1
|
Maia Santos Urbancg Moncorvo F, Avendaño Leon OL, Curti C, Kabri Y, Redon S, Torres-Santos EC, Vanelle P. Enhancing Antileishmanial Activity of Amidoxime-Based Compounds Bearing a 4,5-Dihydrofuran Scaffold: In Vitro Screening Against Leishmania amazonensis. Molecules 2024; 29:5469. [PMID: 39598858 PMCID: PMC11597885 DOI: 10.3390/molecules29225469] [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: 10/21/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
Leishmaniasis, a protozoan disease affecting humans, exposes significant shortcomings in current treatments. In continuation to our previous findings on amidoxime-based antileishmanial compounds bearing a 4,5-dihydrofuran scaffold, twelve new amidoxime derivatives substituted at position 3 with an amide bearing a nitrogen heterocycle were synthesized. This series was designed to replace the sulfone and aryl group on a previously reported HIT. The synthesis of these compounds involved the following three-step pathway: manganese (III) acetate-based cyclization of a β-ketoester, followed by amidation with LiHMDS and a final reaction with hydroxylamine. Three of them, containing either bromine, chlorine, or methyl substitutions and featuring a pyridine moiety, showed an interesting toxicity-activity relationship in vitro. They exhibited IC50 values of 15.0 µM, 16.0 µM, and 17.0 µM against the promastigote form of the parasite and IC50 values of 0.5 µM, 0.6 µM, and 0.3 µM against the intracellular amastigote form, respectively. A selectivity index (SI) greater than 300 was established between the cytotoxic concentrations (in murine macrophages) and the effective concentrations (against the intracellular form of Leishmania amazonensis). This SI is at least seventy times higher than that observed for Pentamidine and twenty-five times higher than that observed for the reference HIT, as previously reported.
Collapse
Affiliation(s)
| | - Oscar Leonardo Avendaño Leon
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, CS30064, CEDEX 05, 13385 Marseille, France; (O.L.A.L.); (C.C.); (Y.K.); (S.R.)
| | - Christophe Curti
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, CS30064, CEDEX 05, 13385 Marseille, France; (O.L.A.L.); (C.C.); (Y.K.); (S.R.)
- Service Central de la Qualité et de l’Information Pharmaceutiques (SCQIP), Pharmacy Department, Assistance Publique—Hôpitaux de Marseille (AP-HM), 147 Bd. Baille, 13006 Marseille, France
| | - Youssef Kabri
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, CS30064, CEDEX 05, 13385 Marseille, France; (O.L.A.L.); (C.C.); (Y.K.); (S.R.)
| | - Sébastien Redon
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, CS30064, CEDEX 05, 13385 Marseille, France; (O.L.A.L.); (C.C.); (Y.K.); (S.R.)
| | - Eduardo Caio Torres-Santos
- Laboratório de Bioquímica de Tripanosomatídeos, Instituto Oswaldo Cruz—FIOCRUZ, Av. Brasil, 4365, Rio de Janeiro 21040-900, Brazil;
| | - Patrice Vanelle
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, CS30064, CEDEX 05, 13385 Marseille, France; (O.L.A.L.); (C.C.); (Y.K.); (S.R.)
- Service Central de la Qualité et de l’Information Pharmaceutiques (SCQIP), Pharmacy Department, Assistance Publique—Hôpitaux de Marseille (AP-HM), 147 Bd. Baille, 13006 Marseille, France
| |
Collapse
|
2
|
Klopp C, Zhang X, Campbell MK, Kvaskoff D, Struwe MA, Warren CR, Bajrami B, Scheidig AJ, Jones AK, Clement B. mARC1 Is the Main Contributor to Metabolic Reduction of N-Hydroxyurea. J Med Chem 2024; 67:18090-18097. [PMID: 39397364 PMCID: PMC11513889 DOI: 10.1021/acs.jmedchem.4c01148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024]
Abstract
N-Hydroxyurea has been known since the 1960s as an antiproliferative drug and is used both in oncology and for treatment of hematological disorders such as sickle cell anemia where very high daily doses are administered. It is assumed that the cellular effect of N-hydroxyurea is caused by inhibition of ribonucleotide reductase, while alternative mechanisms, e.g., generation of nitric oxide, have also been proposed. Despite its many therapeutic applications, the metabolism of hydroxyurea is largely unexplored. The major elimination pathway of N-hydroxyurea is the reduction to urea. Since the mitochondrial amidoxime reducing component (mARC) is known for its N-reductive activity, we investigated the reduction of NHU by this enzyme system. This study presents in vitro and in vivo evidence that this reductive biotransformation is specifically mediated by the mARC1. Inactivation by mARC1 is a possible explanation for the high doses of NHU required for treatment.
