1016/j.jinorgbio.2018.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/18/2017] [Accepted: 01/08/2018] [Indexed: 12/27/2022]
Abstract
Nitric oxide (NO·) is a messenger molecule with diverse physiological roles including host defense, neurotransmission and vascular function. The synthesis of NO· from l-arginine is catalyzed by NO-synthases and occurs in two steps through the intermediary Nω-hydroxy-l-arginine (NHA). In both steps the P450-like reaction cycle is coupled with the redox cycle of the cofactor tetrahydrobiopterin (H4B). The mechanism of the second step is studied by Density Functional Theory calculations to ascertain the canonical sequence of proton and electron transfer (PT and ET) events. The proposed mechanism is controlled by the interplay of two electron donors, H4B and NHA. Consistent with experimental data, the catalytic cycle proceeds through the ferric-hydroperoxide complex (Cpd 0) and the following aqua-ferriheme resting state, and involves interim partial oxidation of H4B. The mechanism starts with formation of Cpd 0 from the ferrous-dioxy reactant complex by PT from the C-ring heme propionate coupled with hole transfer to H4B through the highest occupied π-orbital of NHA as a bridge. This enables PT from NHA+· to the proximal oxygen leading to the shallow ferriheme-H2O2 oxidant. Subsequent Fenton-like peroxide bond cleavage triggered by ET from the NHA-derived iminoxy-radical leads to the protonated Cpd II diradicaloid singlet stabilized by spin delocalization in H4B, and the closed-shell coordination complex of HO- with iminoxy-cation. The complex is converted to the transient C-adduct, which releases intended products upon PT to the ferriheme-HO- complex coupled with ET to the H4B+·. Deferred ET from the substrate or undue ET from/to the cofactor leads to side products.
Collapse
Affiliation(s)
- Igor Shamovsky
- Department of Medicinal Chemistry, IMED RIA, AstraZeneca R&D Gothenburg, Pepparedsleden 1, 431 83 Mölndal, Sweden.
| | - Graham Belfield
- Department of Medicinal Chemistry, IMED RIA, AstraZeneca R&D Gothenburg, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Richard Lewis
- Department of Medicinal Chemistry, IMED RIA, AstraZeneca R&D Gothenburg, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Frank Narjes
- Department of Medicinal Chemistry, IMED RIA, AstraZeneca R&D Gothenburg, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Lena Ripa
- Department of Medicinal Chemistry, IMED RIA, AstraZeneca R&D Gothenburg, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Christian Tyrchan
- Department of Medicinal Chemistry, IMED RIA, AstraZeneca R&D Gothenburg, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Lisa Öberg
- Department of Medicinal Chemistry, IMED RIA, AstraZeneca R&D Gothenburg, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Peter Sjö
- Department of Medicinal Chemistry, IMED RIA, AstraZeneca R&D Gothenburg, Pepparedsleden 1, 431 83 Mölndal, Sweden
| |
Collapse
|
13
|
Abstract
Aerobic organisms have evolved to activate oxygen from the atmosphere, which allows them to catalyze the oxidation of different kinds of substrates. This activation of oxygen is achieved by a metal center (usually iron or copper) buried within a metalloprotein. In the case of iron-containing heme enzymes, the activation of oxygen is achieved by formation of transient iron-oxo (ferryl) intermediates; these intermediates are called Compound I and Compound II. The Compound I and II intermediates were first discovered in the 1930s in horseradish peroxidase, and it is now known that these same species are used across the family of heme enzymes, which include all of the peroxidases, the heme catalases, the P450s, cytochrome c oxidase, and NO synthase. Many years have passed since the first observations, but establishing the chemical nature of these transient ferryl species remains a fundamental question that is relevant to the reactivity, and therefore the usefulness, of these species in biology. This Account summarizes experiments that were conceived and conducted at Leicester and presents our ideas on the chemical nature, stability, and reactivity of these ferryl heme species. We begin by briefly summarizing the early milestones in the field, from the 1940s and 1950s. We present comparisons between the nature and reactivity of the ferryl species in horseradish peroxidase, cytochrome c peroxidase, and ascorbate peroxidase; and we consider different modes of electron delivery to ferryl heme, from different substrates in different peroxidases. We address the question of whether the ferryl heme is best formulated as an (unprotonated) FeIV═O or as a (protonated) FeIV-OH species. A range of spectroscopic approaches (EXAFS, resonance Raman, Mossbauer, and EPR) have been used over many decades to examine this question, and in the last ten years, X-ray crystallography has also been employed. We describe how information from all of these studies has blended together to create an overall picture, and how the recent application of neutron crystallography has directly identified protonation states and has helped to clarify the precise nature of the ferryl heme in cytochrome c peroxidase and ascorbate peroxidase. We draw comparisons between the Compound I and Compound II species that we have observed in peroxidases with those found in other heme systems, notably the P450s, highlighting possible commonality across these heme ferryl systems. The identification of proton locations from neutron structures of these ferryl species opens the door for understanding the proton translocations that need to occur during O-O bond cleavage.
Collapse
Affiliation(s)
- Peter C. E. Moody
- Department
of Molecular and Cell Biology and Leicester Institute of Structural
and Chemical Biology, University of Leicester, Lancaster Road, Leicester LE1 9HN, England
| | - Emma L. Raven
- Department
of Chemistry and Leicester Institute of Structural and Chemical Biology, University of Leicester, University Road, Leicester LE1 7RH, U.K
| |
Collapse
|
14
|
Haque MM, Tejero J, Bayachou M, Kenney CT, Stuehr DJ. A cross-domain charge interaction governs the activity of NO synthase. J Biol Chem 2018; 293:4545-4554. [PMID: 29414777 DOI: 10.1074/jbc.ra117.000635] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 01/17/2018] [Indexed: 11/06/2022] Open
Abstract
NO synthase (NOS) enzymes perform interdomain electron transfer reactions during catalysis that may rely on complementary charge interactions at domain-domain interfaces. Guided by our previous results and a computer-generated domain-docking model, we assessed the importance of cross-domain charge interactions in the FMN-to-heme electron transfer in neuronal NOS (nNOS). We reversed the charge of three residues (Glu-762, Glu-816, and Glu-819) that form an electronegative triad on the FMN domain and then individually reversed the charges of three electropositive residues (Lys-423, Lys-620, and Lys-660) on the oxygenase domain (NOSoxy), to potentially restore a cross-domain charge interaction with the triad, but in reversed polarity. Charge reversal of the triad completely eliminated heme reduction and NO synthesis in nNOS. These functions were partly restored by the charge reversal at oxygenase residue Lys-423, but not at Lys-620 or Lys-660. Full recovery of heme reduction was probably muted by an accompanying change in FMN midpoint potential that made electron transfer to the heme thermodynamically unfavorable. Our results provide direct evidence that cross-domain charge pairing is required for the FMN-to-heme electron transfer in nNOS. The unique ability of charge reversal at position 423 to rescue function indicates that it participates in an essential cross-domain charge interaction with the FMN domain triad. This supports our domain-docking model and suggests that it may depict a productive electron transfer complex formed during nNOS catalysis.
Collapse
Affiliation(s)
- Mohammad Mahfuzul Haque
- From the Departments of Pathobiology, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio 44195
| | - Jesús Tejero
- the Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, and
| | - Mekki Bayachou
- the Department of Chemistry, Cleveland State University, Cleveland, Ohio 44115
| | - Claire T Kenney
- From the Departments of Pathobiology, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio 44195
| | - Dennis J Stuehr
- From the Departments of Pathobiology, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio 44195,
| |
Collapse
|
15
|
Perez CE, Park HB, Crawford JM. Functional Characterization of a Condensation Domain That Links Nonribosomal Peptide and Pteridine Biosynthetic Machineries in Photorhabdus luminescens. Biochemistry 2018; 57:354-361. [PMID: 29111689 DOI: 10.1021/acs.biochem.7b00863] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Nonribosomal peptide synthetases (NRPSs) produce a wide variety of biologically important small molecules. NRPSs can interface with other enzymes to form hybrid biosynthetic systems that expand the structural and functional diversity of their products. The pepteridines are metabolites encoded by an unprecedented pteridine-NRPS-type hybrid biosynthetic gene cluster in Photorhabdus luminescens, but how the distinct enzymatic systems interface to produce these molecules has not been examined at the biochemical level. By an unknown mechanism, the genetic locus can also affect the regulation of other enzymes involved in autoinducer and secondary metabolite biosynthesis. Here, through in vitro protein biochemical assays, we demonstrate that an atypical NRPS condensation (C) domain present in the pathway condenses acyl units derived from α-keto acids onto a free 5,6,7,8-tetrahydropterin core, producing the tertiary cis-amide-containing pepteridines. Solution studies of the chemically synthesized molecules led to the same amide regiochemistries that were observed in the natural products. The biochemical transformations mediated by the C domain destroy the radical scavenging activity of its redox active tetrahydropterin substrate. Secondary metabolite analyses revealed that the pepteridine locus affects select metabolic pathways associated with quorum sensing, antibiosis, and symbiosis. Taken together, the results suggest that the pathway likely regulates cellular redox and specialized metabolic pathways through engagement with the citric acid cycle.
