1
|
Failla M, Molaro MC, Schiano ME, Serafini M, Tiburtini GA, Gianquinto E, Scoccia R, Battisegola C, Rimoli MG, Chegaev K, Ercolano G, Lazzarato L, Spyrakis F, Sodano F. Opportunities and Challenges of Arginase Inhibitors in Cancer: A Medicinal Chemistry Perspective. J Med Chem 2024; 67:19988-20021. [PMID: 39558532 DOI: 10.1021/acs.jmedchem.4c01429] [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: 11/20/2024]
Abstract
The overexpression of two arginase (ARG) isoforms, ARG1 and ARG2, contributes to the onset of numerous disorders, including cardiovascular and immune-mediated diseases, as well as tumors. To elucidate the specific roles of ARG1 and ARG2 without interfering with their physiological functions, it is crucial to develop effective ARG inhibitors that target only one isoform, while maintaining low toxicity and an adequate pharmacokinetic profile. In this context, we present a comprehensive overview of the different generations of ARG inhibitors. Given the general lack of selectivity in most existing inhibitors, we analyzed the structural features and plasticity of the ARG1 and ARG2 binding sites to explore the potential for designing inhibitors with novel binding patterns. We also review ongoing preclinical and clinical studies on selected inhibitors, highlighting both progress and challenges in developing potent, selective ARG inhibitors. Furthermore, we discuss medicinal chemistry strategies that may accelerate the discovery of selective ARG inhibitors.
Collapse
Affiliation(s)
- Mariacristina Failla
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | | | | | - Marta Serafini
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | | | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Riccardo Scoccia
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Chiara Battisegola
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Maria Grazia Rimoli
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Giuseppe Ercolano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Loretta Lazzarato
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Federica Sodano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
2
|
Muller J, Marchisio L, Attia R, Zedet A, Maradan R, Vallet M, Aebischer A, Harakat D, Senejoux F, Ramseyer C, Foley S, Cardey B, Girard C, Pudlo M. A colorimetric assay adapted to fragment screening revealing aurones and chalcones as new arginase inhibitors. RSC Med Chem 2024; 15:1722-1730. [PMID: 38784454 PMCID: PMC11110760 DOI: 10.1039/d3md00713h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/23/2024] [Indexed: 05/25/2024] Open
Abstract
Arginase, a difficult-to-target metalloenzyme, is implicated in a wide range of diseases, including cancer, infectious, and cardiovascular diseases. Despite the medical need, existing inhibitors have limited structural diversity, consisting predominantly of amino acids and their derivatives. The search for innovative arginase inhibitors has now extended to screening approaches. Due to the small and narrow active site of arginase, screening must meet the criteria of fragment-based screening. However, the limited binding capacity of fragments requires working at high concentrations, which increases the risk of interference and false positives. In this study, we investigated three colorimetric assays and selected one based on interference for screening under these challenging conditions. The subsequent adaptation and application to the screening a library of metal chelator fragments resulted in the identification of four compounds with moderate activity. The synthesis and evaluation of a series of compounds from one of the hits led to compound 21a with an IC50 value of 91.1 μM close to the reference compound piceatannol. Finally, molecular modelling supports the potential binding of aurones and chalcones to the active site of arginase, suggesting them as new candidates for the development of novel arginase inhibitors.
Collapse
Affiliation(s)
- Jason Muller
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Luca Marchisio
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Rym Attia
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Andy Zedet
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Robin Maradan
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Maxence Vallet
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Alison Aebischer
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Dominique Harakat
- URCATech, ICMR, CNRS UMR 7312 URCA Bât 18, BP 1039, Cedex 2 51687 Reims France
| | - François Senejoux
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Christophe Ramseyer
- Chrono-environnement UMR6249, CNRS Université de Franche-Comté F-25000 Besançon France
| | - Sarah Foley
- Chrono-environnement UMR6249, CNRS Université de Franche-Comté F-25000 Besançon France
| | - Bruno Cardey
- Chrono-environnement UMR6249, CNRS Université de Franche-Comté F-25000 Besançon France
| | - Corine Girard
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Marc Pudlo
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| |
Collapse
|
3
|
Baj J, Flieger W, Barbachowska A, Kowalska B, Flieger M, Forma A, Teresiński G, Portincasa P, Buszewicz G, Radzikowska-Büchner E, Flieger J. Consequences of Disturbing Manganese Homeostasis. Int J Mol Sci 2023; 24:14959. [PMID: 37834407 PMCID: PMC10573482 DOI: 10.3390/ijms241914959] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023] Open
Abstract
Manganese (Mn) is an essential trace element with unique functions in the body; it acts as a cofactor for many enzymes involved in energy metabolism, the endogenous antioxidant enzyme systems, neurotransmitter production, and the regulation of reproductive hormones. However, overexposure to Mn is toxic, particularly to the central nervous system (CNS) due to it causing the progressive destruction of nerve cells. Exposure to manganese is widespread and occurs by inhalation, ingestion, or dermal contact. Associations have been observed between Mn accumulation and neurodegenerative diseases such as manganism, Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. People with genetic diseases associated with a mutation in the gene associated with impaired Mn excretion, kidney disease, iron deficiency, or a vegetarian diet are at particular risk of excessive exposure to Mn. This review has collected data on the current knowledge of the source of Mn exposure, the experimental data supporting the dispersive accumulation of Mn in the brain, the controversies surrounding the reference values of biomarkers related to Mn status in different matrices, and the competitiveness of Mn with other metals, such as iron (Fe), magnesium (Mg), zinc (Zn), copper (Cu), lead (Pb), calcium (Ca). The disturbed homeostasis of Mn in the body has been connected with susceptibility to neurodegenerative diseases, fertility, and infectious diseases. The current evidence on the involvement of Mn in metabolic diseases, such as type 2 diabetes mellitus/insulin resistance, osteoporosis, obesity, atherosclerosis, and non-alcoholic fatty liver disease, was collected and discussed.
