1
|
Girithar HN, Krishnamurthy S, Carroll L, Guller A, Bilgin AA, Gluch L, Guillemin GJ, Ahn SB, Heng B. Breast cancer metastasis progression is associated with elevated activity of kynurenine monooxygenase and kynureninase. Br J Cancer 2024; 131:1881-1892. [PMID: 39558063 PMCID: PMC11628561 DOI: 10.1038/s41416-024-02889-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/20/2024] Open
Abstract
INTRODUCTION Metastasis remains the major cause of death in breast cancer (BrCa) and lacks specific treatment strategies. The kynurenine pathway (KP) has been suggested as a key mechanism facilitating progression of BrCa. While KP activity has been explored in primary BrCa, its role in metastasis remains unclear. To better understand this, we examined changes in the KP of BrCa with no metastasis compared to BCa that produced local or distant metastases. Given that the cancer cell secretome plays a role in metastasis, we also investigated the relationship between changes in KP activity and serum proteins of patients with local or distant metastases. METHODS To investigate changes in the KP in BrCa, with and without metastasis, we quantified KP metabolites in blood sera collected from patients with stage 1 BrCa (n = 34), BrCa with local metastases (n = 46), BrCa with distant metastases (n = 20) and healthy controls (n = 39). The serum protein profile of the BrCa patients with local or distant metastasis was determined before correlation analyses were carried out to examine the relationship between changes in the KP and cancer serum proteins using SPSS. RESULTS We found that the KP was elevated in BrCa patients with local and distant metastasis compared to healthy controls and stage 1 BrCa patients. The activity of kynurenine monooxygenase (KMO) and kynureninase (KYNU) A was positively associated with disease stage and was higher compared to healthy controls. Proteome analysis in patients with local or distant metastasis revealed the dysregulation of 14 proteins, 9 of which were up-regulated and 5 down-regulated at the distant metastasis stage. Importantly, three of these proteins have not been previously linked to BrCa metastasis. In the correlation studies between the KP profile, cancer serum proteins and metastasis status, KYNU A had the greatest number of significant associations with cancer serum protein, followed by KMO. CONCLUSION Our findings reveal that the KP was regulated differently at various stages of BrCa and was more dysregulated in patients with local or distant metastasis. These KP activity changes showed a significant association with cancer serum proteins in BrCa patients with local or distant metastasis, highlighting the potential role of KP in BrCa metastasis.
Collapse
Affiliation(s)
- Hemaasri-Neya Girithar
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Shivani Krishnamurthy
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Luke Carroll
- Australian Proteome Analysis Facility, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Anna Guller
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- Computational Neurosurgery (CNS) Laboratory, Macquarie Medicine School, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW, Sydney, Australia
| | - Ayse A Bilgin
- Faculty of Sciences and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Laurence Gluch
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- The Strathfield Breast Centre, Strathfield, NSW, Australia
| | - Gilles J Guillemin
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, IPB University, Bogor, Indonesia
| | - Seong Beom Ahn
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Benjamin Heng
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.
| |
Collapse
|
2
|
Li W, Yang L, Chen H, Miao J, Wang Y, Zhou C, Chen Y, Kong Z, Shen C, Wu J, Li J, Zhu L, Li Z, Bian Y. Depression, stress, and tryptophan metabolism through the kynurenine pathway: treatment strategies from the perspective of Chinese herbal medicine. Metab Brain Dis 2024; 40:5. [PMID: 39546044 DOI: 10.1007/s11011-024-01461-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/20/2024] [Indexed: 11/17/2024]
Abstract
The pathogenesis of depression is complex, involving abnormalities in tryptophan (TRP) metabolism through the kynurenine pathway (KP). Moreover, depression is closely related to the hypothalamic-pituitary-adrenal (HPA) axis, the gut-brain axis, neuroinflammation, and stress. These factors collectively influence the multidimensional pathological mechanisms of depression. TRP, a fundamental amino acid, serves as a precursor for neuroactive metabolites vital to physiological functions. Central to TRP metabolism is the KP, and the imbalance between neurotoxic and neuroprotective metabolites is closely related to the onset and progression of depression. Therefore, maintaining the balance of KP metabolites is important. In this review, we have investigated the role of the KP in depression and explored the complexity of KP dysregulation and its therapeutic importance. Here, we highlight how a deeper understanding of the KP and its regulation can pave the way for new treatment strategies. Specifically, we have summarized the latest advances in elucidating the key mechanisms of rate-limiting enzyme inhibitors, providing insights into their potential therapeutic efficacy. In addition, we have explored the emerging field of Chinese herbal medicine, discussing its potential to regulate KP metabolites and alleviate depressive symptoms, thereby expanding the treatment options for depression. Our findings emphasize the multifaceted nature of depression and the necessity of interdisciplinary research to fully utilize KP regulation and Chinese herbal medicine as strategies to advance the treatment of depression.