Collapse
Affiliation(s)
- Cathrin Klopp
- Zoological
Institute − Structural Biology, Kiel
University 24118, Kiel, Germany
- Pharmaceutical
Institute − Medicinal Chemistry, Kiel University 24118, Kiel, Germany
| | - Xiaomei Zhang
- Department
of Cardiometabolic Disease Research, Boehringer
Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Morgan K. Campbell
- Department
of Cardiometabolic Disease Research, Boehringer
Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - David Kvaskoff
- Department
of Drug Discovery Sciences, Discovery Science Technologies (DK, BB), Boehringer Ingelheim Pharma GmbH & Co. 88400, Biberach
an der Riss, Germany
| | - Michel A. Struwe
- Zoological
Institute − Structural Biology, Kiel
University 24118, Kiel, Germany
- Pharmaceutical
Institute − Medicinal Chemistry, Kiel University 24118, Kiel, Germany
| | - Curtis R. Warren
- Department
of Cardiometabolic Disease Research, Boehringer
Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Besnik Bajrami
- Department
of Drug Discovery Sciences, Discovery Science Technologies (DK, BB), Boehringer Ingelheim Pharma GmbH & Co. 88400, Biberach
an der Riss, Germany
| | - Axel J. Scheidig
- Zoological
Institute − Structural Biology, Kiel
University 24118, Kiel, Germany
| | - Amanda K. Jones
- Department
of Cardiometabolic Disease Research, Boehringer
Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Bernd Clement
- Pharmaceutical
Institute − Medicinal Chemistry, Kiel University 24118, Kiel, Germany
| |
Collapse
|
3
|
Adamus JP, Ruszczyńska A, Wyczałkowska-Tomasik A. Molybdenum's Role as an Essential Element in Enzymes Catabolizing Redox Reactions: A Review. Biomolecules 2024; 14:869. [PMID: 39062583 PMCID: PMC11275037 DOI: 10.3390/biom14070869] [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: 06/04/2024] [Revised: 07/05/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Molybdenum (Mo) is an essential element for human life, acting as a cofactor in various enzymes crucial for metabolic homeostasis. This review provides a comprehensive insight into the latest advances in research on molybdenum-containing enzymes and their clinical significance. One of these enzymes is xanthine oxidase (XO), which plays a pivotal role in purine catabolism, generating reactive oxygen species (ROS) capable of inducing oxidative stress and subsequent organ dysfunction. Elevated XO activity is associated with liver pathologies such as non-alcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma (HCC). Aldehyde oxidases (AOs) are also molybdenum-containing enzymes that, similar to XO, participate in drug metabolism, with notable roles in the oxidation of various substrates. However, beneath its apparent efficacy, AOs' inhibition may impact drug effectiveness and contribute to liver damage induced by hepatotoxins. Another notable molybdenum-enzyme is sulfite oxidase (SOX), which catalyzes the conversion of sulfite to sulfate, crucial for the degradation of sulfur-containing amino acids. Recent research highlights SOX's potential as a diagnostic marker for HCC, offering promising sensitivity and specificity in distinguishing cancerous lesions. The newest member of molybdenum-containing enzymes is mitochondrial amidoxime-reducing component (mARC), involved in drug metabolism and detoxification reactions. Emerging evidence suggests its involvement in liver pathologies such as HCC and NAFLD, indicating its potential as a therapeutic target. Overall, understanding the roles of molybdenum-containing enzymes in human physiology and disease pathology is essential for advancing diagnostic and therapeutic strategies for various health conditions, particularly those related to liver dysfunction. Further research into the molecular mechanisms underlying these enzymes' functions could lead to novel treatments and improved patient outcomes.
