1
|
Rebai R, Jasmin L, Boudah A. Identification of Two Flavonoids as New and Safe Inhibitors of Kynurenine Aminotransferase II via Computational and In Vitro Study. Pharmaceuticals (Basel) 2025; 18:76. [PMID: 39861140 PMCID: PMC11768104 DOI: 10.3390/ph18010076] [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: 12/11/2024] [Revised: 01/01/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Kynurenine aminotransferase II (KAT-II) is a target for treating several diseases characterized by an excess of kynurenic acid (KYNA). Although KAT-II inactivators are available, they often lead to adverse side effects due to their irreversible inhibition mechanism. This study aimed to identify potent and safe inhibitors of KAT-II using computational and in vitro approaches. Methods: Virtual screening, MM/GBSA, and molecular dynamics simulations were conducted to identify the top drug candidates, followed by kinetic measurements and in vitro cytotoxicity evaluation. Results: The study showed that two compounds, herbacetin and (-)-Epicatechin exhibited the best scores. Their Glide docking scores are -8.66 kcal/mol and -8.16 kcal/mol, respectively, and their MM/GBSA binding energies are -50.30 kcal/mol and -51.35 kcal/mol, respectively. These scores are superior to those of the standard inhibitor, PF-04859989, which has docking scores of -7.12 kcal/mol and binding energy of -38.41 kcal/mol. ADMET analysis revealed that the selected compounds have favorable pharmacokinetic parameters, moderate bioavailability, and a safe toxicity profile, which supports their potential use. Further, the kinetic study showed that herbacetin and (-)-Epicatechin are reversible KAT-II inhibitors and exhibit a competitive inhibition mechanism. Their half-maximal inhibitory concentrations (IC50) are 5.98 ± 0.18 µM and 8.76 ± 0.76 µM, respectively. The MTT assay for cell toxicity indicated that the two compounds do not affect HepG2 cell viability at the necessary concentration for KAT-II inhibition. Conclusions: These results suggest that herbacetin and (-)-Epicatechin are suitable for KAT-II inhibition and are promising candidates for further development of KAT-II inhibitors.
Collapse
Affiliation(s)
- Redouane Rebai
- Department of Natural and Life Sciences, Faculty of Exact Sciences and Natural and Life Sciences, University Mohamed Khider of Biskra, BP145 RP, Biskra 07000, Algeria
- Laboratory of Biotechnology, National Higher School of Biotechnology, Ville Universitaire (University of Constantine 3), Ali Mendjeli, BP E66, Constantine 25100, Algeria
| | - Luc Jasmin
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco, 707 Parnassus Ave Suite D-1201, San Francisco, CA 94143, USA;
| | - Abdennacer Boudah
- Laboratory of Biotechnology, National Higher School of Biotechnology, Ville Universitaire (University of Constantine 3), Ali Mendjeli, BP E66, Constantine 25100, Algeria
| |
Collapse
|
2
|
Shirley JD, Gillingham JR, Nauta KM, Diwakar S, Carlson EE. kinact/ KI Value Determination for Penicillin-Binding Proteins in Live Cells. ACS Infect Dis 2024; 10:4137-4145. [PMID: 39628314 DOI: 10.1021/acsinfecdis.4c00370] [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] [Indexed: 12/11/2024]
Abstract
Penicillin-binding proteins (PBPs) are an essential family of bacterial enzymes that are covalently inhibited by the β-lactam class of antibiotics. PBP inhibition disrupts peptidoglycan biosynthesis, which results in deficient growth and proliferation, and ultimately leads to lysis. IC50 values are often employed as descriptors of enzyme inhibition and inhibitor selectivity, but can be misleading in the study of time-dependent, covalent inhibitors. Due to this disconnect, the second-order rate constant, kinact/KI, is a more appropriate metric of covalent-inhibitor potency. Despite being the gold standard measurement of potency, kinact/KI values are typically obtained from in vitro assays, which limits assay throughput if investigating an enzyme family with multiple homologues (such as the PBPs). Therefore, we developed a whole-cell kinact/KI assay to define inhibitor potency for the PBPs in Streptococcus pneumoniae using the fluorescent, activity-based probe, Bocillin-FL. Our results align with in vitro kinact/KI data and show a comparable relationship to previously established IC50 values. These results support the validity of our in vivo kinact/KI method as a means of obtaining β-lactam potency for a suite of PBPs to enable structure-activity relationship studies.
Collapse
Affiliation(s)
- Joshua D Shirley
- Department of Medicinal Chemistry, University of Minnesota, 208 Harvard Street SE, Minneapolis, Minnesota 55454, United States
| | - Jacob R Gillingham
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, 321 Church St SE, Minneapolis, Minnesota 55454, United States
| | - Kelsie M Nauta
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Shivani Diwakar
- Department of Pharmacology, University of Minnesota, 321 Church St SE, Minneapolis, Minnesota 55454, United States
| | - Erin E Carlson
- Department of Medicinal Chemistry, University of Minnesota, 208 Harvard Street SE, Minneapolis, Minnesota 55454, United States
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, 321 Church St SE, Minneapolis, Minnesota 55454, United States
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
- Department of Pharmacology, University of Minnesota, 321 Church St SE, Minneapolis, Minnesota 55454, United States
| |
Collapse
|
3
|
Oxenkrug G. Anthranilic Acid-G-Protein Coupled Receptor109A-Cytosolic Phospholipase A2-Myelin-Cognition Cascade: A New Target for the Treatment/Prevention of Cognitive Impairment in Schizophrenia, Dementia, and Aging. Int J Mol Sci 2024; 25:13269. [PMID: 39769034 PMCID: PMC11675959 DOI: 10.3390/ijms252413269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Cognitive impairment is a core feature of neurodevelopmental (schizophrenia) and aging-associated (mild cognitive impairment and Alzheimer's dementia) neurodegenerative diseases. Limited efficacy of current pharmacological treatments warrants further search for new targets for nootropic interventions. The breakdown of myelin, a phospholipids axonal sheath that protects the conduction of nerve impulse between neurons, was proposed as a neuropathological abnormality that precedes and promotes the deposition of amyloid-β in neuritic plaques. The present review of the recent literature and our own pre- and clinical data suggest (for the first time) that the anthranilic acid (AA)-induced activation of microglial-expressed G-protein coupled receptor (GPR109A) inhibits cytosolic phospholipase A2 (cPLA2), an enzyme that triggers the degradation of myelin and consequently attenuates cognitive impairment. The present review suggests that the up-regulation of AA formation is a sex-specific compensatory (adaptive) reaction aimed to prevent/treat cognitive impairment. The AA-GPR109A-cPLA2-myelin-cognition cascade suggests new nootropic interventions, e.g., the administration of pegylated kynureninase, an enzyme that catalyzes AA formation from Kynurenine (Kyn), a tryptophane catabolite; pegylated interferon-alpha; central and peripheral Kyn aminotransferase inhibitors that increase availability of Kyn as a substrate for AA formation; and vagus nerve stimulation. The cascade predicts nootropic activity of exogenous GPR109A agonists that were designed and underwent clinical trials (unsuccessful) as anti-dyslipidemia agents. The proposed cascade might contribute to the pathogenesis of cognitive impairment. Data on AA in neurodegenerative disorders are scarce, and the proposed cascade needs further exploration in pre- and clinical studies.
Collapse
Affiliation(s)
- Gregory Oxenkrug
- Department of Psychiatry, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
4
|
Pocivavsek A, Schwarcz R, Erhardt S. Neuroactive Kynurenines as Pharmacological Targets: New Experimental Tools and Exciting Therapeutic Opportunities. Pharmacol Rev 2024; 76:978-1008. [PMID: 39304346 PMCID: PMC11549936 DOI: 10.1124/pharmrev.124.000239] [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: 07/07/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
Both preclinical and clinical studies implicate functional impairments of several neuroactive metabolites of the kynurenine pathway (KP), the major degradative cascade of the essential amino acid tryptophan in mammals, in the pathophysiology of neurologic and psychiatric diseases. A number of KP enzymes, such as tryptophan 2,3-dioxygenase (TDO2), indoleamine 2,3-dioxygenases (IDO1 and IDO2), kynurenine aminotransferases (KATs), kynurenine 3-monooxygenase (KMO), 3-hydroxyanthranilic acid oxygenase (3-HAO), and quinolinic acid phosphoribosyltransferase (QPRT), control brain KP metabolism in health and disease and are therefore increasingly considered to be promising targets for the treatment of disorders of the nervous system. Understanding the distribution, cellular expression, and regulation of KP enzymes and KP metabolites in the brain is therefore critical for the conceptualization and implementation of successful therapeutic strategies. SIGNIFICANCE STATEMENT: Studies have implicated the kynurenine pathway of tryptophan in the pathophysiology of neurologic and psychiatric diseases. Key enzymes of the kynurenine pathway regulate brain metabolism in both health and disease, making them promising targets for treating these disorders. Therefore, understanding the distribution, cellular expression, and regulation of these enzymes and metabolites in the brain is critical for developing effective therapeutic strategies. This review endeavors to describe these processes in detail.
Collapse
Affiliation(s)
- Ana Pocivavsek
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina (A.P.); Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland (R.S.); and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.E.)
| | - Robert Schwarcz
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina (A.P.); Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland (R.S.); and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.E.)
| | - Sophie Erhardt
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina (A.P.); Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland (R.S.); and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.E.)
| |
Collapse
|
5
|
Alves LDF, Moore JB, Kell DB. The Biology and Biochemistry of Kynurenic Acid, a Potential Nutraceutical with Multiple Biological Effects. Int J Mol Sci 2024; 25:9082. [PMID: 39201768 PMCID: PMC11354673 DOI: 10.3390/ijms25169082] [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: 07/19/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Kynurenic acid (KYNA) is an antioxidant degradation product of tryptophan that has been shown to have a variety of cytoprotective, neuroprotective and neuronal signalling properties. However, mammalian transporters and receptors display micromolar binding constants; these are consistent with its typically micromolar tissue concentrations but far above its serum/plasma concentration (normally tens of nanomolar), suggesting large gaps in our knowledge of its transport and mechanisms of action, in that the main influx transporters characterized to date are equilibrative, not concentrative. In addition, it is a substrate of a known anion efflux pump (ABCC4), whose in vivo activity is largely unknown. Exogeneous addition of L-tryptophan or L-kynurenine leads to the production of KYNA but also to that of many other co-metabolites (including some such as 3-hydroxy-L-kynurenine and quinolinic acid that may be toxic). With the exception of chestnut honey, KYNA exists at relatively low levels in natural foodstuffs. However, its bioavailability is reasonable, and as the terminal element of an irreversible reaction of most tryptophan degradation pathways, it might be added exogenously without disturbing upstream metabolism significantly. Many examples, which we review, show that it has valuable bioactivity. Given the above, we review its potential utility as a nutraceutical, finding it significantly worthy of further study and development.