Collapse
Affiliation(s)
- Corey E Perez
- Department of Chemistry, Yale University , New Haven, Connecticut 06520, United States.,Chemical Biology Institute, Yale University , West Haven, Connecticut 06516, United States
| | - Hyun Bong Park
- Department of Chemistry, Yale University , New Haven, Connecticut 06520, United States.,Chemical Biology Institute, Yale University , West Haven, Connecticut 06516, United States
| | - Jason M Crawford
- Department of Chemistry, Yale University , New Haven, Connecticut 06520, United States.,Chemical Biology Institute, Yale University , West Haven, Connecticut 06516, United States.,Department of Microbial Pathogenesis, Yale School of Medicine , New Haven, Connecticut 06510, United States
| |
Collapse
|
16
|
Weisslocker-Schaetzel M, Lembrouk M, Santolini J, Dorlet P. Revisiting the Val/Ile Mutation in Mammalian and Bacterial Nitric Oxide Synthases: A Spectroscopic and Kinetic Study. Biochemistry 2017; 56:748-756. [PMID: 28074650 DOI: 10.1021/acs.biochem.6b01018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Nitric oxide is produced in mammals by the nitric oxide synthase (NOS) isoforms at a catalytic site comprising a heme associated with a biopterin cofactor. Through genome sequencing, proteins that are highly homologous to the oxygenase domain of NOSs have been identified, in particular in bacteria. The active site is highly conserved except for a valine residue in the distal pocket that is replaced with an isoleucine in bacteria. This switch was previously reported to influence the kinetics of the reaction. We have used the V346I mutant of the mouse inducible NOS (iNOS) as well as the I224V mutant of the NOS from Bacillus subtilis (bsNOS) to study their spectroscopic signatures in solution and look for potential structural differences compared to their respective wild types. Both mutants seem destabilized in the absence of substrate and cofactor. When both substrate and cofactor are present, small differences can be detected with Nω-hydroxy-l-arginine compared to arginine, which is likely due to the differences in the hydrogen bonding network of the distal pocket. Stopped-flow experiments evidence significant changes in the kinetics of the reaction due to the mutation as was already known. We found these effects particularly marked for iNOS. On the basis of these results, we performed rapid freeze-quench experiments to trap the biopterin radical and found the same results that we had obtained for the wild types. Despite differences in kinetics, a radical could be trapped in both steps for the iNOS mutant but only for the first step in the mutant of bsNOS. This strengthens the hypothesis that mammalian and bacterial NOSs may have a different mechanism during the second catalytic step.
Collapse
Affiliation(s)
- Marine Weisslocker-Schaetzel
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay , F-91198 Gif-sur-Yvette cedex, France
| | - Mehdi Lembrouk
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay , F-91198 Gif-sur-Yvette cedex, France
| | - Jérôme Santolini
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay , F-91198 Gif-sur-Yvette cedex, France
| | - Pierre Dorlet
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay , F-91198 Gif-sur-Yvette cedex, France
| |
Collapse
|
17
|
Ramasamy S, Haque MM, Gangoda M, Stuehr DJ. Tetrahydrobiopterin redox cycling in nitric oxide synthase: evidence supports a through-heme electron delivery. FEBS J 2016; 283:4491-4501. [PMID: 27760279 PMCID: PMC5387691 DOI: 10.1111/febs.13933] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/06/2016] [Accepted: 10/17/2016] [Indexed: 11/30/2022]
Abstract
The nitric oxide synthases (NOS) catalyze a two-step oxidation of l-arginine (Arg) to generate NO. In the first step, O2 activation involves one electron being provided to the heme by an enzyme-bound 6R-tetrahydro-l-biopterin cofactor (H4 B), and the H4 B radical must be reduced back to H4 B in order for NOS to continue catalysis. Although an NADPH-derived electron is used to reduce the H4 B radical, how this occurs is unknown. We hypothesized that the NOS flavoprotein domain might reduce the H4 B radical by utilizing the NOS heme porphyrin as a conduit to deliver the electron. This model predicts that factors influencing NOS heme reduction should also influence the extent and rate of H4 B radical reduction in kind. To test this, we utilized single catalytic turnover and stop-freeze methods, along with electron paramagnetic resonance spectroscopy, to measure the rate and extent of reduction of the 5-methyl-H4 B radical formed in neuronal NOS (nNOS) during Arg hydroxylation. We used several nNOS variants that supported either a slower or faster than normal rate of ferric heme reduction. We found that the rates and extents of nNOS heme reduction correlated well with the rates and extents of 5-methyl-H4 B radical reduction among the various nNOS enzymes. This supports a model where the heme porphyrin transfers an electron from the NOS flavoprotein to the H4 B radical formed during catalysis, revealing that the heme plays a dual role in catalyzing O2 activation or electron transfer at distinct points in the reaction cycle.
Collapse
Affiliation(s)
- Somasundaram Ramasamy
- Department of Pathobiology, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Mohammad Mahfuzul Haque
- Department of Pathobiology, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Mahinda Gangoda
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH 44242, USA
| | - Dennis J. Stuehr
- Department of Pathobiology, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio 44195, USA
| |
Collapse
|
18
|
Krzyaniak MD, Cruce AA, Vennam P, Lockart M, Berka V, Tsai AL, Bowman MK. The tetrahydrobiopterin radical interacting with high- and low-spin heme in neuronal nitric oxide synthase - A new indicator of the extent of NOS coupling. Free Radic Biol Med 2016; 101:367-377. [PMID: 27989753 PMCID: PMC5362310 DOI: 10.1016/j.freeradbiomed.2016.10.503] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 10/07/2016] [Accepted: 10/26/2016] [Indexed: 01/10/2023]
Abstract
Reaction intermediates trapped during the single-turnover reaction of the neuronal ferrous nitric oxide synthase oxygenase domain (Fe(II)nNOSOX) show four EPR spectra of free radicals. Fully-coupled nNOSOX with cofactor (tetrahydrobiopterin, BH4) and substrate (l-arginine) forms the typical BH4 cation radical with an EPR spectrum ~4.0mT wide and hyperfine tensors similar to reports for a biopterin cation radical in inducible NOSOX (iNOSOX). With excess thiol, nNOSox lacking BH4 and l-arg is known to produce superoxide. In contrast, we find that nNOSOX with BH4 but no l-arg forms two radicals with rather different, fast (~250μs at 5K) and slower (~500μs at 20K), electron spin relaxation rates and a combined ~7.0mT wide EPR spectrum. Rapid freeze-quench CW- and pulsed-EPR measurements are used to identify these radicals and their origin. These two species are the same radical with identical nuclear hyperfine couplings, but with spin-spin couplings to high-spin (4.0mT component) or low-spin (7.0mT component) Fe(III) heme. Uncoupled reactions of nNOS leave the enzyme in states that can be chemically reduced to sustain unregulated production of NO and reactive oxygen species in ischemia-reperfusion injury. The broad EPR signal is a convenient indicator of uncoupled nNOS reactions producing low-spin Fe(III) heme.
Collapse
Affiliation(s)
- Matthew D Krzyaniak
- Department of Chemistry, The University of Alabama, Tuscaloosa, AL 35487-0336, USA
| | - Alex A Cruce
- Department of Chemistry, The University of Alabama, Tuscaloosa, AL 35487-0336, USA
| | - Preethi Vennam
- Department of Chemistry, The University of Alabama, Tuscaloosa, AL 35487-0336, USA
| | - Molly Lockart
- Department of Chemistry, The University of Alabama, Tuscaloosa, AL 35487-0336, USA
| | - Vladimir Berka
- Division of Hematology, Department of Internal Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Ah-Lim Tsai
- Division of Hematology, Department of Internal Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Michael K Bowman
- Department of Chemistry, The University of Alabama, Tuscaloosa, AL 35487-0336, USA.
| |
Collapse
|
19
|
Hedison TM, Leferink NGH, Hay S, Scrutton NS. Correlating Calmodulin Landscapes with Chemical Catalysis in Neuronal Nitric Oxide Synthase using Time-Resolved FRET and a 5-Deazaflavin Thermodynamic Trap. ACS Catal 2016; 6:5170-5180. [PMID: 27563493 PMCID: PMC4993522 DOI: 10.1021/acscatal.6b01280] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 06/23/2016] [Indexed: 11/28/2022]
Abstract
![]()
A major challenge in enzymology is
the need to correlate the dynamic
properties of enzymes with, and understand the impact on, their catalytic
cycles. This is especially the case with large, multicenter enzymes
such as the nitric oxide synthases (NOSs), where the importance of
dynamics has been inferred from a variety of structural, single-molecule,
and ensemble spectroscopic approaches but where motions have not been
correlated experimentally with mechanistic steps in the reaction cycle.
Here we take such an approach. Using time-resolved spectroscopy employing
absorbance and Förster resonance energy transfer (FRET) and
exploiting the properties of a flavin analogue (5-deazaflavin mononucleotide
(5-dFMN)) and isotopically labeled nicotinamide coenzymes, we correlate
the timing of CaM structural changes when bound to neuronal nitric
oxide synthase (nNOS) with the nNOS catalytic cycle. We show that
remodeling of CaM occurs early in the electron transfer sequence (FAD
reduction), not at later points in the reaction cycle (e.g., FMN reduction).
Conformational changes are tightly correlated with FAD reduction kinetics
and reflect a transient “opening” and then “closure”
of the bound CaM molecule. We infer that displacement of the C-terminal
tail on binding NADPH and subsequent FAD reduction are the likely
triggers of conformational change. By combining the use of cofactor/coenzyme
analogues and time-resolved FRET/absorbance spectrophotometry, we
show how the reaction cycles of complex enzymes can be simplified,
enabling a detailed study of the relationship between protein dynamics
and reaction cycle chemistry—an approach that can also be used
with other complex multicenter enzymes.