Collapse
Affiliation(s)
- Jakub Piotr Adamus
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Anna Ruszczyńska
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, 02-089 Warsaw, Poland
| | | |
Collapse
|
4
|
Hou W, Watson C, Cecconie T, Bolaki MN, Brady JJ, Lu Q, Gatto GJ, Day TA. Biochemical and functional characterization of the p.A165T missense variant of mitochondrial amidoxime-reducing component 1. J Biol Chem 2024; 300:107353. [PMID: 38723751 PMCID: PMC11190489 DOI: 10.1016/j.jbc.2024.107353] [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: 02/28/2024] [Revised: 04/25/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024] Open
Abstract
Recent genome-wide association studies have identified a missense variant p.A165T in mitochondrial amidoxime-reducing component 1 (mARC1) that is strongly associated with protection from all-cause cirrhosis and improved prognosis in nonalcoholic steatohepatitis. The precise mechanism of this protective effect is unknown. Substitution of alanine 165 with threonine is predicted to affect mARC1 protein stability and to have deleterious effects on its function. To investigate the mechanism, we have generated a knock-in mutant mARC1 A165T and a catalytically dead mutant C273A (as a control) in human hepatoma HepG2 cells, enabling characterization of protein subcellular distribution, stability, and biochemical functions of the mARC1 mutant protein expressed from its endogenous locus. Compared to WT mARC1, we found that the A165T mutant exhibits significant mislocalization outside of its traditional location anchored in the mitochondrial outer membrane and reduces protein stability, resulting in lower basal levels. We evaluated the involvement of the ubiquitin proteasome system in mARC1 A165T degradation and observed increased ubiquitination and faster degradation of the A165T variant. In addition, we have shown that HepG2 cells carrying the MTARC1 p.A165T variant exhibit lower N-reductive activity on exogenously added amidoxime substrates in vitro. The data from these biochemical and functional assays suggest a mechanism by which the MTARC1 p.A165T variant abrogates enzyme function which may contribute to its protective effect in liver disease.
Collapse
Affiliation(s)
- Wangfang Hou
- Respiratory and Immunology Biology Unit, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Christian Watson
- Respiratory and Immunology Biology Unit, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Ted Cecconie
- MEDDesign-NCE-MD SPMB US, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | | | | | - Quinn Lu
- Respiratory and Immunology Biology Unit, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Gregory J Gatto
- Respiratory and Immunology Biology Unit, GlaxoSmithKline, Collegeville, Pennsylvania, USA.
| | - Tovah A Day
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Smagris E, Shihanian LM, Mintah IJ, Bigdelou P, Livson Y, Brown H, Verweij N, Hunt C, Johnson RO, Greer TJ, Hartford SA, Hindy G, Sun L, Nielsen JB, Halasz G, Lotta LA, Murphy AJ, Sleeman MW, Gusarova V. Divergent role of Mitochondrial Amidoxime Reducing Component 1 (MARC1) in human and mouse. PLoS Genet 2024; 20:e1011179. [PMID: 38437227 PMCID: PMC10939284 DOI: 10.1371/journal.pgen.1011179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 03/14/2024] [Accepted: 02/09/2024] [Indexed: 03/06/2024] Open
Abstract
Recent human genome-wide association studies have identified common missense variants in MARC1, p.Ala165Thr and p.Met187Lys, associated with lower hepatic fat, reduction in liver enzymes and protection from most causes of cirrhosis. Using an exome-wide association study we recapitulated earlier MARC1 p.Ala165Thr and p.Met187Lys findings in 540,000 individuals from five ancestry groups. We also discovered novel rare putative loss of function variants in MARC1 with a phenotype similar to MARC1 p.Ala165Thr/p.Met187Lys variants. In vitro studies of recombinant human MARC1 protein revealed Ala165Thr substitution causes protein instability and aberrant localization in hepatic cells, suggesting MARC1 inhibition or deletion may lead to hepatoprotection. Following this hypothesis, we generated Marc1 knockout mice and evaluated the effect of Marc1 deletion on liver phenotype. Unexpectedly, our study found that whole-body Marc1 deficiency in mouse is not protective against hepatic triglyceride accumulation, liver inflammation or fibrosis. In attempts to explain the lack of the observed phenotype, we discovered that Marc1 plays only a minor role in mouse liver while its paralogue Marc2 is the main Marc family enzyme in mice. Our findings highlight the major difference in MARC1 physiological function between human and mouse.