Collapse
Affiliation(s)
- Jacek Baj
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (W.F.); (A.F.)
| | - Wojciech Flieger
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (W.F.); (A.F.)
| | - Aleksandra Barbachowska
- Department of Plastic, Reconstructive and Burn Surgery, Medical University of Lublin, 21-010 Łęczna, Poland;
| | - Beata Kowalska
- Department of Water Supply and Wastewater Disposal, Lublin University of Technology, 20-618 Lublin, Poland;
| | - Michał Flieger
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | - Alicja Forma
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (W.F.); (A.F.)
| | - Grzegorz Teresiński
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | - Piero Portincasa
- Clinica Medica A. Murri, Department of Biomedical Sciences & Human Oncology, Medical School, University of Bari, 70124 Bari, Italy;
| | - Grzegorz Buszewicz
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | | | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
4
|
Distinct binding pattern of nor-NOHA inhibitor to liver arginase in aqueous solution – Perspectives from molecular dynamics simulations. J Mol Liq 2023. [DOI: 10.1016/j.molliq.2022.121014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
5
|
Sharma K, Sharma KK, Mahindra A, Sehra N, Bagra N, Aaghaz S, Parmar R, Rathod GK, Jain R. Design, synthesis, and applications of ring-functionalized histidines in peptide-based medicinal chemistry and drug discovery. Med Res Rev 2023. [PMID: 36710510 DOI: 10.1002/med.21936] [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: 03/09/2022] [Revised: 12/12/2022] [Accepted: 01/06/2023] [Indexed: 01/31/2023]
Abstract
Modified and synthetic α-amino acids are known to show diverse applications. Histidine, which possesses numerous applications when subjected to synthetic modifications, is one such amino acid. The utility of modified histidines varies widely from remarkable biological activities to catalysis, and from nanotechnology to polymer chemistry. This renders histidine residue an important place in scientific research. Histidine is a well-studied scaffold and constitutes the active site of various enzymes catalyzing important reactions in the biological systems. A rational modification in histidine structure with a distinctly developed protocol extensively changes its physical and chemical properties. The utilization of modified histidines in search of potent, target selective and proteostable scaffolds is vital in the development of bioactive peptides with enhanced drug-likeliness. This review is a compilation and analysis of reported side-chain ring modifications at histidine followed by applications of ring-modified histidines in the synthesis of various categories of bioactive peptides and peptidomimetics.
Collapse
Affiliation(s)
- Komal Sharma
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India
| | - Krishna K Sharma
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India
| | - Amit Mahindra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India
| | - Naina Sehra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India
| | - Nitin Bagra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India
| | - Shams Aaghaz
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India
| | - Rajesh Parmar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India
| | - Gajanan K Rathod
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India
| | - Rahul Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Nagar, Punjab, India
| |
Collapse
|
6
|
Chang YS, Lin CY, Liu TY, Huang CM, Chung CC, Chen YC, Tsai FJ, Chang JG, Chang SJ. Polygenic risk score trend and new variants on chromosome 1 are associated with male gout in genome-wide association study. Arthritis Res Ther 2022; 24:229. [PMID: 36221101 PMCID: PMC9552457 DOI: 10.1186/s13075-022-02917-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 09/24/2022] [Indexed: 11/30/2022] Open
Abstract
Background Gout is a highly hereditary disease, but not all those carrying well-known risk variants have developing gout attack even in hyperuricemia status. We performed a genome-wide association study (GWAS) and polygenic risk score (PRS) analysis to illustrate the new genetic architectures of gout and asymptomatic hyperuricemia (AH). Methods GWAS was performed to identify variants associated with gout/AH compared with normouricemia. The participants were males, enrolled from the Taiwan Biobank and China Medical University, and divided into discovery (n=39,594) and replication (n=891) cohorts for GWAS. For PRS analysis, the discovery cohort was grouped as base (n=21,814) and target (n=17,780) cohorts, and the score was estimated by grouping the polymorphisms into protective or not for the phenotypes in the base cohort. Results The genes ABCG2 and SLC2A9 were found as the major genetic factors governing gouty and AH, and even in those carrying the rs2231142 (ABCG2) wild-genotype. Surprisingly, variants on chromosome 1, such as rs7546668 (DNAJC16), rs10927807 (AGMAT), rs9286836 (NUDT17), rs4971100 (TRIM46), rs4072037 (MUC1), and rs2974935 (MTX1), showed significant associations with gout in both discovery and replication cohorts (all p-values < 1e−8). Concerning the PRS, the rates of gout and AH increased with increased quartile PRS in those SNPs having risk effects on the phenotypes; on the contrary, gout/AH rates decreased with increased quartile PRS in those protective SNPs. Conclusions We found new variants on chromosome 1 significantly relating to gout, and PRS predicts the risk of developing gout/AH more robustly based on the SNPs’ effect types on the trait. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02917-4.
Collapse
Affiliation(s)
- Ya-Sian Chang
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, China Medical University, Taichung, Taiwan.,Graduate Institute of Integrated Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Chien-Yu Lin
- Graduate Institute of Clinical Medical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Division of Laboratory Medicine, China Medical University Hsinchu Hospital, Zhubei City, Taiwan
| | - Ting-Yuan Liu
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chung-Ming Huang
- Graduate Institute of Integrated Medicine, College of Medicine, China Medical University, Taichung, Taiwan.,Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chin-Chun Chung
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, China Medical University, Taichung, Taiwan.,Graduate Institute of Integrated Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Yu-Chia Chen
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| | - Jan-Gowth Chang
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, China Medical University, Taichung, Taiwan. .,Graduate Institute of Integrated Medicine, College of Medicine, China Medical University, Taichung, Taiwan.
| | - Shun-Jen Chang
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, China Medical University, Taichung, Taiwan. .,Department of Kinesiology, Health and Leisure Studies, National University of Kaohsiung, No. 700, Kaohsiung University Road, Nanzih District, 81148, Kaohsiung, Taiwan.
| |
Collapse
|
7
|
Muller J, Attia R, Zedet A, Girard C, Pudlo M. An Update on Arginase Inhibitors and Inhibitory Assays. Mini Rev Med Chem 2021; 22:1963-1976. [PMID: 34967285 DOI: 10.2174/1389557522666211229105703] [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: 09/20/2021] [Revised: 10/18/2021] [Accepted: 10/22/2021] [Indexed: 11/22/2022]
Abstract
Arginase, which converts arginine into ornithine and urea, is a promising therapeutic target. Arginase is involved in cardiovascular diseases, parasitic infections and, through a critical role in immunity, in some cancers. There is a need to develop effective arginase inhibitors and therefore efforts to identify and optimize new inhibitors are increasing. Several methods of evaluating arginase activity are available, but few directly measure the product. Radiometric assays need to separate urea and dying reactions require acidic conditions and sometimes heating. Hence, there are a variety of different approaches available, and each approach has its own limits and benefits. In this review, we provide an update on arginase inhibitors, followed by a discussion on available arginase assays and alternative methods, with a focus on the intrinsic biases and parameters that are likely to impact results.