Collapse
Affiliation(s)
- Luana de Fátima Alves
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - J. Bernadette Moore
- School of Food Science & Nutrition, University of Leeds, Leeds LS2 9JT, UK;
- Department of Biochemistry, Cell & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
| | - Douglas B. Kell
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
- Department of Biochemistry, Cell & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
| |
Collapse
|
6
|
Shirley JD, Nauta KM, Gillingham JR, Diwakar S, Carlson EE. kinact / KI Value Determination for Penicillin-Binding Proteins in Live Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.05.592586. [PMID: 38746240 PMCID: PMC11092749 DOI: 10.1101/2024.05.05.592586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Penicillin-binding proteins (PBPs) are an essential family of bacterial enzymes that are inhibited by the β-lactam class of antibiotics. PBP inhibition disrupts cell wall biosynthesis, which results in deficient growth and proliferation, and ultimately leads to lysis. IC 50 values are often employed as descriptors of enzyme inhibition and inhibitor selectivity but can be misleading in the study of time-dependent, irreversible inhibitors. Due to this disconnect, the second order rate constant k inact / K I is a more appropriate metric of covalent inhibitor potency. Despite being the gold standard measurement of potency, k inact / K I values are typically obtained from in vitro assays, which limits assay throughput if investigating an enzyme family with multiple homologs (such as the PBPs). Therefore, we developed a whole-cell k inact / K I assay to define inhibitor potency for the PBPs in Streptococcus pneumoniae using the fluorescent activity-based probe Bocillin-FL. Our results align with in vitro k inact / K I data and show a comparable relationship to previously established IC 50 values. These results support the validity of our in vivo k inact / K I method as a means of obtaining a full picture of β-lactam potency for a suite of PBPs. Abstract Figure
Collapse
|
7
|
Liang X, Du W, Huang L, Xiang L, Pan W, Yang F, Zheng F, Xie Y, Geng L, Gong S, Xu W. Helicobacter pylori promotes gastric intestinal metaplasia through activation of IRF3-mediated kynurenine pathway. Cell Commun Signal 2023; 21:141. [PMID: 37328804 PMCID: PMC10273570 DOI: 10.1186/s12964-023-01162-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 05/07/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Metabolic reprogramming is a critical event for cell fate and function, making it an attractive target for clinical therapy. The function of metabolic reprogramming in Helicobacter pylori (H. pylori)-infected gastric intestinal metaplasia remained to be identified. METHODS Xanthurenic acid (XA) was measured in gastric cancer cells treated with H. pylori or H. pylori virulence factor, respectively, and qPCR and WB were performed to detect CDX2 and key metabolic enzymes expression. A subcellular fractionation approach, luciferase and ChIP combined with immunofluorescence were applied to reveal the mechanism underlying H. pylori mediated kynurenine pathway in intestinal metaplasia in vivo and in vitro. RESULTS Herein, we, for the first time, demonstrated that H. pylori contributed to gastric intestinal metaplasia characterized by enhanced Caudal-related homeobox transcription factor-2 (CDX2) and mucin2 (MUC2) expression, which was attributed to activation of kynurenine pathway. H. pylori promoted kynurenine aminotransferase II (KAT2)-mediated kynurenine pathway of tryptophan metabolism, leading to XA production, which further induced CDX2 expression in gastric epithelial cells. Mechanically, H. pylori activated cyclic guanylate adenylate synthase (cGAS)-interferon regulatory factor 3 (IRF3) pathway in gastric epithelial cells, leading to enhance IRF3 nuclear translocation and the binding of IRF3 to KAT2 promoter. Inhibition of KAT2 could significantly reverse the effect of H. pylori on CDX2 expression. Also, the rescue phenomenon was observed in gastric epithelial cells treated with H. pylori after IRF3 inhibition in vitro and in vivo. Most importantly, phospho-IRF3 was confirmed to be a clinical positive relationship with CDX2. CONCLUSION These finding suggested H. pylori contributed to gastric intestinal metaplasia through KAT2-mediated kynurenine pathway of tryptophan metabolism via cGAS-IRF3 signaling, targeting the kynurenine pathway could be a promising strategy to prevent gastric intestinal metaplasia caused by H. pylori infection. Video Abstract.
Collapse
Affiliation(s)
- Xinhua Liang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wenjun Du
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China
| | - Ling Huang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China
| | - Li Xiang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China
- Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wenxu Pan
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China
| | - Fangying Yang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China
| | - Fengfeng Zheng
- Department of Infectious Diseases, The Affiliated Hospital of Putian University, Putian, 351100, China
| | - Yongwu Xie
- Department of Hematology, Zhuhai Center for Maternal and Child Health Care, Zhuhai, China
| | - Lanlan Geng
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China.
- Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Sitang Gong
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China.
- Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Wanfu Xu
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China.
- Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, 510623, China.
| |
Collapse
|
8
|
Chen J, Lin X, Xu F, Chai K, Ren M, Yu Z, Su W, Liu F. An Efficient Continuous Flow Synthesis for the Preparation of N-Arylhydroxylamines: Via a DMAP-Mediated Hydrogenation Process. Molecules 2023; 28:molecules28072968. [PMID: 37049731 PMCID: PMC10096002 DOI: 10.3390/molecules28072968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
The selective hydrogenation of nitroarenes to N-arylhydroxylamines is an important synthetic process in the chemical industry. It is commonly accomplished by using heterogeneous catalytic systems that contain inhibitors, such as DMSO. Herein, DMAP has been identified as a unique additive for increasing hydrogenation activity and product selectivity (up to >99%) under mild conditions in the Pt/C-catalyzed process. Continuous-flow technology has been explored as an efficient approach toward achieving the selective hydrogenation of nitroarenes to N-arylhydroxylamines. The present flow protocol was applied for a vast substrate scope and was found to be compatible with a wide range of functional groups, such as electron-donating groups, carbonyl, and various halogens. Further studies were attempted to show that the improvement in the catalytic activity and selectivity benefited from the dual functions of DMAP; namely, the heterolytic H2 cleavage and competitive adsorption.
Collapse
Affiliation(s)
- Jianli Chen
- College of New Materials Engineering, Jiaxing Nanhu University, Jiaxing 314000, China
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
- Correspondence: (J.C.); (F.L.)
| | - Xinyu Lin
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Feng Xu
- Raybow (Hangzhou) Pharmaceutical Co., Ltd., Hangzhou 310014, China
| | - Kejie Chai
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Minna Ren
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Zhiqun Yu
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Weike Su
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Fengfan Liu
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
- Correspondence: (J.C.); (F.L.)
| |
Collapse
|
9
|
Shen H, Xu X, Bai Y, Wang X, Wu Y, Zhong J, Wu Q, Luo Y, Shang T, Shen R, Xi M, Sun H. Therapeutic potential of targeting kynurenine pathway in neurodegenerative diseases. Eur J Med Chem 2023; 251:115258. [PMID: 36917881 DOI: 10.1016/j.ejmech.2023.115258] [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: 01/25/2023] [Revised: 02/17/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023]
Abstract
Kynurenine pathway (KP), the primary pathway of L-tryptophan (Trp) metabolism in mammals, contains several neuroactive metabolites such as kynurenic acid (KA) and quinolinic acid (QA). Its imbalance involved in aging and neurodegenerative diseases (NDs) has attracted much interest in therapeutically targeting KP enzymes and KP metabolite-associated receptors, especially kynurenine monooxygenase (KMO). Currently, many agents have been discovered with significant improvement in animal models but only one aryl hydrocarbon receptor (AHR) agonist 30 (laquinimod) has entered clinical trials for treating Huntington's disease (HD). In this review, we describe neuroactive KP metabolites, discuss the dysregulation of KP in aging and NDs and summarize the development of KP regulators in preclinical and clinical studies, offering an outlook of targeting KP for NDs treatment in future.
Collapse
Affiliation(s)
- Hualiang Shen
- Zhejiang Engineering Research Center of Fat-soluble Vitamin, Shaoxing University, Shaoxing, 312000, China; College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing, 312000, China
| | - Xinde Xu
- Zhejiang Medicine Co. Ltd., Shaoxing, 312500, China
| | - Yalong Bai
- Zhejiang Medicine Co. Ltd., Shaoxing, 312500, China
| | | | - Yibin Wu
- College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing, 312000, China
| | - Jia Zhong
- College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing, 312000, China
| | - Qiyi Wu
- College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing, 312000, China
| | - Yanjuan Luo
- Zhejiang Engineering Research Center of Fat-soluble Vitamin, Shaoxing University, Shaoxing, 312000, China; College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing, 312000, China
| | - Tianbo Shang
- Zhejiang Engineering Research Center of Fat-soluble Vitamin, Shaoxing University, Shaoxing, 312000, China; College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing, 312000, China
| | - Runpu Shen
- Zhejiang Engineering Research Center of Fat-soluble Vitamin, Shaoxing University, Shaoxing, 312000, China; College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing, 312000, China
| | - Meiyang Xi
- Zhejiang Engineering Research Center of Fat-soluble Vitamin, Shaoxing University, Shaoxing, 312000, China; College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing, 312000, China.
| | - Haopeng Sun
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
10
|
Intson K, Geissah S, McCullumsmith RE, Ramsey AJ. A role for endothelial NMDA receptors in the pathophysiology of schizophrenia. Schizophr Res 2022; 249:63-73. [PMID: 33189520 PMCID: PMC11740474 DOI: 10.1016/j.schres.2020.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022]
Abstract
Numerous genetic and postmortem studies link N-methyl-d-aspartate receptor (NMDAR) dysfunction with schizophrenia, forming the basis of the popular glutamate hypothesis. Neuronal NMDAR abnormalities are consistently reported from both basic and clinical experiments, however, non-neuronal cells also contain NMDARs, and are rarely, if ever, considered in the discussion of glutamate action in schizophrenia. We offer an examination of recent discoveries elucidating the actions and consequences of NMDAR activation in the neuroendothelium. While there has been mixed literature regarding blood flow alterations in the schizophrenia brain, in this review, we posit that some common findings may be explained by neuroendothelial NMDAR dysfunction. In particular, we emphasize that endothelial NMDARs are key mediators of neurovascular coupling, where increased neuronal activity leads to increased blood flow. Based on the broad conclusions that hypoperfusion is a neuroanatomical finding in schizophrenia, we discuss potential mechanisms by which endothelial NMDARs contribute to this disorder. We propose that endothelial NMDAR dysfunction can be a primary cause of neurovascular abnormalities in schizophrenia. Importantly, functional MRI studies using BOLD signal as a proxy for neuron activity should be considered in a new light if neurovascular coupling is impaired in schizophrenia. This review is the first to propose that NMDARs in non-excitable cells play a role in schizophrenia.