Collapse
Affiliation(s)
- Eriks Smagris
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Lisa M. Shihanian
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Ivory J. Mintah
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Parnian Bigdelou
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Yuliya Livson
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Heather Brown
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Niek Verweij
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Charleen Hunt
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | | | - Tyler J. Greer
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | | | - George Hindy
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Luanluan Sun
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Jonas B. Nielsen
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Gabor Halasz
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Luca A. Lotta
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Andrew J. Murphy
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Mark W. Sleeman
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Viktoria Gusarova
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| |
Collapse
|
6
|
Struwe MA, Scheidig AJ, Clement B. The mitochondrial amidoxime reducing component-from prodrug-activation mechanism to drug-metabolizing enzyme and onward to drug target. J Biol Chem 2023; 299:105306. [PMID: 37778733 PMCID: PMC10637980 DOI: 10.1016/j.jbc.2023.105306] [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: 06/19/2023] [Revised: 09/17/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023] Open
Abstract
The mitochondrial amidoxime-reducing component (mARC) is one of five known molybdenum enzymes in eukaryotes. mARC belongs to the MOSC domain superfamily, a large group of so far poorly studied molybdoenzymes. mARC was initially discovered as the enzyme activating N-hydroxylated prodrugs of basic amidines but has since been shown to also reduce a variety of other N-oxygenated compounds, for example, toxic nucleobase analogs. Under certain circumstances, mARC might also be involved in reductive nitric oxide synthesis through reduction of nitrite. Recently, mARC enzymes have received a lot of attention due to their apparent involvement in lipid metabolism and, in particular, because many genome-wide association studies have shown a common variant of human mARC1 to have a protective effect against liver disease. The mechanism linking mARC enzymes with lipid metabolism remains unknown. Here, we give a comprehensive overview of what is currently known about mARC enzymes, their substrates, structure, and apparent involvement in human disease.
Collapse
Affiliation(s)
- Michel A Struwe
- Zoologisches Institut - Strukturbiologie, Christian-Albrechts-Universität Kiel, Kiel, Germany; Pharmazeutisches Institut, Christian-Albrechts-Universität Kiel, Kiel, Germany.
| | - Axel J Scheidig
- Zoologisches Institut - Strukturbiologie, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - Bernd Clement
- Pharmazeutisches Institut, Christian-Albrechts-Universität Kiel, Kiel, Germany
| |
Collapse
|
7
|
Rixen S, Indorf PM, Kubitza C, Struwe MA, Klopp C, Scheidig AJ, Kunze T, Clement B. Reduction of Hydrogen Peroxide by Human Mitochondrial Amidoxime Reducing Component Enzymes. Molecules 2023; 28:6384. [PMID: 37687214 PMCID: PMC10489706 DOI: 10.3390/molecules28176384] [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: 08/01/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
The mitochondrial amidoxime reducing component (mARC) is a human molybdoenzyme known to catalyze the reduction of various N-oxygenated substrates. The physiological function of mARC enzymes, however, remains unknown. In this study, we examine the reduction of hydrogen peroxide (H2O2) by the human mARC1 and mARC2 enzymes. Furthermore, we demonstrate an increased sensitivity toward H2O2 for HEK-293T cells with an MTARC1 knockout, which implies a role of mARC enzymes in the cellular response to oxidative stress. H2O2 is a reactive oxygen species (ROS) formed in all living cells involved in many physiological processes. Furthermore, H2O2 constitutes the first mARC substrate without a nitrogen-oxygen bond, implying that mARC enzymes may have a substrate spectrum going beyond the previously examined N-oxygenated compounds.