Collapse
Affiliation(s)
- Jason Muller
- PEPITE EA4267, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| | - Rym Attia
- PEPITE EA4267, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| | - Andy Zedet
- PEPITE EA4267, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| | - Corine Girard
- PEPITE EA4267, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| | - Marc Pudlo
- PEPITE EA4267, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| |
Collapse
|
8
|
S. Clemente G, van Waarde A, F. Antunes I, Dömling A, H. Elsinga P. Arginase as a Potential Biomarker of Disease Progression: A Molecular Imaging Perspective. Int J Mol Sci 2020; 21:E5291. [PMID: 32722521 PMCID: PMC7432485 DOI: 10.3390/ijms21155291] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022] Open
Abstract
Arginase is a widely known enzyme of the urea cycle that catalyzes the hydrolysis of L-arginine to L-ornithine and urea. The action of arginase goes beyond the boundaries of hepatic ureogenic function, being widespread through most tissues. Two arginase isoforms coexist, the type I (Arg1) predominantly expressed in the liver and the type II (Arg2) expressed throughout extrahepatic tissues. By producing L-ornithine while competing with nitric oxide synthase (NOS) for the same substrate (L-arginine), arginase can influence the endogenous levels of polyamines, proline, and NO•. Several pathophysiological processes may deregulate arginase/NOS balance, disturbing the homeostasis and functionality of the organism. Upregulated arginase expression is associated with several pathological processes that can range from cardiovascular, immune-mediated, and tumorigenic conditions to neurodegenerative disorders. Thus, arginase is a potential biomarker of disease progression and severity and has recently been the subject of research studies regarding the therapeutic efficacy of arginase inhibitors. This review gives a comprehensive overview of the pathophysiological role of arginase and the current state of development of arginase inhibitors, discussing the potential of arginase as a molecular imaging biomarker and stimulating the development of novel specific and high-affinity arginase imaging probes.
Collapse
Affiliation(s)
- Gonçalo S. Clemente
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| | - Aren van Waarde
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| | - Inês F. Antunes
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| | - Alexander Dömling
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV Groningen, The Netherlands;
| | - Philip H. Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| |
Collapse
|
9
|
Functional analysis of the Mn2+ requirement in the catalysis of ureohydrolases arginase and agmatinase - a historical perspective. J Inorg Biochem 2020; 202:110812. [DOI: 10.1016/j.jinorgbio.2019.110812] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/13/2019] [Accepted: 08/25/2019] [Indexed: 12/22/2022]
|
10
|
Chen AY, Adamek RN, Dick BL, Credille CV, Morrison CN, Cohen SM. Targeting Metalloenzymes for Therapeutic Intervention. Chem Rev 2019; 119:1323-1455. [PMID: 30192523 PMCID: PMC6405328 DOI: 10.1021/acs.chemrev.8b00201] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metalloenzymes are central to a wide range of essential biological activities, including nucleic acid modification, protein degradation, and many others. The role of metalloenzymes in these processes also makes them central for the progression of many diseases and, as such, makes metalloenzymes attractive targets for therapeutic intervention. Increasing awareness of the role metalloenzymes play in disease and their importance as a class of targets has amplified interest in the development of new strategies to develop inhibitors and ultimately useful drugs. In this Review, we provide a broad overview of several drug discovery efforts focused on metalloenzymes and attempt to map out the current landscape of high-value metalloenzyme targets.
Collapse
Affiliation(s)
- Allie Y Chen
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Rebecca N Adamek
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Benjamin L Dick
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Cy V Credille
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Christine N Morrison
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Seth M Cohen
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| |
Collapse
|
11
|
Abdelkawy KS, Lack K, Elbarbry F. Pharmacokinetics and Pharmacodynamics of Promising Arginase Inhibitors. Eur J Drug Metab Pharmacokinet 2018; 42:355-370. [PMID: 27734327 DOI: 10.1007/s13318-016-0381-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Up-regulation of arginase activity in several chronic disease conditions, including cancer and hypertension, may suggest new targets for treatment. Recently, the number of new arginase inhibitors with promising therapeutic effects for asthma, cancer, hypertension, diabetes mellitus, and erectile dysfunction has shown a remarkable increase. Arginase inhibitors may be chemical substances, such as boron-based amino acid derivatives, α-difluoromethylornithine (DMFO), and Nω-hydroxy-nor-L-arginine (nor-NOHA) or, of plant origin such as sauchinone, salvianolic acid B (SAB), piceatannol-3-O-β-D-glucopyranoside (PG) and obacunone. Despite their promising therapeutic potential, little is known about pharmacokinetics and pharmacodynamics of some of these agents. Several studies were conducted in different animal species and in vitro systems and reported significant differences in pharmacokinetics and pharmacodynamics of arginase inhibitors. Therefore, extra caution should be considered before extrapolating these studies to human. Physicochemical and pharmacokinetic profiles of some effective arginase inhibitors make it challenging to formulate stable and effective formulation. In this article, existing literature on the pharmacokinetics and pharmacodynamics of arginase inhibitors were reviewed and compared together with emphasis on possible drug interactions and solutions to overcome pharmacokinetics challenges and shortage of arginase inhibitors in clinical practice.
Collapse
Affiliation(s)
| | - Kelsey Lack
- School of Pharmacy, Pacific University, 222 SE 8th Ave., Hillsboro, OR, 97123, USA
| | - Fawzy Elbarbry
- School of Pharmacy, Pacific University, 222 SE 8th Ave., Hillsboro, OR, 97123, USA.
| |
Collapse
|
12
|
Arginase Structure and Inhibition: Catalytic Site Plasticity Reveals New Modulation Possibilities. Sci Rep 2017; 7:13616. [PMID: 29051526 PMCID: PMC5648838 DOI: 10.1038/s41598-017-13366-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/22/2017] [Indexed: 01/23/2023] Open
Abstract
Metalloenzyme arginase is a therapeutically relevant target associated with tumor growth. To fight cancer immunosuppression, arginase activity can be modulated by small chemical inhibitors binding to its catalytic center. To better understand molecular mechanisms of arginase inhibition, a careful computer-aided mechanistic structural investigation of this enzyme was conducted. Using molecular dynamics (MD) simulations in the microsecond range, key regions of the protein active site were identified and their flexibility was evaluated and compared. A cavity opening phenomenon was observed, involving three loops directly interacting with all known ligands, while metal coordinating regions remained motionless. A novel dynamic 3D pharmacophore analysis method termed dynophores has been developed that allows for the construction of a single 3D-model comprising all ligand-enzyme interactions occurring throughout a complete MD trajectory. This new technique for the in silico study of intermolecular interactions allows for loop flexibility analysis coupled with movements and conformational changes of bound ligands. Presented MD studies highlight the plasticity of the size of the arginase active site, leading to the hypothesis that larger ligands can enter the cavity of arginase. Experimental testing of a targeted fragment library substituted by different aliphatic groups validates this hypothesis, paving the way for the design of arginase inhibitors with novel binding patterns.