Collapse
Affiliation(s)
- Katheron Intson
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Salma Geissah
- Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Amy J Ramsey
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
11
|
Exercised accelerated the production of muscle-derived kynurenic acid in skeletal muscle and alleviated the postmenopausal osteoporosis through the Gpr35/NFκB p65 pathway. J Orthop Translat 2022; 35:1-12. [PMID: 35846727 PMCID: PMC9260440 DOI: 10.1016/j.jot.2022.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/22/2022] [Accepted: 03/09/2022] [Indexed: 01/06/2023] Open
Abstract
Background Reduced serum estrogen levels in postmenopausal patients not only aggravate bone loss but also impact myokine secretion. Emerging evidence has revealed the importance of myokines in bone metabolism, and exercise can interfere with the secretion of myokines. However, few studies have explored the impact of exercise on myokine secretion in the postmenopausal osteoporosis (PMOP) process. Methods Ten-weeks-old C57B/L6 female mice were used for constructing the postmenopausal osteoporosis model. The expression levels of kynurenine aminotransferases (Kats) were detected by RT-PCR and Western Blot. The concentration of serum kynurenic acid (Kyna) was detected by HPLC-MS. Micro-CT analysis was used for determine the changes of bone mineral density and the microstructure. The primary osteoblast and osteoclast were isolated from mice to determine the effect and mechanism of Kyna on the bone formation and resorption. Results In our research, we found a lower serum level of muscle-derived kynurenic acid (Kyna) in PMOP model mice, accompanied by a decreased level of kynurenine aminotransferases (Kats) in the gastrocnemius muscle. Moreover, treadmill-running exercise upregulated the muscle levels of KATs and increased the serum concentration of Kyna, which was positively correlated with the alleviation of bone loss. Furthermore, we found that exogenous Kyna treatment alleviated bone mineral loss and microstructure destruction in PMOP mice by inhibiting osteoclast maturation and increasing osteoblast viability. Mechanistically, we observed that Kyna reduced the NFκB p65 phosphorylation level by activating the Gpr35 receptor, which inhibited NFATc1 expression in osteoclasts and upregulated Runx2 expression in osteoblasts. Conclusion Our results revealed that the muscle levels of Kats and serum level of Kyna were negatively correlated with the severity of PMOP. Exercise intervention and exogenous Kyna treatment alleviated the impairment of bone microstructure through the Gpr35 receptor, paving the way for a novel therapeutic intervention in PMOP. The Translational potential of this article This study provides evidences that Kyna could increase the osteoblastgenesis and inhibit the osteoclastgenesis, which could be a novel therapeutic approach for osteoporosis treatment.
Collapse
|
12
|
Shou J, Feng G, He JY, Bai QF, Li X. Photoredox One-Pot Synthesis of 3,4-Dihydroquinolin-2(1H)-ones. Synlett 2022. [DOI: 10.1055/s-0041-1737910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
AbstractA photoredox one-pot strategy for efficient accessing 3,4-dihydroquinolin-2(1H)-ones from anilines, oxalyl chloride, and electron-deficient alkenes is disclosed. The new approach features excellent synthetic efficiency, readily available starting materials, and simple operations. It is compatible with a variety of anilines and electron-deficient alkenes. A broad array of 3,4-dihydroquinolin-2(1H)-ones were prepared.
Collapse
Affiliation(s)
- Jianxin Shou
- Office of Science and Technology, Shaoxing University
| | - Gaofeng Feng
- Zhejiang Key Laboratory of Alternative Technologies for Fine Chemicals Process, Shaoxing University
| | - Jing-Yao He
- Zhejiang Key Laboratory of Alternative Technologies for Fine Chemicals Process, Shaoxing University
| | - Qi-Fan Bai
- Zhejiang Key Laboratory of Alternative Technologies for Fine Chemicals Process, Shaoxing University
| | - Xuewen Li
- Zhejiang Key Laboratory of Alternative Technologies for Fine Chemicals Process, Shaoxing University
| |
Collapse
|
13
|
Maryška M, Svobodová L, Dehaen W, Hrabinová M, Rumlová M, Soukup O, Kuchař M. Heterocyclic Cathinones as Inhibitors of Kynurenine Aminotransferase II-Design, Synthesis, and Evaluation. Pharmaceuticals (Basel) 2021; 14:ph14121291. [PMID: 34959692 PMCID: PMC8708382 DOI: 10.3390/ph14121291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/24/2021] [Accepted: 12/08/2021] [Indexed: 11/16/2022] Open
Abstract
Kynurenic acid is a neuroprotective metabolite of tryptophan formed by kynurenine aminotransferase (KAT) catalyzed transformation of kynurenine. However, its high brain levels are associated with cognitive deficit and with the pathophysiology of schizophrenia. Although several classes of KAT inhibitors have been published, the search for new inhibitor chemotypes is crucial for the process of finding suitable clinical candidates. Therefore, we used pharmacophore modeling and molecular docking, which predicted derivatives of heterocyclic amino ketones as new potential irreversible inhibitors of kynurenine aminotransferase II. Thiazole and triazole-based amino ketones were synthesized within a SAR study and their inhibitory activities were evaluated in vitro. The observed activities confirmed our computational model and, moreover, the best compounds showed sub-micromolar inhibitory activity with 2-alaninoyl-5-(4-fluorophenyl)thiazole having IC50 = 0.097 µM.
Collapse
Affiliation(s)
- Michal Maryška
- Forensic Laboratory of Biologically Active Substances, Department of Chemistry of Natural Compounds, Faculty of Food and Biochemical Technology, University of Chemistry and Technology Prague, Technická 5, 16628 Prague, Czech Republic; (M.M.); (L.S.)
- National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
| | - Lucie Svobodová
- Forensic Laboratory of Biologically Active Substances, Department of Chemistry of Natural Compounds, Faculty of Food and Biochemical Technology, University of Chemistry and Technology Prague, Technická 5, 16628 Prague, Czech Republic; (M.M.); (L.S.)
| | - Wim Dehaen
- CZ-OPENSCREEN: National Infrastructure for Chemical Biology, Department of Informatics and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology Prague, Technická 5, 16628 Prague, Czech Republic;
| | - Martina Hrabinová
- Biomedical Research Center, University Hospital Hradec Králové, Sokolská 581, 50005 Hradec Kralové, Czech Republic; (M.H.); (O.S.)
- Department of Toxicology and Military Pharmacy, University of Defense, Třebešská 1575, 50005 Hradec Králové, Czech Republic
| | - Michaela Rumlová
- Department of Biotechnology, Faculty of Food and Biochemical Technology, University of Chemistry and Technology Prague, Technická 5, 16628 Prague, Czech Republic;
| | - Ondřej Soukup
- Biomedical Research Center, University Hospital Hradec Králové, Sokolská 581, 50005 Hradec Kralové, Czech Republic; (M.H.); (O.S.)
- Department of Toxicology and Military Pharmacy, University of Defense, Třebešská 1575, 50005 Hradec Králové, Czech Republic
| | - Martin Kuchař
- Forensic Laboratory of Biologically Active Substances, Department of Chemistry of Natural Compounds, Faculty of Food and Biochemical Technology, University of Chemistry and Technology Prague, Technická 5, 16628 Prague, Czech Republic; (M.M.); (L.S.)
- National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
- Correspondence: ; Tel.: +420-220-444-431
| |
Collapse
|
14
|
Neef J, Palacios DS. Progress in mechanistically novel treatments for schizophrenia. RSC Med Chem 2021; 12:1459-1475. [PMID: 34671731 PMCID: PMC8459322 DOI: 10.1039/d1md00096a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/18/2021] [Indexed: 11/21/2022] Open
Abstract
Currently available pharmacological treatments for schizophrenia derive their activity mainly by directly modulating the D2 receptor. This mode of action can alleviate the positive symptoms of schizophrenia but do not address the negative or cognitive symptoms of the disease and carry a heavy side effect burden that leads to high levels of patient non-compliance. Novel mechanisms to treat the positive symptoms of schizophrenia with improved tolerability, as well as medicines to treat negative and cognitive symptoms are urgently required. Recent efforts to identify small molecules for schizophrenia with non-D2 mechanisms will be highlighted, with a focus on those that have reached clinical development. Finally, the potential for disease modifying treatments for schizophrenia will also be discussed.
Collapse
Affiliation(s)
- James Neef
- Novartis Institutes for BioMedical Research Inc 22 Windsor St Cambridge MA 02139 USA
| | - Daniel S Palacios
- Novartis Institutes for BioMedical Research Inc 22 Windsor St Cambridge MA 02139 USA
| |
Collapse
|
15
|
Büki A, Kekesi G, Horvath G, Vécsei L. A Potential Interface between the Kynurenine Pathway and Autonomic Imbalance in Schizophrenia. Int J Mol Sci 2021; 22:10016. [PMID: 34576179 PMCID: PMC8467675 DOI: 10.3390/ijms221810016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023] Open
Abstract
Schizophrenia is a neuropsychiatric disorder characterized by various symptoms including autonomic imbalance. These disturbances involve almost all autonomic functions and might contribute to poor medication compliance, worsened quality of life and increased mortality. Therefore, it has a great importance to find a potential therapeutic solution to improve the autonomic disturbances. The altered level of kynurenines (e.g., kynurenic acid), as tryptophan metabolites, is almost the most consistently found biochemical abnormality in schizophrenia. Kynurenic acid influences different types of receptors, most of them involved in the pathophysiology of schizophrenia. Only few data suggest that kynurenines might have effects on multiple autonomic functions. Publications so far have discussed the implication of kynurenines and the alteration of the autonomic nervous system in schizophrenia independently from each other. Thus, the coupling between them has not yet been addressed in schizophrenia, although their direct common points, potential interfaces indicate the consideration of their interaction. The present review gathers autonomic disturbances, the impaired kynurenine pathway in schizophrenia, and the effects of kynurenine pathway on autonomic functions. In the last part of the review, the potential interaction between the two systems in schizophrenia, and the possible therapeutic options are discussed.
Collapse
Affiliation(s)
- Alexandra Büki
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., H-6720 Szeged, Hungary; (A.B.); (G.K.); (G.H.)
| | - Gabriella Kekesi
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., H-6720 Szeged, Hungary; (A.B.); (G.K.); (G.H.)
| | - Gyongyi Horvath
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., H-6720 Szeged, Hungary; (A.B.); (G.K.); (G.H.)
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6., H-6725 Szeged, Hungary
- MTA-SZTE Neuroscience Research Group, H-6725 Szeged, Hungary
- Interdisciplinary Excellence Center, Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6., H-6725 Szeged, Hungary
| |
Collapse
|
16
|
Marszalek-Grabska M, Walczak K, Gawel K, Wicha-Komsta K, Wnorowska S, Wnorowski A, Turski WA. Kynurenine emerges from the shadows – Current knowledge on its fate and function. Pharmacol Ther 2021; 225:107845. [DOI: 10.1016/j.pharmthera.2021.107845] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022]
|
17
|
Bai MY, Lovejoy DB, Guillemin GJ, Kozak R, Stone TW, Koola MM. Galantamine-Memantine Combination and Kynurenine Pathway Enzyme Inhibitors in the Treatment of Neuropsychiatric Disorders. Complex Psychiatry 2021; 7:19-33. [PMID: 35141700 PMCID: PMC8443947 DOI: 10.1159/000515066] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/04/2021] [Indexed: 12/25/2022] Open
Abstract
The kynurenine pathway (KP) is a major route for L-tryptophan (L-TRP) metabolism, yielding a variety of bioactive compounds including kynurenic acid (KYNA), 3-hydroxykynurenine (3-HK), quinolinic acid (QUIN), and picolinic acid (PIC). These tryptophan catabolites are involved in the pathogenesis of many neuropsychiatric disorders, particularly when the KP becomes dysregulated. Accordingly, the enzymes that regulate the KP such as indoleamine 2,3-dioxygenase (IDO)/tryptophan 2,3-dioxygenase, kynurenine aminotransferases (KATs), and kynurenine 3-monooxygenase (KMO) represent potential drug targets as enzymatic inhibition can favorably rebalance KP metabolite concentrations. In addition, the galantamine-memantine combination, through its modulatory effects at the alpha7 nicotinic acetylcholine receptors and N-methyl-D-aspartate receptors, may counteract the effects of KYNA. The aim of this review is to highlight the effectiveness of IDO-1, KAT II, and KMO inhibitors, as well as the galantamine-memantine combination in the modulation of different KP metabolites. KAT II inhibitors are capable of decreasing the KYNA levels in the rat brain by a maximum of 80%. KMO inhibitors effectively reduce the central nervous system (CNS) levels of 3-HK, while markedly boosting the brain concentration of KYNA. Emerging data suggest that the galantamine-memantine combination also lowers L-TRP, kynurenine, KYNA, and PIC levels in humans. Presently, there are only 2 pathophysiological mechanisms (cholinergic and glutamatergic) that are FDA approved for the treatment of cognitive dysfunction for which purpose the galantamine-memantine combination has been designed for clinical use against Alzheimer's disease. The alpha7 nicotinic-NMDA hypothesis targeted by the galantamine-memantine combination has been implicated in the pathophysiology of various CNS diseases. Similarly, KYNA is well capable of modulating the neuropathophysiology of these disorders. This is known as the KYNA-centric hypothesis, which may be implicated in the management of certain neuropsychiatric conditions. In line with this hypothesis, KYNA may be considered as the "conductor of the orchestra" for the major pathophysiological mechanisms underlying CNS disorders. Therefore, there is great opportunity to further explore and compare the biological effects of these therapeutic modalities in animal models with a special focus on their effects on KP metabolites in the CNS and with the ultimate goal of progressing to clinical trials for many neuropsychiatric diseases.