Collapse
Affiliation(s)
- Sophia Rixen
- Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, Kiel University, 24118 Kiel, Germany; (S.R.); (P.M.I.); (M.A.S.); (C.K.); (T.K.)
| | - Patrick M. Indorf
- Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, Kiel University, 24118 Kiel, Germany; (S.R.); (P.M.I.); (M.A.S.); (C.K.); (T.K.)
| | - Christian Kubitza
- Department of Structural Biology, Zoological Institute, Kiel University, 24118 Kiel, Germany; (C.K.); (A.J.S.)
| | - Michel A. Struwe
- Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, Kiel University, 24118 Kiel, Germany; (S.R.); (P.M.I.); (M.A.S.); (C.K.); (T.K.)
- Department of Structural Biology, Zoological Institute, Kiel University, 24118 Kiel, Germany; (C.K.); (A.J.S.)
| | - Cathrin Klopp
- Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, Kiel University, 24118 Kiel, Germany; (S.R.); (P.M.I.); (M.A.S.); (C.K.); (T.K.)
- Department of Structural Biology, Zoological Institute, Kiel University, 24118 Kiel, Germany; (C.K.); (A.J.S.)
| | - Axel J. Scheidig
- Department of Structural Biology, Zoological Institute, Kiel University, 24118 Kiel, Germany; (C.K.); (A.J.S.)
| | - Thomas Kunze
- Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, Kiel University, 24118 Kiel, Germany; (S.R.); (P.M.I.); (M.A.S.); (C.K.); (T.K.)
| | - Bernd Clement
- Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, Kiel University, 24118 Kiel, Germany; (S.R.); (P.M.I.); (M.A.S.); (C.K.); (T.K.)
| |
Collapse
|
8
|
Klopp C, Struwe MA, Plieth C, Clement B, Scheidig AJ. New Design of an Activity Assay Suitable for High-Throughput Screening of Substrates and Inhibitors of the Mitochondrial Amidoxime Reducing Component (mARC). Anal Chem 2023; 95:12452-12458. [PMID: 37549068 DOI: 10.1021/acs.analchem.3c02109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
The mitochondrial amidoxime-reducing component (mARC) is one of the simplest molybdenum-containing enzymes. mARC is among a few known reducing enzymes playing an important role in drug metabolism in mammals. Here, an assay based on the fluorescence of NADH is reported for the rapid detection of substrates and potential inhibitors of mARC. So far unknown inhibitors might be useful for the development of drugs assigned to nonalcoholic fatty liver disease (NAFLD) and similar diseases. Kinetics of reactions catalyzed by mARC can be recorded with high sensitivity and precision. On a microtiter plate scale, the assay presented could be applied for high-throughput screening of substance libraries and detection of novel mARC substrate candidates. For instance, molnupiravir was also identified as a new substrate by this assay. For better comparison for such substances, the inhibitor or substrate-to-BAO ratio was introduced. After normalization of enzyme activities to the standard benzamidoxime, substrates can reproducibly be classified.