Collapse
Affiliation(s)
- Tobias M. Hedison
- Manchester Synthetic Biology
Research Centre for Fine and Speciality Chemicals (SYNBIOCHEM), Manchester
Institute of Biotechnology, The University of Manchester, Manchester M1 7DN, United Kingdom
| | - Nicole G. H. Leferink
- Manchester Synthetic Biology
Research Centre for Fine and Speciality Chemicals (SYNBIOCHEM), Manchester
Institute of Biotechnology, The University of Manchester, Manchester M1 7DN, United Kingdom
| | - Sam Hay
- Manchester Synthetic Biology
Research Centre for Fine and Speciality Chemicals (SYNBIOCHEM), Manchester
Institute of Biotechnology, The University of Manchester, Manchester M1 7DN, United Kingdom
| | - Nigel S. Scrutton
- Manchester Synthetic Biology
Research Centre for Fine and Speciality Chemicals (SYNBIOCHEM), Manchester
Institute of Biotechnology, The University of Manchester, Manchester M1 7DN, United Kingdom
| |
Collapse
|
20
|
Xiang J, Wang Q, Yiu SM, Man WL, Kwong HK, Lau TC. Aerobic Oxidation of an Osmium(III) N-Hydroxyguanidine Complex To Give Nitric Oxide. Inorg Chem 2016; 55:5056-61. [DOI: 10.1021/acs.inorgchem.6b00652] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Jing Xiang
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong
- College of Chemistry and Environmental Engineering, Yangtze University, Jingzhou, Hubei 434020, People’s Republic of China
| | - Qian Wang
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong
| | - Shek-Man Yiu
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong
| | - Wai-Lun Man
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong
| | - Hoi-Ki Kwong
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong
| | - Tai-Chu Lau
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong
| |
Collapse
|
21
|
Brunel A, Lang J, Couture M, Boucher JL, Dorlet P, Santolini J. Oxygen activation in NO synthases: evidence for a direct role of the substrate. FEBS Open Bio 2016; 6:386-97. [PMID: 27419044 PMCID: PMC4856417 DOI: 10.1002/2211-5463.12036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 12/15/2015] [Accepted: 01/13/2016] [Indexed: 12/13/2022] Open
Abstract
Nitric oxide (NO) and the other reactive nitrogen species (RNOS) play crucial patho‐physiological roles at the interface of oxidative stress and signalling processes. In mammals, the NO synthases (NOSs) are the source of these reactive nitrogen species, and so to understand the precise biological role of RNOS and NO requires elucidation of the molecular functioning of NOS. Oxygen activation, which is at the core of NOS catalysis, involves a sophisticated sequence of electron and proton transfers. While electron transfer in NOS has received much attention, the proton transfer processes has been scarcely investigated. Here, we report an original approach that combines fast‐kinetic techniques coupled to resonance Raman spectroscopy with the use of synthetic analogues of NOS substrate. We characterise FeII‐O2 reaction intermediates in the presence of L‐arginine (Arg), alkyl‐ and aryl‐guanidines. The presence of new reaction intermediates, such as ferric haem‐peroxide, that was formerly postulated, was tracked by analysing the oxygen activation reaction at different times and with different excitation wavelengths. Our results suggest that Arg is not a proton donor, but indirectly intervenes in oxygen activation mechanism by modulating the distal H‐bond network and, in particular, by tuning the position and the role of the distal water molecule. This report supports a catalytic model with two proton transfers in step 1 (Arg hydroxylation) but only one proton transfer in step 2 (Nω‐hydroxy‐L‐arginine oxidation).
Collapse
Affiliation(s)
- Albane Brunel
- Laboratoire Stress Oxydant et Détoxication Institute for Integrative Biology of the Cell (I2BC) CEA, CNRS, Université Paris-Saclay Gif-sur-Yvette Cedex France
| | - Jérôme Lang
- Département de biochimie, de microbiologie et de bio-informatique, and PROTEO Pavillon Charles-Eugène Marchand Université Laval Québec Canada
| | - Manon Couture
- Département de biochimie, de microbiologie et de bio-informatique, and PROTEO Pavillon Charles-Eugène Marchand Université Laval Québec Canada
| | | | - Pierre Dorlet
- Laboratoire Stress Oxydant et Détoxication Institute for Integrative Biology of the Cell (I2BC) CEA, CNRS, Université Paris-Saclay Gif-sur-Yvette Cedex France
| | - Jérôme Santolini
- Laboratoire Stress Oxydant et Détoxication Institute for Integrative Biology of the Cell (I2BC) CEA, CNRS, Université Paris-Saclay Gif-sur-Yvette Cedex France
| |
Collapse
|
22
|
Wang ZQ, Haque MM, Binder K, Sharma M, Wei CC, Stuehr DJ. Engineering nitric oxide synthase chimeras to function as NO dioxygenases. J Inorg Biochem 2016; 158:122-130. [PMID: 27013266 DOI: 10.1016/j.jinorgbio.2016.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 03/10/2016] [Accepted: 03/12/2016] [Indexed: 10/22/2022]
Abstract
Nitric oxide synthases (NOSs) catalyze a two-step oxidation of l-arginine to form nitric oxide (NO) and l-citrulline. NOS contains a N-terminal oxygenase domain (NOSoxy) that is the site of NO synthesis, and a C-terminal reductase domain (NOSred) that binds nicotinamide adenine dinucleotide phosphate (NADPH), flavin adenine dinucleotide (FAD), and flavin mononucleotide (FMN) and provides electrons to the NOSoxy heme during catalysis. The three NOS isoforms in mammals inducible NOS (iNOS), neuronal NOS (nNOS), and endothelial NOS (eNOS) share high structural similarity but differ in NO release rates and catalytic properties due to differences in enzyme kinetic parameters. These parameters must be balanced for NOS enzymes to release NO, rather than consume it in a competing, inherent NO dioxygenase reaction. To improve understanding, we drew on a global catalytic model and previous findings to design three NOS chimeras that may predominantly function as NO dioxygenases: iNOSoxy/nNOSred (Wild type (WT) chimera), V346I iNOSoxy/nNOSred (V346I chimera) and iNOSoxy/S1412D nNOSred (S1412D chimera). The WT and S1412D chimeras had higher NO release than the parent iNOS, while the V346I chimera exhibited much lower NO release, consistent with expectations. Measurements indicated that a greater NO dioxygenase activity was achieved, particularly in the V346I chimera, which dioxygenated an estimated two to four NO per NO that it released, while the other chimeras had nearly equivalent NO dioxygenase and NO release activities. Computer simulations of the global catalytic model using the measured kinetic parameters produced results that mimicked the measured outcomes, and this provided further insights on the catalytic behaviors of the chimeras and basis of their increased NO dioxygenase activities.
Collapse
Affiliation(s)
- Zhi-Qiang Wang
- Department of Chemistry and Biochemistry, Kent State University Geauga, Burton, OH 44021, United States.
| | - Mohammad Mahfuzul Haque
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, United States
| | - Katherine Binder
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, United States
| | - Manisha Sharma
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, United States
| | - Chin-Chuan Wei
- Department of Chemistry, Southern Illinois University Edwardsville, Edwardsville, IL 62026, United States
| | - Dennis J Stuehr
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, United States.
| |
Collapse
|
23
|
Heine CL, Schmidt R, Geckl K, Schrammel A, Gesslbauer B, Schmidt K, Mayer B, Gorren ACF. Selective Irreversible Inhibition of Neuronal and Inducible Nitric-oxide Synthase in the Combined Presence of Hydrogen Sulfide and Nitric Oxide. J Biol Chem 2015; 290:24932-44. [PMID: 26296888 PMCID: PMC4599001 DOI: 10.1074/jbc.m115.660316] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Indexed: 11/06/2022] Open
Abstract
Citrulline formation by both human neuronal nitric-oxide synthase (nNOS) and mouse macrophage inducible NOS was inhibited by the hydrogen sulfide (H2S) donor Na2S with IC50 values of ∼2.4·10−5 and ∼7.9·10−5m, respectively, whereas human endothelial NOS was hardly affected at all. Inhibition of nNOS was not affected by the concentrations of l-arginine (Arg), NADPH, FAD, FMN, tetrahydrobiopterin (BH4), and calmodulin, indicating that H2S does not interfere with substrate or cofactor binding. The IC50 decreased to ∼1.5·10−5m at pH 6.0 and increased to ∼8.3·10−5m at pH 8.0. Preincubation of concentrated nNOS with H2S under turnover conditions decreased activity after dilution by ∼70%, suggesting irreversible inhibition. However, when calmodulin was omitted during preincubation, activity was not affected, suggesting that irreversible inhibition requires both H2S and NO. Likewise, NADPH oxidation was inhibited with an IC50 of ∼1.9·10−5m in the presence of Arg and BH4 but exhibited much higher IC50 values (∼1.0–6.1·10−4m) when Arg and/or BH4 was omitted. Moreover, the relatively weak inhibition of nNOS by Na2S in the absence of Arg and/or BH4 was markedly potentiated by the NO donor 1-(hydroxy-NNO-azoxy)-l-proline, disodium salt (IC50 ∼ 1.3–2.0·10−5m). These results suggest that nNOS and inducible NOS but not endothelial NOS are irreversibly inhibited by H2S/NO at modest concentrations of H2S in a reaction that may allow feedback inhibition of NO production under conditions of excessive NO/H2S formation.
Collapse
Affiliation(s)
| | | | - Kerstin Geckl
- From the Departments of Pharmacology and Toxicology and
| | | | - Bernd Gesslbauer
- Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, Karl Franzens University Graz, A-8010 Graz, Austria
| | - Kurt Schmidt
- From the Departments of Pharmacology and Toxicology and
| | - Bernd Mayer
- From the Departments of Pharmacology and Toxicology and
| | | |
Collapse
|
24
|
Tang W, Li H, Poulos TL, Silverman RB. Mechanistic studies of inactivation of inducible nitric oxide synthase by amidines. Biochemistry 2015; 54:2530-8. [PMID: 25811913 DOI: 10.1021/acs.biochem.5b00135] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nitric oxide synthase (NOS) catalyzes the conversion of L-arginine to L-citrulline and nitric oxide. N(5)-(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.