Collapse
Affiliation(s)
- Michael Y. Bai
- Department of Biomedical Sciences, Neuroinflammation Group, Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - David B. Lovejoy
- Department of Biomedical Sciences, Neuroinflammation Group, Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Gilles J. Guillemin
- Department of Biomedical Sciences, Neuroinflammation Group, Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Rouba Kozak
- Neuroscience Drug Discovery Unit, Takeda Pharmaceuticals International Co, Cambridge, Massachusetts, USA
| | - Trevor W. Stone
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
| | - Maju Mathew Koola
- Department of Psychiatry and Behavioral Health, Stony Brook University Renaissance School of Medicine, Stony Brook, Stony Brook, New York, USA
| |
Collapse
|
18
|
Noorbakhsh A, Hosseininezhadian Koushki E, Farshadfar C, Ardalan N. Designing a natural inhibitor against human kynurenine aminotransferase type II and a comparison with PF-04859989: a computational effort against schizophrenia. J Biomol Struct Dyn 2021; 40:7038-7051. [PMID: 33645449 DOI: 10.1080/07391102.2021.1893817] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Kynurenine aminotransferase II (KATII) enzyme has an essential role in L-kynurenine transmission to kynurenic acid (KYNA). High concentration of kynurenic acid associates with schizophrenia and some neurocognitive disorders. Decreasing KYNA production via inhibiting KATII would be an effective method for treating and understanding the related central nervous system (CNS) diseases. This study aimed to discover a potent inhibitor against human KATII (hKATII) in comparison with PF-04859989. We utilized the computational methods of molecular dynamics, virtual screening, docking, and binding free-energy calculations. Initially, the 58722 compounds from three drug libraries, including IBS library, DrugBank library, and Analyticon library, were obtained. At the next stage, these sets of compounds were screened by AutoDock Vina software, and a potent inhibitor (ZINC35466084) was selected. Following the screening, molecular dynamics simulations for both ZINC35466084 and PF-04859989 were performed by GROMACS software. MM-PBSA analysis showed that the amount of binding free energy for ZINC35466084 (-61.26 KJ mol-1) is more potent than PF-04859989 (-43.14 KJ mol-1). Furthermore, the ADME analysis results revealed that the pharmacokinetic parameters of ZINC35466084 are acceptable for human use. Eventually, our data demonstrated that ZINC35466084 is suitable for hKATII inhibition, and it is an appropriate candidate for further studies in the laboratory. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Akbar Noorbakhsh
- Department of Biochemistry, Science and Research Branch, Islamic Azad University, Sanandaj, Iran
| | - Elnaz Hosseininezhadian Koushki
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Chiako Farshadfar
- Department of Biochemistry, Science and Research Branch, Islamic Azad University, Sanandaj, Iran
| | - Noeman Ardalan
- Department of Microbiology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
19
|
Tryptophan Metabolism as a Pharmacological Target. Trends Pharmacol Sci 2020; 42:60-73. [PMID: 33256987 DOI: 10.1016/j.tips.2020.11.006] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023]
Abstract
L-Tryptophan is an essential amino acid required for protein synthesis. It undergoes an extensive and complex metabolism along several pathways, resulting in many bioactive molecules acting in various organs through different action mechanisms. Enzymes involved in its metabolism, metabolites themselves, or their receptors, represent potential therapeutic targets, which are the subject of dynamic research. Disruptions in L-tryptophan metabolism are reported in several neurological, metabolic, psychiatric, and intestinal disorders, paving the way to develop drugs to target it. This review will briefly describe L-tryptophan metabolism and present and discuss the most recent pharmacological developments targeting it.
Collapse
|
20
|
The advantages of describing covalent inhibitor in vitro potencies by IC 50 at a fixed time point. IC 50 determination of covalent inhibitors provides meaningful data to medicinal chemistry for SAR optimization. Bioorg Med Chem 2020; 29:115865. [PMID: 33285410 DOI: 10.1016/j.bmc.2020.115865] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 01/07/2023]
Abstract
Recent years have seen a resurgence in drug discovery efforts aimed at the identification of covalent inhibitors which has led to an explosion of literature reports in this area and most importantly new approved therapies. These reports and breakthroughs highlight the significant investments made across the industry in SAR campaigns to optimize inhibitors. The potency of covalent inhibitors is generally considered to be more accurately described by the time-independent kinetic parameter kinact/Ki rather than a by a simple IC50 since the latter is a time-dependent parameter. Enzyme substrate concentrations are an additional important factor to consider when attempting to translate parameters derived from enzymology experiments to phenotypic behavior in a physiologically relevant cell-based system. Theoretical and experimental investigations into the relationship between IC50, time, substrate concentration and Kinact/Ki provided us with an effective approach to provide meaningful data for SAR optimization. The data we generated for our JAK3 irreversible covalent inhibitor program using IC50 values provided by enzyme assays with long incubations (>1h) coupled with physiological substrate concentration provided the medicinal chemist with optimal information in a rapid and efficient manner. We further document the wide applicability of this method by applying it to other enzymes systems where we have run covalent inhibitor programs.
Collapse
|
21
|
Transplantation of microbiota from drug-free patients with schizophrenia causes schizophrenia-like abnormal behaviors and dysregulated kynurenine metabolism in mice. Mol Psychiatry 2020; 25:2905-2918. [PMID: 31391545 DOI: 10.1038/s41380-019-0475-4] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/28/2019] [Accepted: 06/20/2019] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests that gut microbiota plays a role in the pathogenesis of schizophrenia via the microbiota-gut-brain axis. This study sought to investigate whether transplantation of fecal microbiota from drug-free patients with schizophrenia into specific pathogen-free mice could cause schizophrenia-like behavioral abnormalities. The results revealed that transplantation of fecal microbiota from schizophrenic patients into antibiotic-treated mice caused behavioral abnormalities such as psychomotor hyperactivity, impaired learning and memory in the recipient animals. These mice also showed elevation of the kynurenine-kynurenic acid pathway of tryptophan degradation in both periphery and brain, as well as increased basal extracellular dopamine in prefrontal cortex and 5-hydroxytryptamine in hippocampus, compared with their counterparts receiving feces from healthy controls. Furthermore, colonic luminal filtrates from the mice transplanted with patients' fecal microbiota increased both kynurenic acid synthesis and kynurenine aminotransferase II activity in cultured hepatocytes and forebrain cortical slices. Sixty species of donor-derived bacteria showed significant difference between the mice colonized with the patients' and the controls' fecal microbiota, highlighting 78 differentially enriched functional modules including tryptophan biosynthesis function. In conclusion, our study suggests that the abnormalities in the composition of gut microbiota contribute to the pathogenesis of schizophrenia partially through the manipulation of tryptophan-kynurenine metabolism.
Collapse
|
22
|
Lemos H, Mohamed E, Ou R, McCardle C, Zheng X, McGuire K, Homer NZM, Mole DJ, Huang L, Mellor AL. Co-treatments to Boost IDO Activity and Inhibit Production of Downstream Catabolites Induce Durable Suppression of Experimental Autoimmune Encephalomyelitis. Front Immunol 2020; 11:1256. [PMID: 32625215 PMCID: PMC7311583 DOI: 10.3389/fimmu.2020.01256] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022] Open
Abstract
Reinforcing defective tolerogenic processes slows progression of autoimmune (AI) diseases and has potential to promote drug-free disease remission. Previously, we reported that DNA nanoparticles (DNPs) and cyclic dinucleotides (CDNs) slow progression of experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis, by activating the Stimulator of Interferon Genes (STING) signaling adaptor to stimulate interferon type 1 (IFN-I) production, which induced dendritic cells to express indoleamine 2,3 dioxygenase (IDO) and acquire immune regulatory phenotypes. Here, we show that therapeutic responses to DNPs depend on DNA sensing via cyclic GAMP synthase (cGAS) and interactions between Programmed Death-1 (PD-1) and PD-1 ligands. To investigate how increased tryptophan (Trp) metabolism by IDO promotes therapeutic responses mice were co-treated at EAE onset with DNPs and drugs that inhibit kynurenine aminotransferase-II (KatII) or 3-hydroxyanthranilic acid dioxygenase (HAAO) activity downstream of IDO in the kynurenine (Kyn) pathway. DNP and KatII or HAAO inhibitor co-treatments suppressed EAE progression more effectively than DNPs, while KatII inhibition had no significant therapeutic benefit and HAAO inhibition attenuated but did not prevent EAE progression. Moreover, therapeutic responses to co-treatments were durable as EAE progression did not resume after co-treatment. Thus, using STING agonists to boost IDO activity and manipulating the Kyn pathway downstream of IDO is an effective strategy to enhance tolerogenic responses that overcome autoimmunity to suppress EAE progression.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/drug effects
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Autoimmunity
- B7-H1 Antigen/metabolism
- Chromatography, Liquid
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Enzyme Activation/drug effects
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Kynurenine/metabolism
- Membrane Proteins/agonists
- Metabolic Networks and Pathways
- Metabolome
- Metabolomics/methods
- Mice
- Mice, Knockout
- Nanoparticles
- Programmed Cell Death 1 Receptor/metabolism
- Signal Transduction/drug effects
- Tandem Mass Spectrometry
Collapse
Affiliation(s)
- Henrique Lemos
- Immune Metabolism Laboratory, Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Eslam Mohamed
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Rong Ou
- Immune Metabolism Laboratory, Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Caroline McCardle
- Immune Metabolism Laboratory, Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Xiaozhong Zheng
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Kris McGuire
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Natalie Z. M. Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Centre for Cardiovascular Sciences, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Damian J. Mole
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Lei Huang
- Immune Metabolism Laboratory, Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrew L. Mellor
- Immune Metabolism Laboratory, Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
23
|
Discovery of sulfonamides and 9-oxo-2,8-diazaspiro[5,5]undecane-2-carboxamides as human kynurenine aminotransferase 2 (KAT2) inhibitors. Bioorg Med Chem Lett 2020; 30:127060. [PMID: 32113843 DOI: 10.1016/j.bmcl.2020.127060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/14/2020] [Accepted: 02/21/2020] [Indexed: 02/02/2023]
Abstract
Human kynurenine aminotransferase 2 (KAT2) inhibitors could be potentially used to treat the cognitive deficits associated with bipolar disease and schizophrenia. Although, there has been active drug research activity by several industrial and academic groups in developing KAT2 inhibitors over the years, no such compound has proceeded to the clinics. Here, we report two different chemical series of reversible KAT2 inhibitors with sub-micromolar activities. The first series was identified by a high-throughput screening of a diverse random library and the second one by structure-based virtual screening. Two novel crystal structures of KAT2 complexed with different reversible inhibitors were also deposited to the Protein databank which could be useful for future drug discovery efforts.