Collapse
Affiliation(s)
- Cathrin Klopp
- Pharmaceutical Institute - Medicinal Chemistry, Kiel University, Gutenbergstraße 76, 24118 Kiel, Germany
- Zoological Institute - Structural Biology, Kiel University, Am Botanischen Garten 9, 24118 Kiel, Germany
| | - Michel A Struwe
- Pharmaceutical Institute - Medicinal Chemistry, Kiel University, Gutenbergstraße 76, 24118 Kiel, Germany
- Zoological Institute - Structural Biology, Kiel University, Am Botanischen Garten 9, 24118 Kiel, Germany
| | - Christoph Plieth
- Centre for Biochemistry and Molecular Biology, Kiel University, Am Botanischen Garten 9, 24118 Kiel, Germany
| | - Bernd Clement
- Pharmaceutical Institute - Medicinal Chemistry, Kiel University, Gutenbergstraße 76, 24118 Kiel, Germany
| | - Axel J Scheidig
- Zoological Institute - Structural Biology, Kiel University, Am Botanischen Garten 9, 24118 Kiel, Germany
| |
Collapse
|
9
|
Clement B, Struwe MA. The History of mARC. Molecules 2023; 28:4713. [PMID: 37375270 DOI: 10.3390/molecules28124713] [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: 05/12/2023] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
The mitochondrial amidoxime-reducing component (mARC) is the most recently discovered molybdoenzyme in humans after sulfite oxidase, xanthine oxidase and aldehyde oxidase. Here, the timeline of mARC's discovery is briefly described. The story begins with investigations into N-oxidation of pharmaceutical drugs and model compounds. Many compounds are N-oxidized extensively in vitro, but it turned out that a previously unknown enzyme catalyzes the retroreduction of the N-oxygenated products in vivo. After many years, the molybdoenzyme mARC could finally be isolated and identified in 2006. mARC is an important drug-metabolizing enzyme and N-reduction by mARC has been exploited very successfully for prodrug strategies, that allow oral administration of otherwise poorly bioavailable therapeutic drugs. Recently, it was demonstrated that mARC is a key factor in lipid metabolism and likely involved in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). The exact link between mARC and lipid metabolism is not yet fully understood. Regardless, many now consider mARC a potential drug target for the prevention or treatment of liver diseases. This article focusses on discoveries related to mammalian mARC enzymes. mARC homologues have been studied in algae, plants and bacteria. These will not be discussed extensively here.
Collapse
Affiliation(s)
- Bernd Clement
- Pharmazeutisches Institut, Christian-Albrechts-Universität zu Kiel, Gutenbergstraße 76, 24118 Kiel, Germany
| | - Michel A Struwe
- Pharmazeutisches Institut, Christian-Albrechts-Universität zu Kiel, Gutenbergstraße 76, 24118 Kiel, Germany
- Zoologisches Institut-Strukturbiologie, Zentrum für Biochemie und Molekularbiologie, Christian-Albrechts-Universität zu Kiel, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| |
Collapse
|
10
|
Zapiter J, Harmer JR, Struwe M, Scheidig A, Clement B, Bernhardt PV. Enzyme Electrode Biosensors for N-Hydroxylated Prodrugs Incorporating the Mitochondrial Amidoxime Reducing Component. Anal Chem 2022; 94:9208-9215. [PMID: 35700342 DOI: 10.1021/acs.analchem.2c02232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Human mitochondrial amidoxime reducing component 1 and 2 (mARC1 and mARC2) were immobilised on glassy carbon electrodes using the crosslinker glutaraldehyde. Voltammetry was performed in the presence of the artificial electron transfer mediator methyl viologen, whose redox potential lies negative of the enzymes' MoVI/V and MoV/IV redox potentials which were determined from optical spectroelectrochemical and EPR measurements. Apparent Michaelis constants obtained from catalytic limiting currents at various substrate concentrations were comparable to those previously reported in the literature from enzymatic assays. Kinetic parameters for benzamidoxime reduction were determined from cyclic voltammograms simulated using Digisim. pH dependence and stability of the enzyme electrode with time were also determined from limiting catalytic currents in saturating concentrations of benzamidoxime. The same electrode remained active after at least 9 days. Fabrication of this versatile and cost-effective biosensor is effective in screening new pharmaceutically important substrates and mARC inhibitors.