Collapse
Affiliation(s)
- Wei Tang
- †Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, and Center for Molecular Innovation and Drug Discovery, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208-3113, United States
| | - Huiying Li
- ‡Departments of Molecular Biology and Biochemistry, Chemistry, and Pharmaceutical Sciences, University of California, Irvine, California 92697-3900, United States
| | - Thomas L Poulos
- ‡Departments of Molecular Biology and Biochemistry, Chemistry, and Pharmaceutical Sciences, University of California, Irvine, California 92697-3900, United States
| | - Richard B Silverman
- †Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, and Center for Molecular Innovation and Drug Discovery, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208-3113, United States
| |
Collapse
|
25
|
Leferink NGH, Hay S, Rigby SEJ, Scrutton NS. Towards the free energy landscape for catalysis in mammalian nitric oxide synthases. FEBS J 2014; 282:3016-29. [PMID: 25491181 DOI: 10.1111/febs.13171] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Revised: 12/05/2014] [Accepted: 12/05/2014] [Indexed: 01/30/2023]
Abstract
The general requirement for conformational sampling in biological electron transfer reactions catalysed by multi-domain redox systems has been emphasized in recent years. Crucially, we lack insight into the extent of the conformational space explored and the nature of the energy landscapes associated with these reactions. The nitric oxide synthases (NOS) produce the signalling molecule NO through a series of complex electron transfer reactions. There is accumulating evidence that protein domain dynamics and calmodulin binding are implicated in regulating electron flow from NADPH, through the FAD and FMN cofactors, to the haem oxygenase domain, where NO is generated. Simple models based on static crystal structures of the isolated reductase domain have suggested a role for large-scale motions of the FMN-binding domain in shuttling electrons from the reductase domain to the oxygenase domain. However, detailed insight into the higher-order domain architecture and dynamic structural transitions in NOS enzymes during enzyme turnover is lacking. In this review, we discuss the recent advances made towards mapping the catalytic free energy landscapes of NOS enzymes through integration of both structural techniques (e.g. cryo-electron microscopy) and biophysical techniques (e.g. pulsed-electron paramagnetic resonance). The general picture that emerges from these experiments is that NOS enzymes exist in an equilibrium of conformations, comprising a 'rugged' or 'frustrated' energy landscape, with a key regulatory role for calmodulin in driving vectorial electron transfer by altering the conformational equilibrium. A detailed understanding of these landscapes may provide new opportunities for discovery of isoform-specific inhibitors that bind at the dynamic interfaces of these multi-dimensional energy landscapes.
Collapse
Affiliation(s)
- Nicole G H Leferink
- Manchester Institute of Biotechnology and Faculty of Life Sciences, The University of Manchester, UK
| | - Sam Hay
- Manchester Institute of Biotechnology and Faculty of Life Sciences, The University of Manchester, UK
| | - Stephen E J Rigby
- Manchester Institute of Biotechnology and Faculty of Life Sciences, The University of Manchester, UK
| | - Nigel S Scrutton
- Manchester Institute of Biotechnology and Faculty of Life Sciences, The University of Manchester, UK
| |
Collapse
|
26
|
Das A, Gopalakrishnan B, Druhan LJ, Wang TY, De Pascali F, Rockenbauer A, Racoma I, Varadharaj S, Zweier JL, Cardounel AJ, Villamena FA. Reversal of SIN-1-induced eNOS dysfunction by the spin trap, DMPO, in bovine aortic endothelial cells via eNOS phosphorylation. Br J Pharmacol 2014; 171:2321-34. [PMID: 24405159 DOI: 10.1111/bph.12572] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 12/03/2013] [Accepted: 12/18/2013] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Nitric oxide (NO) derived from eNOS is mostly responsible for the maintenance of vascular homeostasis and its decreased bioavailability is characteristic of reactive oxygen species (ROS)-induced endothelial dysfunction (ED). Because 5,5-dimethyl-1-pyrroline-N-oxide (DMPO), a commonly used spin trap, can control intracellular nitroso-redox balance by scavenging ROS and donating NO, it was employed as a cardioprotective agent against ED but the mechanism of its protection is still not clear. This study elucidated the mechanism of protection by DMPO against SIN-1-induced oxidative injury to bovine aortic endothelial cells (BAEC). EXPERIMENTAL APPROACH BAEC were treated with SIN-1, as a source of peroxynitrite anion (ONOO⁻), and then incubated with DMPO. Cytotoxicity following SIN-1 alone and cytoprotection by adding DMPO was assessed by MTT assay. Levels of ROS and NO generation from HEK293 cells transfected with wild-type and mutant eNOS cDNAs, tetrahydrobiopterin bioavailability, eNOS activity, eNOS and Akt kinase phosphorylation were measured. KEY RESULTS Post-treatment of cells with DMPO attenuated SIN-1-mediated cytotoxicity and ROS generation, restoration of NO levels via increased in eNOS activity and phospho-eNOS levels. Treatment with DMPO alone significantly increased NO levels and induced phosphorylation of eNOS Ser¹¹⁷⁹ via Akt kinase. Transfection studies with wild-type and mutant human eNOS confirmed the dual role of eNOS as a producer of superoxide anion (O₂⁻) with SIN-1 treatment, and a producer of NO in the presence of DMPO. CONCLUSION AND IMPLICATIONS Post-treatment with DMPO of oxidatively challenged cells reversed eNOS dysfunction and could have pharmacological implications in the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Amlan Das
- Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Sharma MC. Comparative pharmacophore modeling and QSAR studies for structural requirements of some substituted 2-aminopyridines derivatives as inhibitors nitric oxide synthases. Interdiscip Sci 2014. [PMID: 25183347 DOI: 10.1007/s12539-013-0038-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Revised: 10/06/2013] [Accepted: 10/14/2014] [Indexed: 06/03/2023]
Abstract
The present studies are an attempt in this direction seeking for the development and comparison of QSAR models of substituted 2-aminopyridines derivatives as inhibitors of nitric oxide synthases by different feature selection methods. Comparing the two different feature selection methods, it is implicit that the model built with the selected variables by simulated annealing (SA) method gives better prediction in case of 2D and 3D QSAR modeling. The QSAR study was carried out on V-life Molecular Design Suite software and the derived best QSAR model was derived by partial component regression (PCR) method. The statistically significant best model with high correlation coefficient (r2 = 0.8408) was selected for further study. The model was further validated by means of crossed squared correlation coefficient (q2 = 0.7270 and pred r2 = 0.7889) which shows model has good predictive ability. 3D-QSAR analysis has been performed on a series of substituted 2-aminopyridines derivatives as which were screened as inhibitors of nitric oxide synthases, using the simulated annealing and step wise k-nearest neighbour Molecular Field Analysis. The best QSAR model showed q2 = 0.8377, r2 = 0.8739 and standard error = 0.1954. It was observed that steric properties predicted by k-nearest neighbour MFA contours can be related to inhibitors of nitric oxide synthases. The predictive ability of the resultant model was evaluated using a test set molecules and the predicted r2 = 0.8159. The distances between the pharmacophore sites were measured in order to confirm their significance to the activities. The results reveal that the acceptor (acc), donor (don), aliphatic and aromatic pharmacophore properties are favorable contours sites for both the activities. The two dimensional and k-nearest neighbour contour plots required for further understanding of the relationship between structural features of substituted 2-aminopyridines derivatives and their activities which should be applicable to design newer potential inducible nitric oxide synthases.
Collapse
Affiliation(s)
- Mukesh C Sharma
- Drug Design and Development Laboratory, School of Pharmacy, Devi Ahilya University, Takshila Campus, Khandwa Road, Indore, 452 001, India,
| |
Collapse
|
28
|
Proteomic analysis of metacyclogenesis in Leishmania infantum wild-type and PTR1 null mutant. EUPA OPEN PROTEOMICS 2014. [DOI: 10.1016/j.euprot.2014.07.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
29
|
Heine C, Kolesnik B, Schmidt R, Werner ER, Mayer B, Gorren ACF. Interaction between neuronal nitric-oxide synthase and tetrahydrobiopterin revisited: studies on the nature and mechanism of tight pterin binding. Biochemistry 2014; 53:1284-95. [PMID: 24512289 PMCID: PMC3944803 DOI: 10.1021/bi401307r] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 02/10/2014] [Indexed: 01/06/2023]
Abstract
Recombinant neuronal nitric-oxide synthase (nNOS) expressed in baculovirus-infected Sf9 cells contains approximately 1 equiv of tightly bound tetrahydrobiopterin (BH4) per dimer and binds a second equivalent with a dissociation constant in the 10(-7)-10(-6) M range. Less is known about the pterin-binding properties of nNOS originating from expression systems such as Escherichia coli that do not produce BH4. We determined the binding properties of E. coli-expressed nNOS for BH4 and several inhibitory pterins by monitoring their effects on enzyme activity. E. coli-expressed nNOS as isolated was activated by BH4 monophasically with EC50 ≈ 2 × 10(-7) M, demonstrating a lack of tight pterin binding. However, overnight incubation with BH4 resulted in tight binding of one BH4 per dimer, yielding an enzyme that resembled Sf9-expressed nNOS. Tight pterin binding was also induced by preincubation with 4-amino-tetrahydrobiopterin, but not by 7,8-dihydrobiopterin or 4-amino-dihydrobiopterin, suggesting that tight-binding site formation requires preincubation with a fully reduced pteridine. Kinetic experiments showed that tight-binding site formation takes approximately 10 min with 1 μM BH4 (2 min with 1 μM 4-amino-BH4) at 4 °C. Anaerobic preincubation experiments demonstrated that O2 is not involved in the process. Gel electrophoretic studies suggest that tight-binding site formation is accompanied by an increase in the strength of the NOS dimer. We propose that incubation of pterin-free nNOS with BH4 creates one tight pterin-binding site per dimer, leaving the other site unaffected, in a reaction that involves redox chemistry.