Collapse
|
13
|
Pudlo M, Demougeot C, Girard-Thernier C. Arginase Inhibitors: A Rational Approach Over One Century. Med Res Rev 2016; 37:475-513. [DOI: 10.1002/med.21419] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/14/2016] [Accepted: 09/22/2016] [Indexed: 12/28/2022]
Affiliation(s)
- Marc Pudlo
- PEPITE - EA4267; University Bourgogne Franche-Comté; Besançon France
| | - Céline Demougeot
- PEPITE - EA4267; University Bourgogne Franche-Comté; Besançon France
| | | |
Collapse
|
14
|
Pham TN, Bordage S, Pudlo M, Demougeot C, Thai KM, Girard-Thernier C. Cinnamide Derivatives as Mammalian Arginase Inhibitors: Synthesis, Biological Evaluation and Molecular Docking. Int J Mol Sci 2016; 17:E1656. [PMID: 27690022 PMCID: PMC5085689 DOI: 10.3390/ijms17101656] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/22/2016] [Accepted: 09/23/2016] [Indexed: 12/24/2022] Open
Abstract
Arginases are enzymes that are involved in many human diseases and have been targeted for new treatments. Here a series of cinnamides was designed, synthesized and evaluated in vitro and in silico for their inhibitory activity against mammalian arginase. Using a microassay on purified liver bovine arginase (b-ARG I), (E)-N-(2-phenylethyl)-3,4-dihydroxycinnamide, also named caffeic acid phenylamide (CAPA), was shown to be slightly more active than our natural reference inhibitor, chlorogenic acid (IC50 = 6.9 ± 1.3 and 10.6 ± 1.6 µM, respectively) but it remained less active that the synthetic reference inhibitor Nω-hydroxy-nor-l-arginine nor-NOHA (IC50 = 1.7 ± 0.2 µM). Enzyme kinetic studies showed that CAPA was a competitive inhibitor of arginase with Ki = 5.5 ± 1 µM. Whereas the activity of nor-NOHA was retained (IC50 = 5.7 ± 0.6 µM) using a human recombinant arginase I (h-ARG I), CAPA showed poorer activity (IC50 = 60.3 ± 7.8 µM). However, our study revealed that the cinnamoyl moiety and catechol function were important for inhibitory activity. Docking results on h-ARG I demonstrated that the caffeoyl moiety could penetrate into the active-site pocket of the enzyme, and the catechol function might interact with the cofactor Mn2+ and several crucial amino acid residues involved in the hydrolysis mechanism of arginase. The results of this study suggest that 3,4-dihydroxycinnamides are worth being considered as potential mammalian arginase inhibitors, and could be useful for further research on the development of new arginase inhibitors.
Collapse
Affiliation(s)
- Thanh-Nhat Pham
- PEPITE EA4267, University Bourgogne Franche-Comté, F-25000 Besançon, France.
| | - Simon Bordage
- PEPITE EA4267, University Bourgogne Franche-Comté, F-25000 Besançon, France.
- University Lille, EA 7394-ICV-Institut Charles Viollette, F-59000 Lille, France.
| | - Marc Pudlo
- PEPITE EA4267, University Bourgogne Franche-Comté, F-25000 Besançon, France.
| | - Céline Demougeot
- PEPITE EA4267, University Bourgogne Franche-Comté, F-25000 Besançon, France.
| | - Khac-Minh Thai
- Department of Medicinal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 41 Dinh Tien Hoang, Dist 1, Ho Chi Minh City 700000, Vietnam.
| | | |
Collapse
|
15
|
Fedoseev P, Sharma N, Khunt R, Ermolat'ev DS, Van der Eycken EV. Iodine-mediated regioselective guanylation-amination of propargylamines towards the synthesis of diversely substituted 2-aminoimidazoles. RSC Adv 2016. [DOI: 10.1039/c6ra13371a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A diversity-oriented approach for the synthesis of 2-aminoimidazoles is presented.
Collapse
Affiliation(s)
- Pavel Fedoseev
- Laboratory for Organic & Microwave-Assisted Chemistry (LOMAC)
- Department of Chemistry
- University of Leuven (KU Leuven)
- Belgium
| | - Nandini Sharma
- Laboratory for Organic & Microwave-Assisted Chemistry (LOMAC)
- Department of Chemistry
- University of Leuven (KU Leuven)
- Belgium
| | - Rupesh Khunt
- Laboratory for Organic & Microwave-Assisted Chemistry (LOMAC)
- Department of Chemistry
- University of Leuven (KU Leuven)
- Belgium
| | - Denis S. Ermolat'ev
- Laboratory for Organic & Microwave-Assisted Chemistry (LOMAC)
- Department of Chemistry
- University of Leuven (KU Leuven)
- Belgium
| | - Erik V. Van der Eycken
- Laboratory for Organic & Microwave-Assisted Chemistry (LOMAC)
- Department of Chemistry
- University of Leuven (KU Leuven)
- Belgium
| |
Collapse
|
16
|
Dražić T, Vazdar K, Vazdar M, Đaković M, Mikecin AM, Kralj M, Malnar M, Hećimović S, Habuš I. Synthesis of new 2-aminoimidazolones with antiproliferative activity via base promoted amino-β-lactam rearrangement. Tetrahedron 2015. [DOI: 10.1016/j.tet.2015.10.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
17
|
Aydin M, Altintas N, Cem Mutlu L, Bilir B, Oran M, Tülübaş F, Topçu B, Tayfur İ, Küçükyalçin V, Kaplan G, Gürel A. Asymmetric dimethylarginine contributes to airway nitric oxide deficiency in patients with COPD. CLINICAL RESPIRATORY JOURNAL 2015; 11:318-327. [PMID: 26076870 DOI: 10.1111/crj.12337] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 05/17/2015] [Accepted: 06/12/2015] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Asymmetric dimethylarginine (ADMA) and nitric oxide (NO) show their mechanism of action reciprocally, the balance between these molecules contributes to the tight regulation of airways tone and function. OBJECTIVES The aim of this study to determine the serum levels of ADMA and NO in patients with chronic obstructive pulmonary disease (COPD) and establish whether their level vary in relation to forced expiratory volume in 1s (FEV1 ), to assess their role in pathophysiology of COPD. MATERIALS AND METHODS This study consisted of 58 patients with COPD and 30 healthy subjects. Serum ADMA and NO levels were measured using enzyme-linked immunosorbent assay and the colorimetric method, respectively. RESULTS Serum ADMA levels were significantly higher, however, NO levels were lower in patients with COPD compared with controls. ADMA levels were inversely correlated with NO levels. Serum ADMA and NO were significantly correlated with FEV1 . Multivariable logistic regression analysis revealed that serum ADMA and NO were independently and significantly associated with the presence of COPD. Multiple linear regression analysis showed that COPD was positively associated with ADMA, additionally COPD and ADMA were independently and inversely associated with NO. NO levels were decreased, ADMA levels were increased compliant with progression of COPD stages. CONCLUSION While circulating ADMA is higher, NO is lower in COPD and both show a strong correlation to the degree of airflow limitation. ADMA seems to be a possible new marker of prognosis of COPD and can be a novel therapeutic target for the treatment of COPD.