Collapse
Affiliation(s)
- Joan Zapiter
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
| | - Jeffrey R Harmer
- Centre for Advanced Imaging, The University of Queensland, Brisbane 4072, Australia
| | - Michel Struwe
- Pharmazeutisches Institut, Universität Kiel, Gutenbergstraße 76, Kiel 24118, Germany.,Zoologisches Institut/Strukturbiologie, Am Botanischen Garten 11, Kiel 24118, Germany
| | - Axel Scheidig
- Zoologisches Institut/Strukturbiologie, Am Botanischen Garten 11, Kiel 24118, Germany
| | - Bernd Clement
- Pharmazeutisches Institut, Universität Kiel, Gutenbergstraße 76, Kiel 24118, Germany
| | - Paul V Bernhardt
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
| |
Collapse
|
11
|
Maiber L, Koprivova A, Bender D, Kopriva S, Fischer-Schrader K. Characterization of the amidoxime reducing components ARC1 and ARC2 from Arabidopsis thaliana. FEBS J 2022; 289:5656-5669. [PMID: 35366369 DOI: 10.1111/febs.16450] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 02/26/2022] [Accepted: 03/31/2022] [Indexed: 02/06/2023]
Abstract
Five molybdenum-dependent enzymes are known in eukaryotes. While four of them are under investigation since decades, the most recently discovered, (mitochondrial) amidoxime reducing component ((m)ARC), has only been characterized in mammals and the green algae Chlamydomonas reinhardtii. While mammalian mARCs have been shown to be involved in various signalling pathways, Chlamydomonas ARC was shown to be a nitric oxide (NO)-forming nitrite reductase. Similar to mammals, higher plants possess two ARC proteins. To test whether plant ARCs have a similar function in NO production to the function they have in C. reinhardtii, we analysed the enzymes from the model plant Arabidopsis thaliana. Both ARC1 and ARC2 from Arabidopsis could reduce N-hydroxylated compounds, while nitrite reduction to form NO could only be demonstrated for ARC2. Searching for physiological electron donors, we found that both ARC enzymes accept electrons from NADH via cytochrome b5 reductase and cytochrome b5 , but only ARC2 is able to accept electrons from nitrate reductase at all. Furthermore, arc-deficient mutant plants were similar to wildtype plants regarding growth and also nitrite-dependent NO-formation. Altogether, our results did not confirm the hypothesis that either ARC1 or ARC2 from Arabidopsis are involved in physiologically relevant nitrite-dependent NO-formation. In contrast, our data suggest that ARC1 and ARC2 have distinct, yet unknown physiological roles in higher plants.
Collapse
Affiliation(s)
- Ludmila Maiber
- Department of Chemistry, Institute for Biochemistry, University of Cologne, Germany
| | - Anna Koprivova
- Institute for Plant Sciences, Cluster of Excellence on Plant Sciences, University of Cologne, Germany
| | - Daniel Bender
- Department of Chemistry, Institute for Biochemistry, University of Cologne, Germany
| | - Stanislav Kopriva
- Institute for Plant Sciences, Cluster of Excellence on Plant Sciences, University of Cologne, Germany
| | | |
Collapse
|
12
|
Kalimuthu P, Harmer JR, Baldauf M, Hassan AH, Kruse T, Bernhardt PV. Electrochemically driven catalysis of the bacterial molybdenum enzyme YiiM. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2022; 1863:148523. [PMID: 34921810 DOI: 10.1016/j.bbabio.2021.148523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/18/2021] [Accepted: 12/07/2021] [Indexed: 12/20/2022]
Abstract
The Mo-dependent enzyme YiiM enzyme from Escherichia coli is a member of the sulfite oxidase family and shares many similarities with the well-studied human mitochondrial amidoxime reducing component (mARC). We have investigated YiiM catalysis using electrochemical and spectroscopic methods. EPR monitored redox potentiometry found the active site redox potentials to be MoVI/V -0.02 V and MoV/IV -0.12 V vs NHE at pH 7.2. In the presence of methyl viologen as an electrochemically reduced electron donor, YiiM catalysis was studied with a range of potential substrates. YiiM preferentially reduces N-hydroxylated compounds such as hydroxylamines, amidoximes, N-hydroxypurines and N-hydroxyureas but shows little or no activity against amine-oxides or sulfoxides. The pH optimum for catalysis was 7.1 and a bell-shaped pH profile was found with pKa values of 6.2 and 8.1 either side of this optimum that are associated with protonation/deprotonations that modulate activity. Simulation of the experimental voltammetry elucidated kinetic parameters associated with YiiM catalysis with the substrates 6-hydroxyaminopurine and benzamidoxime.