Collapse
|
24
|
Yoshida T, Yamasaki S, Kaneko O, Taoka N, Tomimoto Y, Namatame I, Yahata T, Kuromitsu S, Cantley LC, Lyssiotis CA. A covalent small molecule inhibitor of glutamate-oxaloacetate transaminase 1 impairs pancreatic cancer growth. Biochem Biophys Res Commun 2020; 522:633-638. [PMID: 31787239 PMCID: PMC6981064 DOI: 10.1016/j.bbrc.2019.11.130] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 11/19/2019] [Indexed: 10/25/2022]
Abstract
Metabolic programs are rewired in cancer cells to support survival and tumor growth. Among these, recent studies have demonstrated that glutamate-oxaloacetate transaminase 1 (GOT1) plays key roles in maintaining redox homeostasis and proliferation of pancreatic ductal adenocarcinomas (PDA). This suggests that small molecule inhibitors of GOT1 could have utility for the treatment of PDA. However, the development of GOT1 inhibitors has been challenging, and no compound has yet demonstrated selectivity for GOT1-dependent cell metabolism or selective growth inhibition of PDA cell lines. In contrast, potent inhibitors that covalently bind to the transaminase cofactor pyridoxal-5'-phosphate (PLP), within the active site of the enzyme, have been reported for kynurenine aminotransferase (KAT) and gamma-aminobutyric acid aminotransferase (GABA-AT). Given the drug discovery successes with these transaminases, we aimed to identify PLP-dependent suicide substrate-type GOT1 inhibitors. Here, we demonstrate that PF-04859989, a known KAT2 inhibitor, has PLP-dependent inhibitory activity against GOT1 and shows selective growth inhibition of PDA cell lines.
Collapse
Affiliation(s)
- Tomohiro Yoshida
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba-shi, Ibaraki, 305-8585, Japan
| | - Shingo Yamasaki
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba-shi, Ibaraki, 305-8585, Japan
| | - Osamu Kaneko
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba-shi, Ibaraki, 305-8585, Japan
| | - Naofumi Taoka
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba-shi, Ibaraki, 305-8585, Japan
| | - Yusuke Tomimoto
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba-shi, Ibaraki, 305-8585, Japan
| | - Ichiji Namatame
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba-shi, Ibaraki, 305-8585, Japan
| | - Toshiko Yahata
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba-shi, Ibaraki, 305-8585, Japan
| | - Sadao Kuromitsu
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba-shi, Ibaraki, 305-8585, Japan
| | - Lewis C Cantley
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, United States
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, United States; Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, 48109, United States; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, United States.
| |
Collapse
|
25
|
Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat Rev Drug Discov 2019; 18:379-401. [PMID: 30760888 DOI: 10.1038/s41573-019-0016-5] [Citation(s) in RCA: 875] [Impact Index Per Article: 145.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
L-Tryptophan (Trp) metabolism through the kynurenine pathway (KP) is involved in the regulation of immunity, neuronal function and intestinal homeostasis. Imbalances in Trp metabolism in disorders ranging from cancer to neurodegenerative disease have stimulated interest in therapeutically targeting the KP, particularly the main rate-limiting enzymes indoleamine-2,3-dioxygenase 1 (IDO1), IDO2 and tryptophan-2,3-dioxygenase (TDO) as well as kynurenine monooxygenase (KMO). However, although small-molecule IDO1 inhibitors showed promise in early-stage cancer immunotherapy clinical trials, a phase III trial was negative. This Review summarizes the physiological and pathophysiological roles of Trp metabolism, highlighting the vast opportunities and challenges for drug development in multiple diseases.
Collapse
|
26
|
Kynurenines and the Endocannabinoid System in Schizophrenia: Common Points and Potential Interactions. Molecules 2019; 24:molecules24203709. [PMID: 31619006 PMCID: PMC6832375 DOI: 10.3390/molecules24203709] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/09/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022] Open
Abstract
Schizophrenia, which affects around 1% of the world’s population, has been described as a complex set of symptoms triggered by multiple factors. However, the exact background mechanisms remain to be explored, whereas therapeutic agents with excellent effectivity and safety profiles have yet to be developed. Kynurenines and the endocannabinoid system (ECS) play significant roles in both the development and manifestation of schizophrenia, which have been extensively studied and reviewed previously. Accordingly, kynurenines and the ECS share multiple features and mechanisms in schizophrenia, which have yet to be reviewed. Thus, the present study focuses on the main common points and potential interactions between kynurenines and the ECS in schizophrenia, which include (i) the regulation of glutamatergic/dopaminergic/γ-aminobutyric acidergic neurotransmission, (ii) their presence in astrocytes, and (iii) their role in inflammatory mechanisms. Additionally, promising pharmaceutical approaches involving the kynurenine pathway and the ECS will be reviewed herein.
Collapse
|
27
|
Biber K, Bhattacharya A, Campbell BM, Piro JR, Rohe M, Staal RGW, Talanian RV, Möller T. Microglial Drug Targets in AD: Opportunities and Challenges in Drug Discovery and Development. Front Pharmacol 2019; 10:840. [PMID: 31507408 PMCID: PMC6716448 DOI: 10.3389/fphar.2019.00840] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022] Open
Abstract
Alzheimer’s disease (AD) is a large and increasing unmet medical need with no disease-modifying treatment currently available. Genetic evidence from genome-wide association studies (GWASs) and gene network analysis has clearly revealed a key role of the innate immune system in the brain, of which microglia are the most important element. Single-nucleotide polymorphisms (SNPs) in genes predominantly expressed in microglia have been associated with altered risk of developing AD. Furthermore, microglia-specific pathways are affected on the messenger RNA (mRNA) expression level in post-mortem AD tissue and in mouse models of AD. Together these findings have increased the interest in microglia biology, and numerous scientific reports have proposed microglial molecules and pathways as drug targets for AD. Target identification and validation are generally the first steps in drug discovery. Both target validation and drug lead identification for central nervous system (CNS) targets and diseases entail additional significant obstacles compared to peripheral targets and diseases. This makes CNS drug discovery, even with well-validated targets, challenging. In this article, we will illustrate the special challenges of AD drug discovery by discussing the viability/practicality of possible microglia drug targets including cluster of differentiation 33 (CD33), KCa3.1, kynurenines, ionotropic P2 receptor 7 (P2X7), programmed death-1 (PD-1), Toll-like receptors (TLRs), and triggering receptor expressed in myeloid cells 2 (TREM2).
Collapse
Affiliation(s)
- Knut Biber
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Ludwigshafen, Germany
| | | | | | - Justin R Piro
- AbbVie Foundational Neuroscience Center, Cambridge, MA, United States
| | - Michael Rohe
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Ludwigshafen, Germany
| | | | - Robert V Talanian
- AbbVie Foundational Neuroscience Center, Cambridge, MA, United States
| | - Thomas Möller
- AbbVie Foundational Neuroscience Center, Cambridge, MA, United States
| |
Collapse
|
28
|
Yoshida Y, Fujigaki H, Kato K, Yamazaki K, Fujigaki S, Kunisawa K, Yamamoto Y, Mouri A, Oda A, Nabeshima T, Saito K. Selective and competitive inhibition of kynurenine aminotransferase 2 by glycyrrhizic acid and its analogues. Sci Rep 2019; 9:10243. [PMID: 31308447 PMCID: PMC6629613 DOI: 10.1038/s41598-019-46666-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/03/2019] [Indexed: 12/15/2022] Open
Abstract
The enzyme kynurenine aminotransferase (KAT) catalyses the conversion of kynurenine (KYN) to kynurenic acid (KYNA). Although the isozymes KAT1–4 have been identified, KYNA is mainly produced by KAT2 in brain tissues. KNYA is an antagonist of N-methyl-D-aspartate and α-7-nicotinic acetylcholine receptors, and accumulation of KYNA in the brain has been associated with the pathology of schizophrenia. Therefore, KAT2 could be exploited as a therapeutic target for the management of schizophrenia. Although currently available KAT2 inhibitors irreversibly bind to pyridoxal 5′-phosphate (PLP), inhibition via this mechanism may cause adverse side effects because of the presence of other PLP-dependent enzymes. Therefore, we identified novel selective KAT2 inhibitors by screening approximately 13,000 molecules. Among these, glycyrrhizic acid (GL) and its analogues, glycyrrhetinic acid (GA) and carbenoxolone (CBX), were identified as KAT2 inhibitors. These compounds were highly selective for KAT2 and competed with its substrate KYN, but had no effects on the other 3 KAT isozymes. Furthermore, we demonstrated that in complex structures that were predicted in docking calculations, GL, GA and CBX were located on the same surface as the aromatic ring of KYN. These results indicate that GL and its analogues are highly selective and competitive inhibitors of KAT2.