Collapse
Affiliation(s)
- Murat Aydin
- Department of Biochemistry, School of Medicine, Namik Kemal University, Tekirdağ, Turkey
| | - Nejat Altintas
- Department of Pulmonary and Sleep Medicine, School of Medicine, Namik Kemal University, Tekirdağ, Turkey
| | - Levent Cem Mutlu
- Department of Pulmonary and Sleep Medicine, School of Medicine, Namik Kemal University, Tekirdağ, Turkey
| | - Bulent Bilir
- Department of Internal Medicine, School of Medicine, Namik Kemal University, Tekirdağ, Turkey
| | - Mustafa Oran
- Department of Internal Medicine, School of Medicine, Namik Kemal University, Tekirdağ, Turkey
| | - Feti Tülübaş
- Department of Biochemistry, School of Medicine, Namik Kemal University, Tekirdağ, Turkey
| | - Birol Topçu
- Department of Biostatistics, School of Medicine, Namik Kemal University, Tekirdağ, Turkey
| | - İsmail Tayfur
- Department of Biochemistry, School of Medicine, Namik Kemal University, Tekirdağ, Turkey
| | - Volkan Küçükyalçin
- Department of Biochemistry, School of Medicine, Namik Kemal University, Tekirdağ, Turkey
| | - Gizem Kaplan
- Department of Pulmonary and Sleep Medicine, School of Medicine, Namik Kemal University, Tekirdağ, Turkey
| | - Ahmet Gürel
- Department of Biochemistry, School of Medicine, Namik Kemal University, Tekirdağ, Turkey
| |
Collapse
|
18
|
|
19
|
Osteoporosis: From osteoscience to neuroscience and beyond. Mech Ageing Dev 2015; 145:26-38. [DOI: 10.1016/j.mad.2015.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 02/02/2015] [Accepted: 02/03/2015] [Indexed: 11/17/2022]
|
20
|
Bartelle BB, Mana MD, Suero-Abreu GA, Rodriguez JJ, Turnbull DH. Engineering an effective Mn-binding MRI reporter protein by subcellular targeting. Magn Reson Med 2014; 74:1750-7. [PMID: 25522343 DOI: 10.1002/mrm.25566] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/24/2014] [Accepted: 11/17/2014] [Indexed: 12/23/2022]
Abstract
PURPOSE Manganese (Mn) is an effective contrast agent and biologically active metal, which has been widely used for Mn-enhanced MRI (MEMRI). The purpose of this study was to develop and test a Mn binding protein for use as a genetic reporter for MEMRI. METHODS The bacterial Mn-binding protein, MntR was identified as a candidate reporter protein. MntR was engineered for expression in mammalian cells, and targeted to different subcellular organelles, including the Golgi Apparatus where cellular Mn is enriched. Transfected HEK293 cells and B16 melanoma cells were tested in vitro and in vivo, using immunocytochemistry, MR imaging and relaxometry. RESULTS Subcellular targeting of MntR to the cytosol, endoplasmic reticulum and Golgi apparatus was verified with immunocytochemistry. After targeting to the Golgi, MntR expression produced robust R1 changes and T1 contrast in cells, in vitro and in vivo. Co-expression with the divalent metal transporter DMT1, a previously described Mn-based reporter, further enhanced contrast in B16 cells in culture, but in the in vivo B16 tumor model tested was not significantly better than MntR alone. CONCLUSION This second-generation reporter system both expands the capabilities of genetically encoded reporters for imaging with MEMRI and provides important insights into the mechanisms of Mn biology which create endogenous MEMRI contrast.
Collapse
Affiliation(s)
- Benjamin B Bartelle
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, New York, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Miyeko D Mana
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, New York, USA.,Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Giselle A Suero-Abreu
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, New York, USA
| | - Joe J Rodriguez
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, New York, USA
| | - Daniel H Turnbull
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, New York, USA.,Departments of Radiology and Pathology, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
21
|
Evans V, Mahon MF, Webster RL. A mild, copper-catalysed amide deprotection strategy: use of tert-butyl as a protecting group. Tetrahedron 2014. [DOI: 10.1016/j.tet.2014.07.080] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
22
|
Čechová D, Martišková A, Padělková Z, Gal’a L, Dlháň L, Valigura D, Valko M, Boča R, Moncol J. Manganese(II) one-dimensional coordination polymers with nitrobenzoato or nitrosalicylato bridges: Syntheses, crystal structures, and magnetic properties. Polyhedron 2014. [DOI: 10.1016/j.poly.2014.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
23
|
McGeary RP, Schenk G, Guddat LW. The applications of binuclear metallohydrolases in medicine: Recent advances in the design and development of novel drug leads for purple acid phosphatases, metallo-β-lactamases and arginases. Eur J Med Chem 2014; 76:132-44. [DOI: 10.1016/j.ejmech.2014.02.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 01/28/2014] [Accepted: 02/06/2014] [Indexed: 10/25/2022]
|
24
|
Sutradhar M, Martins LMDRS, Guedes da Silva MFC, Alegria ECBA, Liu CM, Pombeiro AJL. Dinuclear Mn(ii,ii) complexes: magnetic properties and microwave assisted oxidation of alcohols. Dalton Trans 2014; 43:3966-77. [DOI: 10.1039/c3dt52774c] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
25
|
North ML, Grasemann H, Khanna N, Inman MD, Gauvreau GM, Scott JA. Increased ornithine-derived polyamines cause airway hyperresponsiveness in a mouse model of asthma. Am J Respir Cell Mol Biol 2013; 48:694-702. [PMID: 23470627 DOI: 10.1165/rcmb.2012-0323oc] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Up-regulation of arginase contributes to airways hyperresponsiveness (AHR) in asthma by reducing L-arginine bioavailability for the nitric oxide (NO) synthase isozymes. The product of arginase activity, L-ornithine, can be metabolized into polyamines by ornithine decarboxylase. We tested the hypothesis that increases in L-ornithine-derived polyamines contribute to AHR in mouse models of allergic airways inflammation. After measuring significantly increased polyamine levels in sputum samples from human subjects with asthma after allergen challenge, we used acute and subacute ovalbumin sensitization and challenge mouse models of allergic airways inflammation and naive mice to investigate the relationship of AHR to methacholine and polyamines in the lung. We found that spermine levels were elevated significantly in lungs from the acute model, which exhibits robust AHR, but not in the subacute murine model of asthma, which does not develop AHR. Intratracheal administration of spermine significantly augmented airways responsiveness to methacholine in both naive mice and mice with subacute airways inflammation, and reduced nitrite/nitrate levels in lung homogenates, suggesting that the AHR developed as a consequence of inhibition of constitutive NO production in the airways. Chronic inhibition of polyamine synthesis using an ornithine decarboxylase inhibitor significantly reduced polyamine levels, restored nitrite/nitrate levels to normal, and abrogated the AHR to methacholine in the acute model of allergic airways inflammation. We demonstrate that spermine increases airways responsiveness to methacholine, likely through inhibition of constitutive NO synthesis. Thus, inhibition of polyamine production may represent a new therapeutic target to treat airway obstruction in allergic asthma.