Collapse
Affiliation(s)
- Palraj Kalimuthu
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, 4072, Australia
| | - Jeffrey R Harmer
- Centre for Advanced Imaging, University of Queensland, Brisbane, 4072, Australia
| | - Milena Baldauf
- Department of Plant Biology, Technische Universitaet Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
| | - Ahmed H Hassan
- Department of Plant Biology, Technische Universitaet Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
| | - Tobias Kruse
- Department of Plant Biology, Technische Universitaet Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
| | - Paul V Bernhardt
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, 4072, Australia.
| |
Collapse
|
13
|
Indorf P, Patzak A, Lichtenberger F. Drug metabolism in animal models and humans: Translational aspects and chances for individual therapy. Acta Physiol (Oxf) 2021; 233:e13734. [PMID: 34637592 DOI: 10.1111/apha.13734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Patrick Indorf
- Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, Institute of Vegetative Physiology Charité—Universitätsmedizin Berlin Berlin Germany
| | - Andreas Patzak
- Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, Institute of Vegetative Physiology Charité—Universitätsmedizin Berlin Berlin Germany
| | - Falk‐Bach Lichtenberger
- Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, Institute of Vegetative Physiology Charité—Universitätsmedizin Berlin Berlin Germany
| |
Collapse
|
14
|
Maccallini C, Marinelli L, Indorf P, Cacciatore I, Fantacuzzi M, Clement B, Di Stefano A, Amoroso R. A Novel Prodrug of a nNOS Inhibitor with Improved Pharmacokinetic Potential. ChemMedChem 2020; 15:2157-2163. [PMID: 32783298 PMCID: PMC7756445 DOI: 10.1002/cmdc.202000349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/05/2020] [Indexed: 12/15/2022]
Abstract
Under different pathological conditions, aberrant induction of neuronal nitric oxide synthase (nNOS) generates overproduction of NO that can cause irreversible cell damage. The aim of this study was to develop an amidoxime prodrug of a potent nNOS inhibitor, the benzhydryl acetamidine. We synthesized the benzhydryl acetamidoxime, which was evaluated in vitro to ascertain the potential NOS inhibitory activity, as well as conducting bioconversion into the parent acetamidine. The prodrug was also profiled for in vitro physicochemical properties, by determining the lipophilicity, passive permeation through the human gastrointestinal tract and across the blood-brain barrier by PAMPA, and chemical, enzymatic, and plasma stability. The obtained data demonstrate that the amidoxime prodrug shows an improved pharmacokinetic profile with respect to the acetamidine nNOS inhibitor, thus suggesting that it could be a promising lead compound to treat all those pathological conditions in which nNOS activity is dysregulated.
Collapse
Affiliation(s)
- Cristina Maccallini
- Department of PharmacyUniversity “G. d'Annunzio” of Chieti - Pescaravia dei Vestini 3166100ChietiItaly
| | - Lisa Marinelli
- Department of PharmacyUniversity “G. d'Annunzio” of Chieti - Pescaravia dei Vestini 3166100ChietiItaly
| | - Patrick Indorf
- Pharmaceutical InstituteUniversity of KielGutenbergstraße. 7624118KielGermany
| | - Ivana Cacciatore
- Department of PharmacyUniversity “G. d'Annunzio” of Chieti - Pescaravia dei Vestini 3166100ChietiItaly
| | - Marialuigia Fantacuzzi
- Department of PharmacyUniversity “G. d'Annunzio” of Chieti - Pescaravia dei Vestini 3166100ChietiItaly
| | - Bernd Clement
- Pharmaceutical InstituteUniversity of KielGutenbergstraße. 7624118KielGermany
| | - Antonio Di Stefano
- Department of PharmacyUniversity “G. d'Annunzio” of Chieti - Pescaravia dei Vestini 3166100ChietiItaly
| | - Rosa Amoroso
- Department of PharmacyUniversity “G. d'Annunzio” of Chieti - Pescaravia dei Vestini 3166100ChietiItaly
| |
Collapse
|