Collapse
Affiliation(s)
- Yukihiro Yoshida
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Sciences, Aichi, 470-1192, Japan
| | - Hidetsugu Fujigaki
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Sciences, Aichi, 470-1192, Japan.
| | - Koichi Kato
- College of Pharmacy, Kinjo Gakuin University, Aichi, 463-8521, Japan.,Faculty of Pharmacy, Meijo University, Aichi, 468-8503, Japan
| | - Kyoka Yamazaki
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Sciences, Aichi, 470-1192, Japan
| | - Suwako Fujigaki
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Sciences, Aichi, 470-1192, Japan
| | - Kazuo Kunisawa
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, Aichi, 470-1192, Japan
| | - Yasuko Yamamoto
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Sciences, Aichi, 470-1192, Japan
| | - Akihiro Mouri
- Department of Regulatory Science, Fujita Health University Graduate School of Health Sciences, Aichi, 470-1192, Japan.,Japanese Drug Organization of Appropriate Use and Research, Aichi, 468-0069, Japan
| | - Akifumi Oda
- Faculty of Pharmacy, Meijo University, Aichi, 468-8503, Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, Aichi, 470-1192, Japan.,Japanese Drug Organization of Appropriate Use and Research, Aichi, 468-0069, Japan
| | - Kuniaki Saito
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Sciences, Aichi, 470-1192, Japan.,Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, Aichi, 470-1192, Japan.,Japanese Drug Organization of Appropriate Use and Research, Aichi, 468-0069, Japan.,Human Health Sciences, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| |
Collapse
|
29
|
Investigating KYNA production and kynurenergic manipulation on acute mouse brain slice preparations. Brain Res Bull 2019; 146:185-191. [DOI: 10.1016/j.brainresbull.2018.12.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/19/2018] [Accepted: 12/26/2018] [Indexed: 01/09/2023]
|
30
|
Rossi F, Miggiano R, Ferraris DM, Rizzi M. The Synthesis of Kynurenic Acid in Mammals: An Updated Kynurenine Aminotransferase Structural KATalogue. Front Mol Biosci 2019; 6:7. [PMID: 30873412 PMCID: PMC6400995 DOI: 10.3389/fmolb.2019.00007] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 02/06/2019] [Indexed: 01/25/2023] Open
Abstract
Kynurenic acid (KYNA) is a bioactive compound that is produced along the kynurenine pathway (KP) during tryptophan degradation. In a few decades, KYNA shifted from being regarded a poorly characterized by-product of the KP to being considered a main player in many aspects of mammalian physiology, including the control of glutamatergic and cholinergic synaptic transmission, and the coordination of immunomodulation. The renewed attention being paid to the study of KYNA homeostasis is justified by the discovery of selective and potent inhibitors of kynurenine aminotransferase II, which is considered the main enzyme responsible for KYNA synthesis in the mammalian brain. Since abnormally high KYNA levels in the central nervous system have been associated with schizophrenia and cognitive impairment, these inhibitors promise the development of novel anti-psychotic and pro-cognitive drugs. Here, we summarize the currently available structural information on human and rodent kynurenine aminotransferases (KATs) as the result of global efforts aimed at describing the full complement of mammalian isozymes. These studies highlight peculiar features of KATs that can be exploited for the development of isozyme-specific inhibitors. Together with the optimization of biochemical assays to measure individual KAT activities in complex samples, this wealth of knowledge will continue to foster the identification and rational design of brain penetrant small molecules to attenuate KYNA synthesis, i.e., molecules capable of lowering KYNA levels without exposing the brain to the harmful withdrawal of KYNA-dependent neuroprotective actions.
Collapse
Affiliation(s)
- Franca Rossi
- Biochemistry and Biocrystallography Unit, DSF-Dipartimento di Scienze del Farmaco, University of Piemonte Orientale, Novara, Italy
| | - Riccardo Miggiano
- Biochemistry and Biocrystallography Unit, DSF-Dipartimento di Scienze del Farmaco, University of Piemonte Orientale, Novara, Italy
| | - Davide M Ferraris
- Biochemistry and Biocrystallography Unit, DSF-Dipartimento di Scienze del Farmaco, University of Piemonte Orientale, Novara, Italy
| | - Menico Rizzi
- Biochemistry and Biocrystallography Unit, DSF-Dipartimento di Scienze del Farmaco, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
31
|
Effect of substituents on 3(S)-amino-1-hydroxy-3,4-dihydroquinolin-2(1H)-one: a DFT study. Theor Chem Acc 2019. [DOI: 10.1007/s00214-018-2403-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
32
|
Chang C, Fonseca KR, Li C, Horner W, Zawadzke LE, Salafia MA, Welch KA, Strick CA, Campbell BM, Gernhardt SS, Rong H, Sawant-Basak A, Liras J, Dounay A, Tuttle JB, Verhoest P, Maurer TS. Quantitative Translational Analysis of Brain Kynurenic Acid Modulation via Irreversible Kynurenine Aminotransferase II Inhibition. Mol Pharmacol 2018; 94:823-833. [PMID: 29853495 DOI: 10.1124/mol.118.111625] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/24/2018] [Indexed: 11/22/2022] Open
Abstract
Kynurenic acid (KYNA) plays a significant role in maintaining normal brain function, and abnormalities in KYNA levels have been associated with various central nervous system disorders. Confirmation of its causality in human diseases requires safe and effective modulation of central KYNA levels in the clinic. The kynurenine aminotransferases (KAT) II enzyme represents an attractive target for pharmacologic modulation of central KYNA levels; however, KAT II and KYNA turnover kinetics, which could contribute to the duration of pharmacologic effect, have not been reported. In this study, the kinetics of central KYNA-lowering effect in rats and nonhuman primates (NHPs, Cynomolgus macaques) was investigated using multiple KAT II irreversible inhibitors as pharmacologic probes. Mechanistic pharmacokinetic-pharmacodynamic analysis of in vivo responses to irreversible inhibition quantitatively revealed that 1) KAT II turnover is relatively slow [16-76 hours' half-life (t1/2)], whereas KYNA is cleared more rapidly from the brain (<1 hour t1/2) in both rats and NHPs, 2) KAT II turnover is slower in NHPs than in rats (76 hours vs. 16 hours t1/2, respectively), and 3) the percent contribution of KAT II to KYNA formation is constant (∼80%) across rats and NHPs. Additionally, modeling results enabled establishment of in vitro-in vivo correlation for both enzyme turnover rates and drug potencies. In summary, quantitative translational analysis confirmed the feasibility of central KYNA modulation in humans. Model-based analysis, where system-specific properties and drug-specific properties are mechanistically separated from in vivo responses, enabled quantitative understanding of the KAT II-KYNA pathway, as well as assisted development of promising candidates to test KYNA hypothesis in humans.
Collapse
Affiliation(s)
- Cheng Chang
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Kari R Fonseca
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Cheryl Li
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Weldon Horner
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Laura E Zawadzke
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Michelle A Salafia
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Kathryn A Welch
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Christine A Strick
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Brian M Campbell
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Steve S Gernhardt
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Haojing Rong
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Aarti Sawant-Basak
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Jennifer Liras
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Amy Dounay
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Jamison B Tuttle
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Patrick Verhoest
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Tristan S Maurer
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| |
Collapse
|
33
|
Jayawickrama GS, Nematollahi A, Sun G, Church WB. Improvement of kynurenine aminotransferase-II inhibitors guided by mimicking sulfate esters. PLoS One 2018; 13:e0196404. [PMID: 29689093 PMCID: PMC5915280 DOI: 10.1371/journal.pone.0196404] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/12/2018] [Indexed: 01/08/2023] Open
Abstract
The mammalian kynurenine aminotransferase (KAT) enzymes are a family of related isoforms that are pyridoxal 5’-phosphate-dependent, responsible for the irreversible transamination of kynurenine to kynurenic acid. Kynurenic acid is implicated in human diseases such as schizophrenia where it is found in elevated levels and consequently KAT-II, as the isoform predominantly responsible for kynurenic acid production in the brain, has been targeted for the development of specific inhibitors. One class of compounds that have also shown inhibitory activity towards the KAT enzymes are estrogens and their sulfate esters. Estradiol disulfate in particular is very strongly inhibitory and it appears that the 17-sulfate makes a significant contribution to its potency. The work here demonstrates that the effect of this moiety can be mirrored in existing KAT-II inhibitors, from the development of two novel inhibitors, JN-01 and JN-02. Both inhibitors were based on NS-1502 (IC50: 315 μM), but the deliberate placement of a sulfonamide group significantly improved the potency of JN-01 (IC50: 73.8 μM) and JN-02 (IC50: 112.8 μM) in comparison to the parent compound. This 3–4 fold increase in potency shows the potential of these moieties to be accommodated in the KAT-II active site and the effect they can have on improving inhibitors, and the environments in the KAT-II have been suitably modelled using docking calculations.
Collapse
Affiliation(s)
- Gayan S. Jayawickrama
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, The University of Sydney, Sydney, New South Wales, Australia
| | - Alireza Nematollahi
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, The University of Sydney, Sydney, New South Wales, Australia
| | - Guanchen Sun
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, The University of Sydney, Sydney, New South Wales, Australia
| | - William Bret Church
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, The University of Sydney, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
34
|
Jayawickrama GS, Nematollahi A, Sun G, Church WB. Fragment Screening of Human Kynurenine Aminotransferase-II. SLAS DISCOVERY 2018. [PMID: 29537924 DOI: 10.1177/2472555218764620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Kynurenine aminotransferase-II (KAT-II) is a pyridoxal 5'-phosphate (PLP)-dependent enzyme that acts in the tryptophan metabolic pathway by catalyzing the transamination of kynurenine into kynurenic acid (KYNA). It is one of four isoforms in the KAT family, of which it is the primary homologue responsible for KYNA production in the mammalian brain. KAT-II is targeted for inhibition as KYNA is implicated in diseases such as schizophrenia, where it is found in elevated concentrations. Previously, many different approaches have been taken to develop KAT-II inhibitors, and herein fragment-based drug design (FBDD) approaches have been exploited to provide further lead compounds that can be designed into novel inhibitors. Surface plasmon resonance (SPR) was used to screen a fragment library containing 1000 compounds, of which 41 hits were identified. These hits were further evaluated with SPR, and 18 were selected for inhibition studies. From these hits, two fragments, F6037-0164 and F0037-7280, were pursued and determined to have an IC50 of 524.5 (± 25.6) μM and 115.2 (± 4.5) μM, respectively. This strategy shows the viability of using FBDD in gleaning knowledge about KAT-II inhibition and generating leads for the production of KAT-II inhibitors.
Collapse
Affiliation(s)
- Gayan S Jayawickrama
- 1 Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, The University of Sydney, Sydney, NSW, Australia
| | - Alireza Nematollahi
- 1 Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, The University of Sydney, Sydney, NSW, Australia
| | - Guanchen Sun
- 1 Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, The University of Sydney, Sydney, NSW, Australia
| | - W Bret Church
- 1 Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
35
|
Xu Y, Cuccui J, Denman C, Maharjan T, Wren BW, Wagner GK. Structure-activity relationships in a new class of non-substrate-like covalent inhibitors of the bacterial glycosyltransferase LgtC. Bioorg Med Chem 2018; 26:2973-2983. [PMID: 29602676 DOI: 10.1016/j.bmc.2018.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/03/2018] [Accepted: 03/04/2018] [Indexed: 02/07/2023]
Abstract
Lipooligosaccharide (LOS) structures in the outer core of Gram-negative mucosal pathogens such as Neisseria meningitidis and Haemophilus influenzae contain characteristic glycoepitopes that contribute significantly to bacterial virulence. An important example is the digalactoside epitope generated by the retaining α-1,4-galactosyltransferase LgtC. These digalactosides camouflage the pathogen from the host immune system and increase its serum resistance. Small molecular inhibitors of LgtC are therefore sought after as chemical tools to study bacterial virulence, and as potential candidates for anti-virulence drug discovery. We have recently discovered a new class of non-substrate-like inhibitors of LgtC. The new inhibitors act via a covalent mode of action, targeting a non-catalytic cysteine residue in the LgtC active site. Here, we describe, for the first time, structure-activity relationships for this new class of glycosyltransferase inhibitors. We have carried out a detailed analysis of the inhibition kinetics to establish the relative contribution of the non-covalent binding and the covalent inactivation steps for overall inhibitory activity. Selected inhibitors were also evaluated against a serum-resistant strain of Haemophilus influenzae, but did not enhance the killing effect of human serum.