Collapse
Affiliation(s)
- Michelle L North
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
26
|
Cloots RHE, Sankaranarayanan S, de Theije CC, Poynter ME, Terwindt E, van Dijk P, Hakvoort TBM, Lamers WH, Köhler SE. Ablation of Arg1 in hematopoietic cells improves respiratory function of lung parenchyma, but not that of larger airways or inflammation in asthmatic mice. Am J Physiol Lung Cell Mol Physiol 2013; 305:L364-76. [PMID: 23831616 DOI: 10.1152/ajplung.00341.2012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Asthma is a chronic inflammatory disease of the small airways, with airway hyperresponsiveness (AHR) and inflammation as hallmarks. Recent studies suggest a role for arginase in asthma pathogenesis, possibly because arginine is the substrate for both arginase and NO synthase and because NO modulates bronchial tone and inflammation. Our objective was to investigate the importance of increased pulmonary arginase 1 expression on methacholine-induced AHR and lung inflammation in a mouse model of allergic asthma. Arginase 1 expression in the lung was ablated by crossing Arg1(fl/fl) with Tie2Cre(tg/-) mice. Mice were sensitized and then challenged with ovalbumin. Lung function was measured with the Flexivent. Adaptive changes in gene expression, chemokine and cytokine secretion, and lung histology were quantified with quantitative PCR, ELISA, and immunohistochemistry. Arg1 deficiency did not affect the allergic response in lungs and large-airway resistance, but it improved peripheral lung function (tissue elastance and resistance) and attenuated adaptive increases in mRNA expression of arginine-catabolizing enzymes Arg2 and Nos2, arginine transporters Slc7a1 and Slc7a7, chemokines Ccl2 and Ccl11, cytokines Tnfa and Ifng, mucus-associated epithelial markers Clca3 and Muc5ac, and lung content of IL-13 and CCL11. However, expression of Il4, Il5, Il10, and Il13 mRNA; lung content of IL-4, IL-5, IL-10, TNF-α, and IFN-γ protein; and lung pathology were not affected. Correlation analysis showed that Arg1 ablation disturbed the coordinated pulmonary response to ovalbumin challenges, suggesting arginine (metabolite) dependence of this response. Arg1 ablation in the lung improved peripheral lung function and affected arginine metabolism but had little effect on airway inflammation.
Collapse
Affiliation(s)
- Roy H E Cloots
- Department of Anatomy & Embryology and NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Bratt JM, Zeki AA, Last JA, Kenyon NJ. Competitive metabolism of L-arginine: arginase as a therapeutic target in asthma. J Biomed Res 2013; 25:299-308. [PMID: 23554705 PMCID: PMC3596726 DOI: 10.1016/s1674-8301(11)60041-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Revised: 06/24/2011] [Accepted: 07/21/2011] [Indexed: 12/20/2022] Open
Abstract
Exhaled breath nitric oxide (NO) is an accepted asthma biomarker. Lung concentrations of NO and its amino acid precursor, L-arginine, are regulated by the relative expressions of the NO synthase (NOS) and arginase isoforms. Increased expression of arginase I and NOS2 occurs in murine models of allergic asthma and in biopsies of asthmatic airways. Although clinical trials involving the inhibition of NO-producing enzymes have shown mixed results, small molecule arginase inhibitors have shown potential as a therapeutic intervention in animal and cell culture models. Their transition to clinical trials is hampered by concerns regarding their safety and potential toxicity. In this review, we discuss the paradigm of arginase and NOS competition for their substrate L-arginine in the asthmatic airway. We address the functional role of L-arginine in inflammation and the potential role of arginase inhibitors as therapeutics.
Collapse
Affiliation(s)
- Jennifer M Bratt
- Department of Internal Medicine, Division of Pulmonary and Critical Care and Sleep Medicine, University of California, Davis, CA 95616, USA
| | | | | | | |
Collapse
|
28
|
Nagagarajan S, Xue F, MacKerell AD. Impact of substrate protonation and tautomerization states on interactions with the active site of arginase I. J Chem Inf Model 2013; 53:452-60. [PMID: 23327293 DOI: 10.1021/ci300506y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Human arginase is a binuclear manganese metalloenzyme that participates in the urea cycle. Arginase catalyzes the hydrolysis of L-arginine into L-ornithine and urea and is linked to several disorders such as asthma and cancer. Currently, the protonation and tautomerization state of the substrate when bound to the active site, which contains two manganese ions, is not known. Knowledge of the charge-dependent behavior of arginine in the arginase I environment would be of utility toward understanding the catalytic mechanism and designing inhibitors of this enzyme. The arginine(+/0) species, including all possible neutral tautomers, were modeled using an aminoimidazole analog as template. All-atom molecular dynamics simulations were then performed on each of the charged and neutral species. In addition, a hydroxide ion was included in selected simulations to test its importance. Results show that the positively charged state of arginine is stable in the active site of arginase I, with that stabilization facilitated by the presence of hydroxide. Glu277 is indicated to play a role in stabilizing arginine in the active site and facilitating its ability to assume a catalytically competent conformation in the presence of hydroxide. The reported interactions and modeled arginine-bound arginase I structures can be used as a tool for structure-based inhibitor design, as experimental data on the structure of the substrate-enzyme complex is lacking.