Collapse
Affiliation(s)
- Christian
L. Heine
- Department
of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, A-8010, Graz, Austria
| | - Bernd Kolesnik
- Department
of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, A-8010, Graz, Austria
| | - Renate Schmidt
- Department
of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, A-8010, Graz, Austria
| | - Ernst R. Werner
- Division
of Biological Chemistry, Biocenter, Innsbruck
Medical University, A-6020, Innsbruck, Austria
| | - Bernd Mayer
- Department
of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, A-8010, Graz, Austria
| | - Antonius C. F. Gorren
- Department
of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, A-8010, Graz, Austria
| |
Collapse
|
30
|
Xu X, Wollenberger U, Qian J, Lettau K, Jung C, Liu S. Electrochemically driven biocatalysis of the oxygenase domain of neuronal nitric oxide synthase in indium tin oxide nanoparticles/polyvinyl alcohol nanocomposite. Bioelectrochemistry 2013; 94:7-12. [PMID: 23727770 DOI: 10.1016/j.bioelechem.2013.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 04/02/2013] [Accepted: 04/23/2013] [Indexed: 11/16/2022]
Abstract
Nitric oxide synthase (NOS) plays a critical role in a number of key physiological and pathological processes. Investigation of electron-transfer reactions in NOS would contribute to a better understanding of the nitric oxide (NO) synthesis mechanism. Herein, we describe an electrochemically driven catalytic strategy, using a nanocomposite that consisted of the oxygenase domain of neuronal NOS (D290nNOSoxy), indium tin oxide (ITO) nanoparticles and polyvinyl alcohol (PVA). Fast direct electron transfer between electrodes and D290nNOSoxy was observed with the heterogeneous electron transfer rate constant (ket) of 154.8 ± 0.1s(-1) at the scan rate of 5 Vs(-1). Moreover, the substrate N(ω)-hydroxy-L-arginine (NHA) was used to prove the concept of electrochemically driven biocatalysis of D290nNOSoxy. In the presence of the oxygen cosubstrate and tetrahydrobiopterin (BH4) cofactor, the addition of NHA caused the decreases of both oxidation current at +0.1 V and reduction current at potentials ranging from -0.149 V to -0.549 V vs Ag/AgCl. Thereafter, a series of control experiments such as in the absence of BH4 or D290nNOSoxy were performed. All the results demonstrated that D290nNOSoxy biocatalysis was successfully driven by electrodes in the presence of BH4 and oxygen. This novel bioelectronic system showed potential for further investigation of NOS and biosensor applications.
Collapse
Affiliation(s)
- Xuan Xu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | | | | | | | | | | |
Collapse
|
31
|
Whitsett J, Filho AR, Sethumadhavan S, Celinska J, Widlansky M, Vásquez-Vivar J. Human endothelial dihydrofolate reductase low activity limits vascular tetrahydrobiopterin recycling. Free Radic Biol Med 2013; 63:143-50. [PMID: 23707606 PMCID: PMC3748942 DOI: 10.1016/j.freeradbiomed.2013.04.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/09/2013] [Accepted: 04/27/2013] [Indexed: 11/19/2022]
Abstract
Tetrahydrobiopterin (BH₄) is required for NO synthesis and inhibition of superoxide release from endothelial NO synthase. Clinical trials using BH₄ to treat endothelial dysfunction have produced mixed results. Poor outcomes may be explained by the rapid systemic and cellular oxidation of BH₄. One of the oxidation products of BH₄, 7,8-dihydrobiopterin (7,8-BH₂), is recycled back to BH₄ by dihydrofolate reductase (DHFR). This enzyme is ubiquitously distributed and shows a wide range of activity depending on species-specific factors and cell type. Information about the kinetics and efficiency of BH4 recycling in human endothelial cells receiving BH₄ treatment is lacking. To characterize this reaction, we applied a novel multielectrode coulometric HPLC method that enabled the direct quantification of 7,8-BH₂ and BH₄, which is not possible with fluorescence-based methodologies. We found that basal untreated BH₄ and 7,8-BH₂ concentrations in human endothelial cells (ECs) are lower than in bovine and murine endothelioma cells. Treatment of human ECs with BH₄ transiently increased intracellular BH₄ while accumulating the more stable 7,8-BH₂. This was different from bovine or murine ECs, which resulted in preferential BH₄ increase. Using BH₄ diastereomers, 6S-BH₄ and 6R-BH₄, the narrow contribution of enzymatic DHFR recycling to total intracellular BH₄ was demonstrated. Reduction of 7,8-BH₂ to BH₄ occurs at very slow rates in cells and needs supraphysiological levels of 7,8-BH₂, indicating this reaction is kinetically limited. Activity assays verified that human DHFR has very low affinity for 7,8-BH₂ (DHF7,8-BH₂) and folic acid inhibits 7,8-BH₂ recycling. We conclude that low activity of endothelial DHFR is an important factor limiting the benefits of BH4 therapies, which may be further aggravated by folate supplements.
Collapse
Affiliation(s)
- Jennifer Whitsett
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
- Redox Biology Program, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Artur Rangel Filho
- Department of Pathology, Jackson Memorial Hospital, University of Miami Leonard M. Miller School of Medicine, Miami, Florida 33136
| | | | - Joanna Celinska
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Michael Widlansky
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Jeannette Vásquez-Vivar
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
- Redox Biology Program, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| |
Collapse
|
32
|
Jiang N, Kuznetsov A, Nocek JM, Hoffman BM, Crane BR, Hu X, Beratan DN. Distance-independent charge recombination kinetics in cytochrome c-cytochrome c peroxidase complexes: compensating changes in the electronic coupling and reorganization energies. J Phys Chem B 2013; 117:9129-41. [PMID: 23895339 PMCID: PMC3809023 DOI: 10.1021/jp401551t] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Charge recombination rate constants vary no more than 3-fold for interprotein ET in the Zn-substituted wild type (WT) cytochrome c peroxidase (CcP):cytochrome c (Cc) complex and in complexes with four mutants of the Cc protein (i.e., F82S, F82W, F82Y, and F82I), despite large differences in the ET distance. Theoretical analysis indicates that charge recombination for all complexes involves a combination of tunneling and hopping via Trp191. For three of the five structures (WT and F82S(W)), the protein favors hopping more than that in the other two structures that have longer heme → ZnP distances (F82Y(I)). Experimentally observed biexponential ET kinetics is explained by the complex locking in alternative coupling pathways, where the acceptor hole state is either primarily localized on ZnP (slow phase) or on Trp191 (fast phase). The large conformational differences between the CcP:Cc interface for the F82Y(I) mutants compared to that the WT and F82S(W) complexes are predicted to change the reorganization energies for the CcP:Cc ET reactions because of changes in solvent exposure and interprotein ET distances. Since the recombination reaction is likely to occur in the inverted Marcus regime, an increased reorganization energy compensates the decreased role for hopping recombination (and the longer transfer distance) in the F82Y(I) mutants. Taken together, coupling pathway and reorganization energy effects for the five protein complexes explain the observed insensitivity of recombination kinetics to donor-acceptor distance and docking pose and also reveals how hopping through aromatic residues can accelerate long-range ET.
Collapse
Affiliation(s)
- Nan Jiang
- Department of Chemistry, Duke University, Durham, NC 27708
| | | | - Judith M. Nocek
- Department of Chemistry, Northwestern University, Evanston, IL 60208
| | - Brian M. Hoffman
- Department of Chemistry, Northwestern University, Evanston, IL 60208
| | - Brian R. Crane
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
| | - Xiangqian Hu
- Department of Chemistry, Duke University, Durham, NC 27708
| | - David N. Beratan
- Department of Chemistry, Duke University, Durham, NC 27708
- Department of Biochemistry, Duke University, Durham, NC 27708
- Department of Physics, Duke University, Durham, NC 27708
| |
Collapse
|
33
|
Haque MM, Tejero J, Bayachou M, Wang ZQ, Fadlalla M, Stuehr DJ. Thermodynamic characterization of five key kinetic parameters that define neuronal nitric oxide synthase catalysis. FEBS J 2013; 280:4439-53. [PMID: 23789902 DOI: 10.1111/febs.12404] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 06/14/2013] [Accepted: 06/17/2013] [Indexed: 11/30/2022]
Abstract
NO synthase (NOS) enzymes convert L-arginine to NO in two sequential reactions whose rates (k(cat1) and k(cat2)) are both limited by the rate of ferric heme reduction (k(r)). An enzyme ferric heme-NO complex forms as an immediate product complex and then undergoes either dissociation (at a rate that we denote as k(d)) to release NO in a productive manner, or reduction (k(r)) to form a ferrous heme-NO complex that must react with O2 (at a rate that we denote as k(ox)) in a NO dioxygenase reaction that regenerates the ferric enzyme. The interplay of these five kinetic parameters (k(cat1), k(cat2), k(r), k(d) and k(ox)) determines NOS specific activity, O2 concentration response, and pulsatile versus steady-state NO generation. In the present study, we utilized stopped-flow spectroscopy and single catalytic turnover methods to characterize the individual temperature dependencies of the five kinetic parameters of rat neuronal NOS. We then incorporated the measured kinetic values into computer simulations of the neuronal NOS reaction using a global kinetic model to comprehensively model its temperature-dependent catalytic behaviours. The results obtained provide new mechanistic insights and also reveal that the different temperature dependencies of the five kinetic parameters significantly alter neuronal NOS catalytic behaviours and NO release efficiency as a function of temperature.