Collapse
Affiliation(s)
- Yong Xu
- King's College London, Department of Chemistry, Faculty of Natural & Mathematical Sciences, Britannia House, 7 Trinity Street, London SE1 1DB, UK
| | - Jon Cuccui
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, UK
| | - Carmen Denman
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, UK
| | - Tripty Maharjan
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, UK
| | - Brendan W Wren
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, UK
| | - Gerd K Wagner
- King's College London, Department of Chemistry, Faculty of Natural & Mathematical Sciences, Britannia House, 7 Trinity Street, London SE1 1DB, UK.
| |
Collapse
|
36
|
Pellicciari R, Liscio P, Giacchè N, De Franco F, Carotti A, Robertson J, Cialabrini L, Katsyuba E, Raffaelli N, Auwerx J. α-Amino-β-carboxymuconate-ε-semialdehyde Decarboxylase (ACMSD) Inhibitors as Novel Modulators of De Novo Nicotinamide Adenine Dinucleotide (NAD +) Biosynthesis. J Med Chem 2018; 61:745-759. [PMID: 29345930 DOI: 10.1021/acs.jmedchem.7b01254] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
NAD+ has a central function in linking cellular metabolism to major cell-signaling and gene-regulation pathways. Defects in NAD+ homeostasis underpin a wide range of diseases, including cancer, metabolic disorders, and aging. Although the beneficial effects of boosting NAD+ on mitochondrial fitness, metabolism, and lifespan are well established, to date, no therapeutic enhancers of de novo NAD+ biosynthesis have been reported. Herein we report the discovery of 3-[[[5-cyano-1,6-dihydro-6-oxo-4-(2-thienyl)-2-pyrimidinyl]thio]methyl]phenylacetic acid (TES-1025, 22), the first potent and selective inhibitor of human ACMSD (IC50 = 0.013 μM) that increases NAD+ levels in cellular systems. The results of physicochemical-property, ADME, and safety profiling, coupled with in vivo target-engagement studies, support the hypothesis that ACMSD inhibition increases de novo NAD+ biosynthesis and position 22 as a first-class molecule for the evaluation of the therapeutic potential of ACMSD inhibition in treating disorders with perturbed NAD+ supply or homeostasis.
Collapse
Affiliation(s)
| | - Paride Liscio
- TES Pharma S.r.l. , IT-06073 Corciano, Perugia, Italy
| | | | | | - Andrea Carotti
- Department of Pharmaceutical Sciences, University of Perugia , IT-06123 Perugia, Italy
| | | | - Lucia Cialabrini
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche , IT-60131 Ancona, Italy
| | - Elena Katsyuba
- Laboratory of Integrative and Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne CH-1015 Lausanne, Switzerland
| | - Nadia Raffaelli
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche , IT-60131 Ancona, Italy
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne CH-1015 Lausanne, Switzerland
| |
Collapse
|
37
|
Jayawickrama GS, Nematollahi A, Sun G, Gorrell MD, Church WB. Inhibition of human kynurenine aminotransferase isozymes by estrogen and its derivatives. Sci Rep 2017; 7:17559. [PMID: 29242525 PMCID: PMC5730616 DOI: 10.1038/s41598-017-17979-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/23/2017] [Indexed: 12/27/2022] Open
Abstract
The kynurenine aminotransferase (KAT) enzymes are pyridoxal 5′-phosphate-dependent homodimers that catalyse the irreversible transamination of kynurenine into kynurenic acid (KYNA) in the tryptophan metabolic pathway. Kynurenic acid is implicated in cognitive diseases such as schizophrenia, and several inhibitors have been reported that selectively target KAT-II as it is primarily responsible for kynurenic acid production in the human brain. Not only is schizophrenia a sexually dimorphic condition, but women that have schizophrenia have reduced estrogen levels in their serum. Estrogens are also known to interact in the kynurenine pathway therefore exploring these interactions can yield a better understanding of the condition and improve approaches in ameliorating its effects. Enzyme inhibitory assays and binding studies showed that estradiol disulfate is a strong inhibitor of KAT-I and KAT-II (IC50: 291.5 μM and 26.3 μM, respectively), with estradiol, estradiol 3-sulfate and estrone sulfate being much weaker (IC50 > 2 mM). Therefore it is possible that estrogen levels can dictate the balance of kynurenic acid in the brain. Inhibition assay results and modelling suggests that the 17-sulfate moiety in estradiol disulfate is very important in improving its potency as an inhibitor, increasing the inhibition by approximately 10–100 fold compared to estradiol.
Collapse
Affiliation(s)
- Gayan S Jayawickrama
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
| | - Alireza Nematollahi
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
| | - Guanchen Sun
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
| | - Mark D Gorrell
- Molecular Hepatology Laboratory, Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - W Bret Church
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
38
|
Wang Q, An J, Alper H, Xiao WJ, Beauchemin AM. Catalytic substitution/cyclization sequences of O-substituted Isocyanates: synthesis of 1-alkoxybenzimidazolones and 1-alkoxy-3,4-dihydroquinazolin-2(1H)-ones. Chem Commun (Camb) 2017; 53:13055-13058. [PMID: 29165447 DOI: 10.1039/c7cc07926e] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
O-Substituted isocyanates (O-isocyanates) have rarely been used in organic synthesis, given their tendency to undergo side reactions (e.g., trimerization). Herein, we show that masked (blocked) O-isocyanate precursors allow one-pot or cascade reaction sequences featuring base-catalyzed substitution with 2-iodoanilines and 2-iodobenzylamines followed by copper-catalyzed cyclization, to form benzimidazolones and 3,4-dihydroquinazolin-2(1H)-ones. This work shows that O-isocyanates can serve as efficient building blocks for the synthesis of hydroxylamine-containing heterocycles.
Collapse
Affiliation(s)
- Qiang Wang
- CCNU-uOttawa Joint Research Centre, Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University, 152 Luoyu Road, Wuhan, Hubei 430079, China
| | | | | | | | | |
Collapse
|
39
|
Pettinger J, Jones K, Cheeseman MD. Lysine-Targeting Covalent Inhibitors. Angew Chem Int Ed Engl 2017; 56:15200-15209. [PMID: 28853194 DOI: 10.1002/anie.201707630] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 08/23/2017] [Indexed: 12/11/2022]
Abstract
Targeted covalent inhibitors have gained widespread attention in drug discovery as a validated method to circumvent acquired resistance in oncology. This strategy exploits small-molecule/protein crystal structures to design tightly binding ligands with appropriately positioned electrophilic warheads. Whilst most focus has been on targeting binding-site cysteine residues, targeting nucleophilic lysine residues can also represent a viable approach to irreversible inhibition. However, owing to the basicity of the ϵ-amino group in lysine, this strategy generates a number of specific challenges. Herein, we review the key principles for inhibitor design, give historical examples, and present recent developments that demonstrate the potential of lysine targeting for future drug discovery.
Collapse
Affiliation(s)
- Jonathan Pettinger
- Cancer Research, UK, Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Keith Jones
- Cancer Research, UK, Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Matthew D Cheeseman
- Cancer Research, UK, Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP, UK
| |
Collapse
|
40
|
Affiliation(s)
- Jonathan Pettinger
- Cancer Research, UK, Cancer Therapeutics Unit; The Institute of Cancer Research; London SW7 3RP Großbritannien
| | - Keith Jones
- Cancer Research, UK, Cancer Therapeutics Unit; The Institute of Cancer Research; London SW7 3RP Großbritannien
| | - Matthew D. Cheeseman
- Cancer Research, UK, Cancer Therapeutics Unit; The Institute of Cancer Research; London SW7 3RP Großbritannien
| |
Collapse
|
41
|
Jacobs KR, Castellano-Gonzalez G, Guillemin GJ, Lovejoy DB. Major Developments in the Design of Inhibitors along the Kynurenine Pathway. Curr Med Chem 2017; 24:2471-2495. [PMID: 28464785 PMCID: PMC5748880 DOI: 10.2174/0929867324666170502123114] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/13/2017] [Accepted: 04/18/2017] [Indexed: 12/20/2022]
Abstract
Disrupted kynurenine pathway (KP) metabolism has been implicated in the progression of neurodegenerative disease, psychiatric disorders and cancer. Modulation of enzyme activity along this pathway may therefore offer potential new therapeutic strategies for these conditions. Considering their prominent positions in the KP, the enzymes indoleamine 2,3-dioxygenase, kynurenine 3-monooxygenase and kynurenine aminotransferase, appear the most attractive targets. Already, increasing interest in this pathway has led to the identification of a number of potent and selective enzyme inhibitors with promising pre-clinical data and the elucidation of several enzyme crystal structures provides scope to rationalize the molecular mechanisms of inhibitor activity. The field seems poised to yield one or more inhibitors that should find clinical utility.
Collapse
Affiliation(s)
- Kelly R Jacobs
- Neuroinflammation Group, Department of Biomedical Research, Faculty of Medicine and Health Sciences, Macquarie University, Sydney. Australia
| | - Gloria Castellano-Gonzalez
- Neuroinflammation Group, Department of Biomedical Research, Faculty of Medicine and Health Sciences, Macquarie University, Sydney. Australia
| | - Gilles J Guillemin
- Department of Biomedical Research, Faculty of Medicine and Health Science, Macquarie University, 2 Technology Place, Sydney. Australia
| | - David B Lovejoy
- Department of Biomedical Research, Faculty of Medicine and Health Science, Macquarie University, 2 Technology Place, Sydney. Australia
| |
Collapse
|
42
|
Crystal structure and mechanistic analysis of a novel human kynurenine aminotransferase-2 reversible inhibitor. Med Chem Res 2017. [DOI: 10.1007/s00044-017-1950-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
43
|
Abnormal kynurenine pathway of tryptophan catabolism in cardiovascular diseases. Cell Mol Life Sci 2017; 74:2899-2916. [PMID: 28314892 DOI: 10.1007/s00018-017-2504-2] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/26/2017] [Accepted: 03/08/2017] [Indexed: 02/06/2023]
Abstract
Kynurenine pathway (KP) is the primary path of tryptophan (Trp) catabolism in most mammalian cells. The KP generates several bioactive catabolites, such as kynurenine (Kyn), kynurenic acid (KA), 3-hydroxykynurenine (3-HK), xanthurenic acid (XA), and 3-hydroxyanthranilic acid (3-HAA). Increased catabolite concentrations in serum are associated with several cardiovascular diseases (CVD), including heart disease, atherosclerosis, and endothelial dysfunction, as well as their risk factors, including hypertension, diabetes, obesity, and aging. The first catabolic step in KP is primarily controlled by indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO). Following this first step, the KP has two major branches, one branch is mediated by kynurenine 3-monooxygenase (KMO) and kynureninase (KYNU) and is responsible for the formation of 3-HK, 3-HAA, and quinolinic acid (QA); and another branch is controlled by kynurenine amino-transferase (KAT), which generates KA. Uncontrolled Trp catabolism has been demonstrated in distinct CVD, thus, understanding the underlying mechanisms by which regulates KP enzyme expression and activity is paramount. This review highlights the recent advances on the effect of KP enzyme expression and activity in different tissues on the pathological mechanisms of specific CVD, KP is an inflammatory sensor and modulator in the cardiovascular system, and KP catabolites act as the potential biomarkers for CVD initiation and progression. Moreover, the biochemical features of critical KP enzymes and principles of enzyme inhibitor development are briefly summarized, as well as the therapeutic potential of KP enzyme inhibitors against CVD is briefly discussed.