Collapse
Affiliation(s)
- Shanthi Nagagarajan
- Department of Pharmaceutical Sciences, University of Maryland, School of Pharmacy, 20 Penn Street HSFII, Baltimore, Maryland 21201, USA
| | | | | |
Collapse
|
29
|
Penton PC, Wang X, Amatullah H, Cooper J, Godri K, North ML, Khanna N, Scott JA, Chow CW. Spleen tyrosine kinase inhibition attenuates airway hyperresponsiveness and pollution-induced enhanced airway response in a chronic mouse model of asthma. J Allergy Clin Immunol 2012; 131:512-20.e1-10. [PMID: 22981792 DOI: 10.1016/j.jaci.2012.07.039] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 07/24/2012] [Accepted: 07/25/2012] [Indexed: 12/31/2022]
Abstract
BACKGROUND Asthma is a chronic inflammatory disease characterized by airways hyperresponsiveness (AHR), reversible airflow obstruction, airway remodeling, and episodic exacerbations caused by air pollutants, such as particulate matter (PM; PM <2.5 μm in diameter [PM(2.5)]) and ozone (O(3)). Spleen tyrosine kinase (Syk), an immunoregulatory kinase, has been implicated in the pathogenesis of asthma. OBJECTIVE We sought to evaluate the effect of Syk inhibition on AHR in a chronic mouse model of allergic airways inflammation and pollutant exposure. METHODS We used a 12-week chronic ovalbumin (OVA) sensitization and challenge mouse model of airways inflammation followed by exposure to PM(2.5) plus O(3). Respiratory mechanics and methacholine (MCh) responsiveness were assessed by using the flexiVent system. The Syk inhibitor NVP-QAB-205 was nebulized intratracheally by using a treatment-based protocol 15 minutes before assessment of MCh responsiveness. RESULTS Syk expression increased significantly in the airway epithelia of OVA-sensitized and OVA-challenged (OVA/OVA) mice compared with OVA-sensitized but PBS-challenged (OVA/PBS) control mice. OVA/OVA mice exhibited AHR to MCh, which was attenuated by a single administration of NVP-QAB-205 (0.3 and 3 mg/kg). PM(2.5) plus O(3) significantly augmented AHR to MCh in the OVA/OVA mice, which was abrogated by NVP-QAB-205. Total inflammatory cell counts were significantly higher in the bronchoalveolar lavage fluid from OVA/OVA than OVA/PBS mice and were unaffected by PM(2.5) plus O(3) or NVP-QAB-205. CONCLUSION NVP-QAB-205 reduced AHR and the enhanced response to PM(2.5) plus O(3) to normal levels in an established model of chronic allergic airways inflammation, suggesting that Syk inhibitors have promise as a therapy for asthma.
Collapse
Affiliation(s)
- Patricia Castellanos Penton
- Division of Respirology, Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Li F, Kang Q, Shan H, Chen L, Xie J. Regioselective N-Alkylation of 2-Aminoimidazoles with Alcohols to 2-(N-Alkylamino)imidazoles Catalyzed by the [Cp*IrCl2]2/K2CO3 System. European J Org Chem 2012. [DOI: 10.1002/ejoc.201200698] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
31
|
Amatullah H, North ML, Akhtar US, Rastogi N, Urch B, Silverman FS, Chow CW, Evans GJ, Scott JA. Comparative cardiopulmonary effects of size-fractionated airborne particulate matter. Inhal Toxicol 2012; 24:161-71. [PMID: 22356274 DOI: 10.3109/08958378.2011.650235] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Strong epidemiological evidence exists linking particulate matter (PM) exposures with hospital admissions of individuals for cardiopulmonary symptoms. The PM size is important in influencing the extent of infiltration into the respiratory tract and systemic circulation and directs the differential physiological impacts. OBJECTIVE To investigate the differential effects of the quasi-ultrafine (PM(0.2)), fine (PM(0.15-2.5)), and coarse PM (PM(2.5-10)) size fractions on pulmonary and cardiac function. METHODS Female BALB/c mice were exposed to HEPA-filtered laboratory air or concentrated coarse, fine, or quasi-ultrafine PM using Harvard Ambient Particle Concentrators in conjunction with our nose-only exposure system. These exposures were conducted as part of the "Health Effects of Aerosols in Toronto (HEAT)" campaign. Following a 4 h exposure, mice underwent assessment of respiratory function and recording of electrocardiograms using the flexiVent® system. RESULTS Exposure to coarse and fine PM resulted in a significant reduction in quasistatic compliance of the lung. Baseline total respiratory resistance and maximum responsiveness to methacholine were augmented after coarse PM exposures but were not affected by quasi-ultrafine PM exposures. In contrast, quasi-ultrafine PM alone had a significant effect on heart rate and in reducing heart rate variability. CONCLUSION These findings indicate that coarse and fine PM influence lung function and airways responsiveness, while ultrafine PM can perturb cardiac function. This study supports the hypothesis that coarse and fine PM exerts its predominant physiologic effects at the site of deposition in the airways, whereas ultrafine PM likely crosses the alveolar epithelial barrier into the systemic circulation to affect cardiovascular function.
Collapse
Affiliation(s)
- Hajera Amatullah
- Division of Occupational and Environmental Health, Dalla Lana School of Public Health, University of Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Scott JA, North ML, Rafii M, Huang H, Pencharz P, Subbarao P, Belik J, Grasemann H. Asymmetric dimethylarginine is increased in asthma. Am J Respir Crit Care Med 2011; 184:779-85. [PMID: 21719758 DOI: 10.1164/rccm.201011-1810oc] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
RATIONALE Asymmetric dimethylarginine (ADMA) is an endogenous nitric oxide synthase (NOS) inhibitor that competes with L-arginine for binding to NOS. It has been suggested that ADMA contributes to inflammation, collagen deposition, nitrosative stress, and lung function in murine models. OBJECTIVES To test the hypothesis that ADMA is increased in asthma and that NOS inhibition by ADMA contributes to airways obstruction. METHODS We assessed alterations of L-arginine, ADMA, and symmetric dimethylarginine (SDMA) levels in a murine model of allergic airways inflammation using LC-tandem mass spectrometry. Based on the levels of ADMA observed in the murine model, we further tested the direct effects of nebulized inhaled ADMA on airways responsiveness in naive control mice. We also assessed alterations of L-arginine, ADMA, and SDMA in humans in adult lung specimens and sputum samples from pediatric patients with asthma. MEASUREMENTS AND MAIN RESULTS ADMA was increased in lungs from the murine model of allergic airways inflammation. Exogenous administration of ADMA to naive mice, at doses consistent with the levels observed in the allergically inflamed lungs, resulted in augmentation of the airways responsiveness to methacholine. ADMA levels were also increased in human asthma lungs and sputum samples. CONCLUSIONS ADMA levels are increased in asthma and contribute to NOS-related pathophysiology.