Collapse
|
34
|
Charge-pairing interactions control the conformational setpoint and motions of the FMN domain in neuronal nitric oxide synthase. Biochem J 2013; 450:607-17. [PMID: 23289611 DOI: 10.1042/bj20121488] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The NOS (nitric oxide synthase; EC 1.14.13.39) enzymes contain a C-terminal flavoprotein domain [NOSred (reductase domain of NOS)] that binds FAD and FMN, and an N-terminal oxygenase domain that binds haem. Evidence suggests that the FMN-binding domain undergoes large conformational motions to shuttle electrons between the NADPH/FAD-binding domain [FNR (ferredoxin NADP-reductase)] and the oxygenase domain. Previously we have shown that three residues on the FMN domain (Glu762, Glu816 and Glu819) that make charge-pairing interactions with the FNR help to slow electron flux through nNOSred (neuronal NOSred). In the present study, we show that charge neutralization or reversal at each of these residues alters the setpoint [Keq(A)] of the NOSred conformational equilibrium to favour the open (FMN-deshielded) conformational state. Moreover, computer simulations of the kinetic traces of cytochrome c reduction by the mutants suggest that they have higher conformational transition rates (1.5-4-fold) and rates of interflavin electron transfer (1.5-2-fold) relative to wild-type nNOSred. We conclude that the three charge-pairing residues on the FMN domain govern electron flux through nNOSred by stabilizing its closed (FMN-shielded) conformational state and by retarding the rate of conformational switching between its open and closed conformations.
Collapse
|
35
|
Tejero J, Stuehr D. Tetrahydrobiopterin in nitric oxide synthase. IUBMB Life 2013; 65:358-65. [PMID: 23441062 DOI: 10.1002/iub.1136] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 12/25/2012] [Indexed: 11/10/2022]
Abstract
SUMMARY Nitric oxide synthase (NOS) is a critical enzyme for the production of the messenger molecule nitric oxide (NO) from L-arginine. NOS enzymes require tetrahydrobiopterin as a cofactor for NO synthesis. Besides being one of the few enzymes to use this cofactor, the role of tetrahydrobiopterin in NOS catalytic mechanism is different from other enzymes: during the catalytic cycle of NOS, tetrahydrobiopterin forms a radical species that is again reduced, thus effectively regenerating after each NO synthesis cycle. In this review, we summarize our current knowledge about the role of tetrahydrobiopterin in the structure, function, and catalytic mechanism of NOS enzymes.
Collapse
Affiliation(s)
- Jesús Tejero
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | |
Collapse
|
36
|
Pharmacological approaches to the treatment of oxidative stress-induced cardiovascular dysfunctions. Future Med Chem 2013; 5:465-78. [DOI: 10.4155/fmc.13.15] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Cardiovascular diseases are a growing major global health problem. Our understanding of the mechanisms of pathophysiology of cardiovascular diseases has been gaining significant advances and a wealth of knowledge implicates oxidative stress as a key causative agent. However, to date, most efforts to treat heart failure using conventional antioxidant therapies have been less than encouraging. With increasing incidences of cardiovascular disease in young as well as in aging populations, and the problem of long-term diminishing efficacy of conventional therapeutics, the need for new treatments has never been greater. In this review, a variety of therapeutic targets and compounds applied to treat cardiovascular diseases via inhibition of oxidative stress are presented.
Collapse
|
37
|
Sabat J, Egawa T, Lu C, Stuehr DJ, Gerfen GJ, Rousseau DL, Yeh SR. Catalytic intermediates of inducible nitric-oxide synthase stabilized by the W188H mutation. J Biol Chem 2013; 288:6095-106. [PMID: 23269673 PMCID: PMC3816742 DOI: 10.1074/jbc.m112.403238] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 12/12/2012] [Indexed: 11/06/2022] Open
Abstract
Nitric-oxide synthase (NOS) catalyzes nitric oxide (NO) synthesis via a two-step process: L-arginine (L-Arg) → N-hydroxy-L-arginine → citrulline + NO. In the active site the heme is coordinated by a thiolate ligand, which accepts a H-bond from a nearby tryptophan residue, Trp-188. Mutation of Trp-188 to histidine in murine inducible NOS was shown to retard NO synthesis and allow for transient accumulation of a new intermediate with a Soret maximum at 420 nm during the L-Arg hydroxylation reaction (Tejero, J., Biswas, A., Wang, Z. Q., Page, R. C., Haque, M. M., Hemann, C., Zweier, J. L., Misra, S., and Stuehr, D. J. (2008) J. Biol. Chem. 283, 33498-33507). However, crystallographic data showed that the mutation did not perturb the overall structure of the enzyme. To understand how the proximal mutation affects the oxygen chemistry, we carried out biophysical studies of the W188H mutant. Our stopped-flow data showed that the 420-nm intermediate was not only populated during the L-Arg reaction but also during the N-hydroxy-L-arginine reaction. Spectroscopic data and structural analysis demonstrated that the 420-nm intermediate is a hydroxide-bound ferric heme species that is stabilized by an out-of-plane distortion of the heme macrocycle and a cation radical centered on the tetrahydrobiopterin cofactor. The current data add important new insights into the previously proposed catalytic mechanism of NOS (Li, D., Kabir, M., Stuehr, D. J., Rousseau, D. L., and Yeh, S. R. (2007) J. Am. Chem. Soc. 129, 6943-6951).
Collapse
Affiliation(s)
- Joseph Sabat
- From the Department of Physiology and Biophysics, Albert
Einstein College of Medicine, Bronx, New York 10461 and
| | - Tsuyoshi Egawa
- From the Department of Physiology and Biophysics, Albert
Einstein College of Medicine, Bronx, New York 10461 and
| | - Changyuan Lu
- From the Department of Physiology and Biophysics, Albert
Einstein College of Medicine, Bronx, New York 10461 and
| | - Dennis J. Stuehr
- the Department of Immunology, Lerner Research Institute,
Cleveland Clinic, Cleveland, Ohio 44195
| | - Gary J. Gerfen
- From the Department of Physiology and Biophysics, Albert
Einstein College of Medicine, Bronx, New York 10461 and
| | - Denis L. Rousseau
- From the Department of Physiology and Biophysics, Albert
Einstein College of Medicine, Bronx, New York 10461 and
| | - Syun-Ru Yeh
- From the Department of Physiology and Biophysics, Albert
Einstein College of Medicine, Bronx, New York 10461 and
| |
Collapse
|
38
|
Inui Y, Shiro M, Kusukawa T, Fukuzumi S, Kojima T. A triangular prismatic hexanuclear iridium(III) complex bridged by flavin analogues showing reversible redox processes. Dalton Trans 2013; 42:2773-8. [PMID: 23235491 DOI: 10.1039/c2dt32535g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
[Ir(6)(μ-alloCl(2)(2-))(3)(Cp*)(6)(OH)(3)](PF(6))(3) (1) having 7,8-dichloroalloxazine dianion (alloCl(2)(2-)) as bridging ligands was synthesized and characterized by X-ray crystallography, spectroscopic and electrochemical measurements. The alloxazine ligands showed unprecedented coordination modes to link the six Ir(III) centres. The complex exhibited remarkable stability and reversible six-electron redox processes at the bridging alloxazine ligands in organic solvents. The first reversible reduction process occurred on each of three alloxazine ligands in 1 to produce a three-electron-reduced species, [Ir(III)(6)Cp*(6)(μ-alloCl(2)˙(3-))(3)(OH)(3)], and was observed as an apparent one-step reduction process at -0.65 V (vs. Fc(0/+)). The second reversible reduction process on each of the three alloxazine ligands in 1 was recorded at almost the same potential, -0.78 V (vs. Fc(0/+)), to afford the six-electron-reduced form, [Ir(III)(6)Cp*(6)(μ-alloCl(2)(4-))(3)(OH)(3)](3-). The radical anion of the alloxazine derivative was detected by EPR measurements at room temperature. After the six-electron reduction of 1 with cobaltocene, the backward oxidation processes of reduced forms with p-chloranil were traced by UV-Vis spectroscopy to confirm the recovery of the original spectrum of 1.
Collapse
Affiliation(s)
- Yuji Inui
- Department of Material and Life Science, Graduate School of Engineering, Osaka University, and ALCA (JST), 2-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | | | | | |
Collapse
|
39
|
Leach AG, Olsson LL, Warner DJ. A monomeric form of iNOS can rationalise observed SAR for inhibitors of dimerisation: quantum mechanics and docking compared. MEDCHEMCOMM 2013. [DOI: 10.1039/c2md20159c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Many interaction types bind iNOS and inhibitors of its dimerization: docking methods reproduce geometries and quantum mechanics lipophilicity ligand efficiencies.
Collapse
|
40
|
Baydar M, Capan Z, Girgin G, Palabiyik SS, Sahin G, Fuchs D, Baydar T. Evaluation of tetrahydrobiopterin pathway in operating room workers: changes in biopterin status and tryptophan metabolism. BULLETIN OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2012; 89:1125-8. [PMID: 23052583 DOI: 10.1007/s00128-012-0845-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 09/24/2012] [Indexed: 06/01/2023]
Abstract
The aim of the study was to evaluate the effect of anesthetics as operating room contaminants on tetrahydrobiopterin pathway in 40 operating room personnel and 30 healthy controls by measuring biopterin, dihydrobiopterin reductase, tryptophan, kynurenine and serotonin. Biopterin concentrations were 124 ± 12.3 µmol/mol creatinine in workers and 88 ± 5.7 µmol/mol creatinine in controls whereas kynurenine concentrations were 1.75 ± 0.09 µM and 1.95 ± 0.06 µM, respectively (both, p < 0.05). It can be claimed that enhanced biopterin and diminished kynurenine levels may play a triggering role in disruption of metabolic events in operating room personnel.