Collapse
|
44
|
Nadvi NA, Salam NK, Park J, Akladios FN, Kapoor V, Collyer CA, Gorrell MD, Church WB. High resolution crystal structures of human kynurenine aminotransferase-I bound to PLP cofactor, and in complex with aminooxyacetate. Protein Sci 2017; 26:727-736. [PMID: 28097769 DOI: 10.1002/pro.3119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 01/05/2017] [Accepted: 01/09/2017] [Indexed: 11/06/2022]
Abstract
In this study, we report two high-resolution structures of the pyridoxal 5' phosphate (PLP)-dependent enzyme kynurenine aminotransferase-I (KAT-I). One is the native structure with the cofactor in the PLP form bound to Lys247 with the highest resolution yet available for KAT-I at 1.28 Å resolution, and the other with the general PLP-dependent aminotransferase inhibitor, aminooxyacetate (AOAA) covalently bound to the cofactor at 1.54 Å. Only small conformational differences are observed in the vicinity of the aldimine (oxime) linkage with which the PLP forms the Schiff base with Lys247 in the 1.28 Å resolution native structure, in comparison to other native PLP-bound structures. We also report the inhibition of KAT-1 by AOAA and aminooxy-phenylpropionic acid (AOPP), with IC50s of 13.1 and 5.7 μM, respectively. The crystal structure of the enzyme in complex with the inhibitor AOAA revealed that the cofactor is the PLP form with the external aldimine linkage. The location of this oxime with the PLP, which forms in place of the native internal aldimine linkage of PLP of the native KAT-I, is away from the position of the native internal aldimine, with the free Lys247 substantially retaining the orientation of the native structure. Tyr101, at the active site, was observed in two conformations in both structures.
Collapse
Affiliation(s)
- Naveed A Nadvi
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, University of Sydney, Sydney, New South Wales, Australia.,Molecular Hepatology, Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Noeris K Salam
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, University of Sydney, Sydney, New South Wales, Australia
| | - Joohong Park
- Molecular Hepatology, Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Fady N Akladios
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, University of Sydney, Sydney, New South Wales, Australia
| | - Vimal Kapoor
- School of Medicine and Pharmacology, The University of Western Australia, Perth, Western, Australia, Australia
| | - Charles A Collyer
- School of Molecular Bioscience, University of Sydney, Sydney, New South Wales, Australia
| | - Mark D Gorrell
- Molecular Hepatology, Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - William Bret Church
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
45
|
Schwarcz R, Stone TW. The kynurenine pathway and the brain: Challenges, controversies and promises. Neuropharmacology 2017; 112:237-247. [PMID: 27511838 PMCID: PMC5803785 DOI: 10.1016/j.neuropharm.2016.08.003] [Citation(s) in RCA: 277] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/29/2016] [Accepted: 08/05/2016] [Indexed: 12/29/2022]
Abstract
Research on the neurobiology of the kynurenine pathway has suffered years of relative obscurity because tryptophan degradation, and its involvement in both physiology and major brain diseases, was viewed almost exclusively through the lens of the well-established metabolite serotonin. With increasing recognition that kynurenine and its metabolites can affect and even control a variety of classic neurotransmitter systems directly and indirectly, interest is expanding rapidly. Moreover, kynurenine pathway metabolism itself is modulated in conditions such as infection and stress, which are known to induce major changes in well-being and behaviour, so that kynurenines may be instrumental in the etiology of psychiatric and neurological disorders. It is therefore likely that the near future will not only witness the discovery of additional physiological and pathological roles for brain kynurenines, but also ever-increasing interest in drug development based on these roles. In particular, targeting the kynurenine pathway with new specific agents may make it possible to prevent disease by appropriate pharmacological or genetic manipulations. The following overview focuses on areas of kynurenine research which are either controversial, of major potential therapeutic interest, or just beginning to receive the degree of attention which will clarify their relevance to neurobiology and medicine. It also highlights technical issues so that investigators entering the field, and new research initiatives, are not misdirected by inappropriate experimental approaches or incorrect interpretations at this time of skyrocketing interest in the subject matter. This article is part of the Special Issue entitled 'The Kynurenine Pathway in Health and Disease'.
Collapse
Affiliation(s)
- Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Trevor W Stone
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
46
|
Notarangelo FM, Pocivavsek A. Elevated kynurenine pathway metabolism during neurodevelopment: Implications for brain and behavior. Neuropharmacology 2017; 112:275-285. [PMID: 26944732 PMCID: PMC5010529 DOI: 10.1016/j.neuropharm.2016.03.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 02/26/2016] [Accepted: 03/01/2016] [Indexed: 11/20/2022]
Abstract
The kynurenine pathway (KP) of tryptophan degradation contains several neuroactive metabolites that may influence brain function in health and disease. Mounting focus has been dedicated to investigating the role of these metabolites during neurodevelopment and elucidating their involvement in the pathophysiology of psychiatric disorders with a developmental component, such as schizophrenia. In this review, we describe the changes in KP metabolism in the brain from gestation until adulthood and illustrate how environmental and genetic factors affect the KP during development. With a particular focus on kynurenic acid, the antagonist of α7 nicotinic acetylcholine (α7nACh) and N-methyl-d-aspartate (NMDA) receptors, both implicated in modulating brain development, we review animal models designed to ascertain the role of perinatal KP elevation on long-lasting biochemical, neuropathological, and behavioral deficits later in life. We present new data demonstrating that combining perinatal choline-supplementation, to potentially increase activation of α7nACh receptors during development, with embryonic kynurenine manipulation is effective in attenuating cognitive impairments in adult rat offspring. With these findings in mind, we conclude the review by discussing the advancement of therapeutic interventions that would target not only symptoms, but potentially the root cause of central nervous system diseases that manifest from a perinatal KP insult. This article is part of the Special Issue entitled 'The Kynurenine Pathway in Health and Disease'.
Collapse
Affiliation(s)
- Francesca M Notarangelo
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ana Pocivavsek
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
47
|
Strelow JM. A Perspective on the Kinetics of Covalent and Irreversible Inhibition. SLAS DISCOVERY 2016; 22:3-20. [PMID: 27703080 DOI: 10.1177/1087057116671509] [Citation(s) in RCA: 215] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The clinical and commercial success of covalent drugs has prompted a renewed and more deliberate pursuit of covalent and irreversible mechanisms within drug discovery. A covalent mechanism can produce potent inhibition in a biochemical, cellular, or in vivo setting. In many cases, teams choose to focus on the consequences of the covalent event, defined by an IC50 value. In a biochemical assay, the IC50 may simply reflect the target protein concentration in the assay. What has received less attention is the importance of the rate of covalent modification, defined by kinact/KI. The kinact/KI is a rate constant describing the efficiency of covalent bond formation resulting from the potency (KI) of the first reversible binding event and the maximum potential rate (kinact) of inactivation. In this perspective, it is proposed that the kinact/KI should be employed as a critical parameter to identify covalent inhibitors, interpret structure-activity relationships (SARs), translate activity from biochemical assays to the cell, and more accurately define selectivity. It is also proposed that a physiologically relevant kinact/KI and an (unbound) AUC generated from a pharmacokinetic profile reflecting direct exposure of the inhibitor to the target protein are two critical determinants of in vivo covalent occupancy. A simple equation is presented to define this relationship and improve the interpretation of covalent and irreversible kinetics.
Collapse
Affiliation(s)
- John M Strelow
- 1 Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| |
Collapse
|
48
|
Study of the Activity and Possible Mechanism of Action of a Reversible Inhibitor of Recombinant Human KAT-2: A Promising Lead in Neurodegenerative and Cognitive Disorders. Molecules 2016; 21:molecules21070856. [PMID: 27367665 PMCID: PMC6273595 DOI: 10.3390/molecules21070856] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/24/2016] [Accepted: 06/27/2016] [Indexed: 01/23/2023] Open
Abstract
Abnormal levels of kynurenic acid (KYNA) in the human brain are believed to be connected to several central nervous system (CNS) diseases, therefore compounds which affect the production of this crucial metabolite are of interest in CNS drug development. The majority of KYNA production is accounted for by kynurenine aminotransferase-2 (KAT-2) in the mammalian brain; hence this enzyme is one of the most interesting targets with which to modulate KYNA levels. Recently developed human KAT-2 inhibitors with high potencies are known to irreversibly bind to the enzyme cofactor, pyridoxal-5′-phosphate (PLP), which may lead to severe side effects due to the abundance of PLP-dependent enzymes. In this study, we report a reversible and competitive inhibitor of KAT-2. Its inhibitory activities were examined using HPLC and surface plasmon resonance (SPR) and compare favorably with other recently reported KAT-2 inhibitors. Our inhibitor, NS-1502, demonstrates suitable inhibitory activity, almost 10 times more potent than the known reversible KAT-2, (S)-ESBA.
Collapse
|
49
|
Kynurenine Aminotransferase Isozyme Inhibitors: A Review. Int J Mol Sci 2016; 17:ijms17060946. [PMID: 27314340 PMCID: PMC4926479 DOI: 10.3390/ijms17060946] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 06/08/2016] [Accepted: 06/10/2016] [Indexed: 12/22/2022] Open
Abstract
Kynurenine aminotransferase isozymes (KATs 1–4) are members of the pyridoxal-5’-phosphate (PLP)-dependent enzyme family, which catalyse the permanent conversion of l-kynurenine (l-KYN) to kynurenic acid (KYNA), a known neuroactive agent. As KATs are found in the mammalian brain and have key roles in the kynurenine pathway, involved in different categories of central nervous system (CNS) diseases, the KATs are prominent targets in the quest to treat neurodegenerative and cognitive impairment disorders. Recent studies suggest that inhibiting these enzymes would produce effects beneficial to patients with these conditions, as abnormally high levels of KYNA are observed. KAT-1 and KAT-3 share the highest sequence similarity of the isozymes in this family, and their active site pockets are also similar. Importantly, KAT-2 has the major role of kynurenic acid production (70%) in the human brain, and it is considered therefore that suitable inhibition of this isozyme would be most effective in managing major aspects of CNS diseases. Human KAT-2 inhibitors have been developed, but the most potent of them, chosen for further investigations, did not proceed in clinical studies due to the cross toxicity caused by their irreversible interaction with PLP, the required cofactor of the KAT isozymes, and any other PLP-dependent enzymes. As a consequence of the possibility of extensive undesirable adverse effects, it is also important to pursue KAT inhibitors that reversibly inhibit KATs and to include a strategy that seeks compounds likely to achieve substantial interaction with regions of the active site other than the PLP. The main purpose of this treatise is to review the recent developments with the inhibitors of KAT isozymes. This treatise also includes analyses of their crystallographic structures in complex with this enzyme family, which provides further insight for researchers in this and related studies.
Collapse
|
50
|
Zádori D, Veres G, Szalárdy L, Klivényi P, Fülöp F, Toldi J, Vécsei L. Inhibitors of the kynurenine pathway as neurotherapeutics: a patent review (2012–2015). Expert Opin Ther Pat 2016; 26:815-32. [DOI: 10.1080/13543776.2016.1189531] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|