Collapse
Affiliation(s)
- Jeremy A Scott
- Division of Occupational Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Ilies M, Di Costanzo L, Dowling DP, Thorn KJ, Christianson DW. Binding of α,α-disubstituted amino acids to arginase suggests new avenues for inhibitor design. J Med Chem 2011; 54:5432-43. [PMID: 21728378 PMCID: PMC3150614 DOI: 10.1021/jm200443b] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Arginase is a binuclear manganese metalloenzyme that hydrolyzes L-arginine to form L-ornithine and urea, and aberrant arginase activity is implicated in various diseases such as erectile dysfunction, asthma, atherosclerosis, and cerebral malaria. Accordingly, arginase inhibitors may be therapeutically useful. Continuing our efforts to expand the chemical space of arginase inhibitor design and inspired by the binding of 2-(difluoromethyl)-L-ornithine to human arginase I, we now report the first study of the binding of α,α-disubstituted amino acids to arginase. Specifically, we report the design, synthesis, and assay of racemic 2-amino-6-borono-2-methylhexanoic acid and racemic 2-amino-6-borono-2-(difluoromethyl)hexanoic acid. X-ray crystal structures of human arginase I and Plasmodium falciparum arginase complexed with these inhibitors reveal the exclusive binding of the L-stereoisomer; the additional α-substituent of each inhibitor is readily accommodated and makes new intermolecular interactions in the outer active site of each enzyme. Therefore, this work highlights a new region of the protein surface that can be targeted for additional affinity interactions, as well as the first comparative structural insights on inhibitor discrimination between a human and a parasitic arginase.
Collapse
Affiliation(s)
- Monica Ilies
- Department of Chemistry, Drexel University, Philadelphia, PA 19104-2875, United States
| | - Luigi Di Costanzo
- RCSB Protein Data Bank, Department of Chemistry and Chemical Biology, Rutgers The State University of New Jersey, Piscataway, NJ 08854-8087, United States
| | - Daniel P. Dowling
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139-4307, United States
| | | | - David W. Christianson
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104-6323 United States
| |
Collapse
|
34
|
Ilies M, Dowling DP, Lombardi PM, Christianson DW. Synthesis of a new trifluoromethylketone analogue of l-arginine and contrasting inhibitory activity against human arginase I and histone deacetylase 8. Bioorg Med Chem Lett 2011; 21:5854-8. [PMID: 21875805 DOI: 10.1016/j.bmcl.2011.07.100] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 07/25/2011] [Accepted: 07/26/2011] [Indexed: 01/12/2023]
Abstract
As part of our continuing search for new amino acid inhibitors of metalloenzymes, we now report the synthesis and biological evaluation of the trifluoromethylketone analogue of L-arginine, (S)-2-amino-8,8,8-trifluoro-7-oxo-octanoic acid (10). While this novel amino acid was initially designed as a potential inhibitor of human arginase I, it exhibits no measurable inhibitory activity against this enzyme. Surprisingly, however, 10 is a potent inhibitor of human histone deacetylase 8, with IC(50)=1.5 ± 0.2 μM. Additionally, 10 weakly inhibits the related bacterial enzyme, acetylpolyamine amidohydrolase, with IC(50)=110 ± 30 μM. The lack of inhibitory activity against human arginase I may result from unfavorable interactions of the bulky trifluoromethyl group of 10 in the constricted active site. Since the active site of histone deacetylase 8 is less constricted, we hypothesize that it accommodates 10 as the gem-diol, which mimics the tetrahedral intermediate and its flanking transition states in catalysis. Therefore, we suggest that 10 represents a new lead in the design of an amino acid or peptide-based inhibitor of histone deacetylases with simpler structure than previously studied trifluoromethylketones.
Collapse
Affiliation(s)
- Monica Ilies
- Department of Chemistry, Drexel University, Philadelphia, PA 19104-2875, USA
| | | | | | | |
Collapse
|
35
|
Grasemann H, Al-Saleh S, Scott JA, Shehnaz D, Mehl A, Amin R, Rafii M, Pencharz P, Belik J, Ratjen F. Asymmetric Dimethylarginine Contributes to Airway Nitric Oxide Deficiency in Patients with Cystic Fibrosis. Am J Respir Crit Care Med 2011; 183:1363-8. [DOI: 10.1164/rccm.201012-1995oc] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
36
|
Newman J, Pearce L, Lesburg CA, Strickland C, Peat TS. Crystallization of an apo form of human arginase: using all the tools in the toolbox simultaneously. Acta Crystallogr Sect F Struct Biol Cryst Commun 2011; 67:90-3. [PMID: 21206033 PMCID: PMC3079981 DOI: 10.1107/s1744309110046208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Accepted: 11/09/2010] [Indexed: 11/10/2022]
Abstract
Arginase (EC 3.5.3.1) is an aminohydrolase that acts on L-arginine to produce urea and ornithine. Two isotypes of the enzyme are found in humans. Type I is predominantly produced in the liver and is a homotrimer of 35 kDa subunits. Human arginase (hArginase) I is seen to be up-regulated in many diseases and is a potential therapeutic target for many diverse indications. Previous reports of crystallization and structure determination of hArginase have always included inhibitors of the enzyme: here, the first case of a true apo crystal form of the enzyme which is suitable for small-molecule soaking is reported. The crystals belonged to space group P2(1)2(1)2(1) and have approximate unit-cell parameters a=53, b=67.5, c=250 Å. The crystals showed slightly anisotropic diffraction to beyond 2.0 Å resolution.
Collapse
Affiliation(s)
- Janet Newman
- Materials Science and Engineering, CSIRO, 343 Royal Parade, Parkville, VIC 3052, Australia.
| | | | | | | | | |
Collapse
|
37
|
Stone EM, Chantranupong L, Georgiou G. The second-shell metal ligands of human arginase affect coordination of the nucleophile and substrate. Biochemistry 2010; 49:10582-8. [PMID: 21053939 PMCID: PMC2998210 DOI: 10.1021/bi101542t] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
![]()
The active sites of eukaryotic arginase enzymes are strictly conserved, especially the first- and second-shell ligands that coordinate the two divalent metal cations that generate a hydroxide molecule for nucleophilic attack on the guanidinium carbon of l-arginine and the subsequent production of urea and l-ornithine. Here by using comprehensive pairwise saturation mutagenesis of the first- and second-shell metal ligands in human arginase I, we demonstrate that several metal binding ligands are actually quite tolerant to amino acid substitutions. Of >2800 double mutants of first- and second-shell residues analyzed, we found more than 80 unique amino acid substitutions, of which four were in first-shell residues. Remarkably, certain second-shell mutations could modulate the binding of both the nucleophilic water/hydroxide molecule and substrate or product ligands, resulting in activity greater than that of the wild-type enzyme. The data presented here constitute the first comprehensive saturation mutagenesis analysis of a metallohydrolase active site and reveal that the strict conservation of the second-shell metal binding residues in eukaryotic arginases does not reflect kinetic optimization of the enzyme during the course of evolution.
Collapse
Affiliation(s)
- Everett M Stone
- Department of Chemical Engineering, University of Texas, Austin, Texas 78712, United States
| | | | | |
Collapse
|