Collapse
Affiliation(s)
- Mustafa Baydar
- Clinic of Anesthesiology, Numune Hospital, Ankara, Turkey
| | | | | | | | | | | | | |
Collapse
|
41
|
Iyanagi T, Xia C, Kim JJP. NADPH-cytochrome P450 oxidoreductase: prototypic member of the diflavin reductase family. Arch Biochem Biophys 2012; 528:72-89. [PMID: 22982532 PMCID: PMC3606592 DOI: 10.1016/j.abb.2012.09.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Revised: 09/01/2012] [Accepted: 09/03/2012] [Indexed: 12/31/2022]
Abstract
NADPH-cytochrome P450 oxidoreductase (CYPOR) and nitric oxide synthase (NOS), two members of the diflavin oxidoreductase family, are multi-domain enzymes containing distinct FAD and FMN domains connected by a flexible hinge. FAD accepts a hydride ion from NADPH, and reduced FAD donates electrons to FMN, which in turn transfers electrons to the heme center of cytochrome P450 or NOS oxygenase domain. Structural analysis of CYPOR, the prototype of this enzyme family, has revealed the exact nature of the domain arrangement and the role of residues involved in cofactor binding. Recent structural and biophysical studies of CYPOR have shown that the two flavin domains undergo large domain movements during catalysis. NOS isoforms contain additional regulatory elements within the reductase domain that control electron transfer through Ca(2+)-dependent calmodulin (CaM) binding. The recent crystal structure of an iNOS Ca(2+)/CaM-FMN construct, containing the FMN domain in complex with Ca(2+)/CaM, provided structural information on the linkage between the reductase and oxgenase domains of NOS, making it possible to model the holo iNOS structure. This review summarizes recent advances in our understanding of the dynamics of domain movements during CYPOR catalysis and the role of the NOS diflavin reductase domain in the regulation of NOS isozyme activities.
Collapse
Affiliation(s)
- Takashi Iyanagi
- Department of Biochemistry, Medical College of Wisconsin, USA
- Department of Life Science, The Himeji Institute of Technology, University of Hyogo, Japan
| | - Chuanwu Xia
- Department of Biochemistry, Medical College of Wisconsin, USA
| | - Jung-Ja P. Kim
- Department of Biochemistry, Medical College of Wisconsin, USA
| |
Collapse
|
42
|
Potential implication of the chemical properties and bioactivity of nitrone spin traps for therapeutics. Future Med Chem 2012; 4:1171-207. [PMID: 22709256 DOI: 10.4155/fmc.12.74] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Nitrone therapeutics has been employed in the treatment of oxidative stress-related diseases such as neurodegeneration, cardiovascular disease and cancer. The nitrone-based compound NXY-059, which is the first drug to reach clinical trials for the treatment of acute ischemic stroke, has provided promise for the development of more robust pharmacological agents. However, the specific mechanism of nitrone bioactivity remains unclear. In this review, we present a variety of nitrone chemistry and biological activity that could be implicated for the nitrone's pharmacological activity. The chemistries of spin trapping and spin adduct reveal insights on the possible roles of nitrones for altering cellular redox status through radical scavenging or nitric oxide donation, and their biological effects are presented. An interdisciplinary approach towards the development of novel synthetic antioxidants with improved pharmacological properties encompassing theoretical, synthetic, biochemical and in vitro/in vivo studies is covered.
Collapse
|
43
|
Haque MM, Fadlalla MA, Aulak KS, Ghosh A, Durra D, Stuehr DJ. Control of electron transfer and catalysis in neuronal nitric-oxide synthase (nNOS) by a hinge connecting its FMN and FAD-NADPH domains. J Biol Chem 2012; 287:30105-16. [PMID: 22722929 PMCID: PMC3436266 DOI: 10.1074/jbc.m112.339697] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 06/13/2012] [Indexed: 01/19/2023] Open
Abstract
In nitric-oxide synthases (NOSs), two flexible hinges connect the FMN domain to the rest of the enzyme and may guide its interactions with partner domains for electron transfer and catalysis. We investigated the role of the FMN-FAD/NADPH hinge in rat neuronal NOS (nNOS) by constructing mutants that either shortened or lengthened this hinge by 2, 4, and 6 residues. Shortening the hinge progressively inhibited electron flux through the calmodulin (CaM)-free and CaM-bound nNOS to cytochrome c, whereas hinge lengthening relieved repression of electron flux in CaM-free nNOS and had no impact or slowed electron flux through CaM-bound nNOS to cytochrome c. How hinge length influenced heme reduction depended on whether enzyme flavins were pre-reduced with NADPH prior to triggering heme reduction. Without pre-reduction, changing the hinge length was deleterious; with pre-reduction, the hinge shortening was deleterious, and hinge lengthening increased heme reduction rates beyond wild type. Flavin fluorescence and stopped-flow kinetic studies on CaM-bound enzymes suggested hinge lengthening slowed the domain-domain interaction needed for FMN reduction. All hinge length changes lowered NO synthesis activity and increased uncoupled NADPH consumption. We conclude that several aspects of catalysis are sensitive to FMN-FAD/NADPH hinge length and that the native hinge allows a best compromise among the FMN domain interactions and associated electron transfer events to maximize NO synthesis and minimize uncoupled NADPH consumption.
Collapse
Affiliation(s)
- Mohammad Mahfuzul Haque
- From the Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Mohammed A. Fadlalla
- From the Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Kulwant S. Aulak
- From the Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Arnab Ghosh
- From the Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Deborah Durra
- From the Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Dennis J. Stuehr
- From the Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| |
Collapse
|
44
|
Brunel A, Santolini J, Dorlet P. Electron paramagnetic resonance characterization of tetrahydrobiopterin radical formation in bacterial nitric oxide synthase compared to mammalian nitric oxide synthase. Biophys J 2012; 103:109-17. [PMID: 22828337 PMCID: PMC3388219 DOI: 10.1016/j.bpj.2012.05.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 05/21/2012] [Accepted: 05/22/2012] [Indexed: 11/24/2022] Open
Abstract
H(4)B is an essential catalytic cofactor of the mNOSs. It acts as an electron donor and activates the ferrous heme-oxygen complex intermediate during Arg oxidation (first step) and NOHA oxidation (second step) leading to nitric oxide and citrulline as final products. However, its role as a proton donor is still debated. Furthermore, its exact involvement has never been explored for other NOSs such as NOS-like proteins from bacteria. This article proposes a comparative study of the role of H(4)B between iNOS and bsNOS. In this work, we have used freeze-quench to stop the arginine and NOHA oxidation reactions and trap reaction intermediates. We have characterized these intermediates using multifrequency electron paramagnetic resonance. For the first time, to our knowledge, we report a radical formation for a nonmammalian NOS. The results indicate that bsNOS, like iNOS, has the capacity to generate a pterin radical during Arg oxidation. Our current electron paramagnetic resonance data suggest that this radical is protonated indicating that H(4)B may not transfer any proton. In the 2nd step, the radical trapped for iNOS is also suggested to be protonated as in the 1st step, whereas it was not possible to trap a radical for the bsNOS 2nd step. Our data highlight potential differences for the catalytic mechanism of NOHA oxidation between mammalian and bacterial NOSs.
Collapse
Key Words
- arg, l-arginine
- epr, electron paramagnetic resonance
- feiino, ferrous heme-nitrosyl complex
- feiio2, ferrous heme-oxygen complex
- feiiino, ferric heme-nitrosyl complex
- h4b, (6r)-5,6,7,8-tetrahydro-l-biopterin
- hs-5c, high-spin hexacoordinated iron
- no, nitric oxide
- noha, nω-hydroxy-l-arginine
- nos, nitric oxide synthase
- nosoxy, oxygenase domain of nos
- bacnos, bacterial nos-like proteins
- enos, endothelial nitric oxide synthase
- inos, inducible nitric oxide synthase
- mnos, mammalian nitric oxide synthase
- nnos, neuronal nitric oxide synthase
- bsnos, nos–like protein isolated from bacillus subtilis
- cpet, concerted proton electron transfer
Collapse
Affiliation(s)
| | | | - Pierre Dorlet
- CNRS, Laboratoire Stress Oxydant et Détoxication, Gif-sur-Yvette, France and CEA, iBiTec-S, Gif-sur-Yvette, France
| |
Collapse
|
45
|
Inui Y, Miyazaki S, Ohkubo K, Fukuzumi S, Kojima T. Regulation of Redox Potential of a Pterin Derivative Bound to a Ruthenium(II) Complex by Intermolecular Hydrogen Bonding with Nucleobases. Angew Chem Int Ed Engl 2012. [DOI: 10.1002/ange.201108827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
46
|
Biondi R, Ambrosio G, De Pascali F, Tritto I, Capodicasa E, Druhan LJ, Hemann C, Zweier JL. HPLC analysis of tetrahydrobiopterin and its pteridine derivatives using sequential electrochemical and fluorimetric detection: application to tetrahydrobiopterin autoxidation and chemical oxidation. Arch Biochem Biophys 2012; 520:7-16. [PMID: 22286026 PMCID: PMC3307828 DOI: 10.1016/j.abb.2012.01.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 01/03/2012] [Accepted: 01/10/2012] [Indexed: 02/07/2023]
Abstract
Tetrahydrobiopterin (BH(4)) is an essential cofactor of endothelial nitric oxide (NO) synthase and when depleted, endothelial dysfunction results with decreased production of NO. BH(4) is also an anti-oxidant being a good "scavenger" of oxidative species. NADPH oxidase, xanthine oxidase, and mitochondrial enzymes producing reactive oxygen species (ROS) can induce elevated oxidant stress and cause BH(4) oxidation and subsequent decrease in NO production and bioavailability. In order to define the process of ROS-mediated BH(4) degradation, a sensitive method for monitoring pteridine redox-state changes is required. Considering that the conventional fluorescence method is an indirect method requiring conversion of all pteridines to oxidized forms, it would be beneficial to use a rapid quantitative assay for the individual detection of BH(4) and its related pteridine metabolites. To study, in detail, the BH(4) oxidative pathways, a rapid direct sensitive HPLC assay of BH(4) and its pteridine derivatives was adapted using sequential electrochemical and fluorimetric detection. We examined BH(4) autoxidation, hydrogen peroxide- and superoxide-driven oxidation, and Fenton reaction hydroxyl radical-driven BH(4) transformation. We demonstrate that the formation of the primary two-electron oxidation product, dihydrobiopterin (BH(2)), predominates with oxygen-induced BH(4) autoxidation and superoxide-catalyzed oxidation, while the irreversible metabolites, pterin and dihydroxanthopterin (XH(2)), are largely produced during hydroxyl radical-driven BH(4) oxidation.
Collapse
Affiliation(s)
- Roberto Biondi
- Division of Cardiology, University of Perugia School of Medicine, 06100 Perugia
- Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, College of Medicine, Ohio State University, Columbus, OH 43210
- Azienda Ospedaliera “S.Maria” Terni
- Dipartimento di Medicina Clinica Sperimentale, University of Perugia School of Medicine, 06100 Perugia
| | - Giuseppe Ambrosio
- Division of Cardiology, University of Perugia School of Medicine, 06100 Perugia
- Dipartimento di Medicina Clinica Sperimentale, University of Perugia School of Medicine, 06100 Perugia
| | - Francesco De Pascali
- Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, College of Medicine, Ohio State University, Columbus, OH 43210
| | - Isabella Tritto
- Division of Cardiology, University of Perugia School of Medicine, 06100 Perugia
- Dipartimento di Medicina Clinica Sperimentale, University of Perugia School of Medicine, 06100 Perugia
| | - Enrico Capodicasa
- Dipartimento di Medicina Clinica Sperimentale, University of Perugia School of Medicine, 06100 Perugia
| | - Lawrence J. Druhan
- Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, College of Medicine, Ohio State University, Columbus, OH 43210
- Department of Anesthesiology, College of Medicine, Ohio State University, Columbus, OH 43210
| | - Craig Hemann
- Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, College of Medicine, Ohio State University, Columbus, OH 43210
| | - Jay L. Zweier
- Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, College of Medicine, Ohio State University, Columbus, OH 43210
| |
Collapse
|
47
|
Inui Y, Miyazaki S, Ohkubo K, Fukuzumi S, Kojima T. Regulation of Redox Potential of a Pterin Derivative Bound to a Ruthenium(II) Complex by Intermolecular Hydrogen Bonding with Nucleobases. Angew Chem Int Ed Engl 2012; 51:4623-7. [DOI: 10.1002/anie.201108827] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Indexed: 11/10/2022]
|
48
|
Oxygen activation in neuronal NO synthase: resolving the consecutive mono-oxygenation steps. Biochem J 2012; 443:505-14. [PMID: 22300432 DOI: 10.1042/bj20111644] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The vital signalling molecule NO is produced by mammalian NOS (nitric oxide synthase) enzymes in two steps. L-arginine is converted into NOHA (Nω-hydroxy-L-arginine), which is converted into NO and citrulline. Both steps are thought to proceed via similar mechanisms in which the cofactor BH4 (tetrahydrobiopterin) activates dioxygen at the haem site by electron transfer. The subsequent events are poorly understood due to the lack of stable intermediates. By analogy with cytochrome P450, a haem-iron oxo species may be formed, or direct reaction between a haem-peroxy intermediate and substrate may occur. The two steps may also occur via different mechanisms. In the present paper we analyse the two reaction steps using the G586S mutant of nNOS (neuronal NOS), which introduces an additional hydrogen bond in the active site and provides an additional proton source. In the mutant enzyme, BH4 activates dioxygen as in the wild-type enzyme, but an interesting intermediate haem species is then observed. This may be a stabilized form of the active oxygenating species. The mutant is able to perform step 2 (reaction with NOHA), but not step 1 (with L-arginine) indicating that the extra hydrogen bond enables it to discriminate between the two mono-oxygenation steps. This implies that the two steps follow different chemical mechanisms.
Collapse
|
49
|
Wang ZQ, Tejero J, Wei CC, Haque MM, Santolini J, Fadlalla M, Biswas A, Stuehr DJ. Arg375 tunes tetrahydrobiopterin functions and modulates catalysis by inducible nitric oxide synthase. J Inorg Biochem 2012; 108:203-15. [PMID: 22173094 PMCID: PMC3306459 DOI: 10.1016/j.jinorgbio.2011.11.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2011] [Revised: 11/12/2011] [Accepted: 11/14/2011] [Indexed: 10/15/2022]
Abstract
NO synthase enzymes (NOS) support unique single-electron transitions of a bound H(4)B cofactor during catalysis. Previous studies showed that both the pterin structure and surrounding protein residues impact H(4)B redox function during catalysis. A conserved Arg residue (Arg375 in iNOS) forms hydrogen bonds with the H(4)B ring. In order to understand the role of this residue in modulating the function of H(4)B and overall NO synthesis of the enzyme, we generated and characterized three mutants R375D, R375K and R375N of the oxygenase domain of inducible NOS (iNOSoxy). The mutations affected the dimer stability of iNOSoxy and its binding affinity toward substrates and H(4)B to varying degrees. Optical spectra of the ferric, ferrous, ferrous dioxy, ferrous-NO, ferric-NO, and ferrous-CO forms of each mutant were similar to the wild-type. However, mutants displayed somewhat lower heme midpoint potentials and faster ferrous heme-NO complex reactivity with O(2). Unlike the wild-type protein, mutants could not oxidize NOHA to nitrite in a H(2)O(2)-driven reaction. Mutation could potentially change the ferrous dioxy decay rate, H(4)B radical formation rate, and the amount of the Arg hydroxylation during single turnover Arg hydroxylation reaction. All mutants were able to form heterodimers with the iNOS G450A full-length protein and displayed lower NO synthesis activities and uncoupled NADPH consumption. We conclude that the conserved residue Arg375 (1) regulates the tempo and extent of the electron transfer between H(4)B and ferrous dioxy species and (2) controls the reactivity of the heme-based oxidant formed after electron transfer from H(4)B during steady state NO synthesis and H(2)O(2)-driven NOHA oxidation. Thus, Arg375 modulates the redox function of H(4)B and is important in controlling the catalytic function of NOS enzymes.
Collapse
Affiliation(s)
- Zhi-Qiang Wang
- Department of Chemistry and Biochemistry, Kent State University at Tuscarawas, New Philadelphia, Ohio, 44663
| | - Jesús Tejero
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| | - Chin-Chuan Wei
- Department of Chemistry, Southern Illinois University Edwardsville, Edwardsville, IL, 62026
| | - Mohammad Mahfuzul Haque
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| | - Jerome Santolini
- iBiTec-S; LSOD, C. E. A. Saclay; 91191 Gif-sur-Yvette Cedex, France
| | - Mohammed Fadlalla
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| | - Ashis Biswas
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| | - Dennis J. Stuehr
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| |
Collapse
|
50
|
Feng C. Mechanism of Nitric Oxide Synthase Regulation: Electron Transfer and Interdomain Interactions. Coord Chem Rev 2012; 256:393-411. [PMID: 22523434 PMCID: PMC3328867 DOI: 10.1016/j.ccr.2011.10.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Nitric oxide synthase (NOS), a flavo-hemoprotein, tightly regulates nitric oxide (NO) synthesis and thereby its dual biological activities as a key signaling molecule for vasodilatation and neurotransmission at low concentrations, and also as a defensive cytotoxin at higher concentrations. Three NOS isoforms, iNOS, eNOS and nNOS (inducible, endothelial, and neuronal NOS), achieve their key biological functions by tight regulation of interdomain electron transfer (IET) process via interdomain interactions. In particular, the FMN-heme IET is essential in coupling electron transfer in the reductase domain with NO synthesis in the heme domain by delivery of electrons required for O(2) activation at the catalytic heme site. Compelling evidence indicates that calmodulin (CaM) activates NO synthesis in eNOS and nNOS through a conformational change of the FMN domain from its shielded electron-accepting (input) state to a new electron-donating (output) state, and that CaM is also required for proper alignment of the domains. Another exciting recent development in NOS enzymology is the discovery of importance of the the FMN domain motions in modulating reactivity and structure of the catalytic heme active site (in addition to the primary role of controlling the IET processes). In the absence of a structure of full-length NOS, an integrated approach of spectroscopic (e.g. pulsed EPR, MCD, resonance Raman), rapid kinetics (laser flash photolysis and stopped flow) and mutagenesis methods is critical to unravel the molecular details of the interdomain FMN/heme interactions. This is to investigate the roles of dynamic conformational changes of the FMN domain and the docking between the primary functional FMN and heme domains in regulating NOS activity. The recent developments in understanding of mechanisms of the NOS regulation that are driven by the combined approach are the focuses of this review. An improved understanding of the role of interdomain FMN/heme interaction and CaM binding may serve as the basis for the design of new selective inhibitors of NOS isoforms.
Collapse
Affiliation(s)
- Changjian Feng
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131 (USA) , Tel: 505-925-4326
| |
Collapse
|