Collapse
Affiliation(s)
- Wen Li
- School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lili Yang
- Jingwen Library, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Provincial Engineering Center of TCM External Medication Researching and Industrializing, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Haozhi Chen
- School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jia Miao
- School of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yutong Wang
- School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Changlin Zhou
- School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yanqi Chen
- School of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ziyang Kong
- School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chengyue Shen
- School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiafei Wu
- School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jinyi Li
- School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Luoying Zhu
- School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhengjun Li
- College of Health Economics Management, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yaoyao Bian
- School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Provincial Engineering Center of TCM External Medication Researching and Industrializing, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
3
|
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
|
4
|
Stone TW, Williams RO. Tryptophan metabolism as a 'reflex' feature of neuroimmune communication: Sensor and effector functions for the indoleamine-2, 3-dioxygenase kynurenine pathway. J Neurochem 2024; 168:3333-3357. [PMID: 38102897 DOI: 10.1111/jnc.16015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/16/2023] [Accepted: 11/08/2023] [Indexed: 12/17/2023]
Abstract
Although the central nervous system (CNS) and immune system were regarded as independent entities, it is now clear that immune system cells can influence the CNS, and neuroglial activity influences the immune system. Despite the many clinical implications for this 'neuroimmune interface', its detailed operation at the molecular level remains unclear. This narrative review focuses on the metabolism of tryptophan along the kynurenine pathway, since its products have critical actions in both the nervous and immune systems, placing it in a unique position to influence neuroimmune communication. In particular, since the kynurenine pathway is activated by pro-inflammatory mediators, it is proposed that physical and psychological stressors are the stimuli of an organismal protective reflex, with kynurenine metabolites as the effector arm co-ordinating protective neural and immune system responses. After a brief review of the neuroimmune interface, the general perception of tryptophan metabolism along the kynurenine pathway is expanded to emphasize this environmentally driven perspective. The initial enzymes in the kynurenine pathway include indoleamine-2,3-dioxygenase (IDO1), which is induced by tissue damage, inflammatory mediators or microbial products, and tryptophan-2,3-dioxygenase (TDO), which is induced by stress-induced glucocorticoids. In the immune system, kynurenic acid modulates leucocyte differentiation, inflammatory balance and immune tolerance by activating aryl hydrocarbon receptors and modulates pain via the GPR35 protein. In the CNS, quinolinic acid activates N-methyl-D-aspartate (NMDA)-sensitive glutamate receptors, whereas kynurenic acid is an antagonist: the balance between glutamate, quinolinic acid and kynurenic acid is a significant regulator of CNS function and plasticity. The concept of kynurenine and its metabolites as mediators of a reflex coordinated protection against stress helps to understand the variety and breadth of their activity. It should also help to understand the pathological origin of some psychiatric and neurodegenerative diseases involving the immune system and CNS, facilitating the development of new pharmacological strategies for treatment.
Collapse
Affiliation(s)
- Trevor W Stone
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Richard O Williams
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| |
Collapse
|
5
|
León-Letelier RA, Dou R, Vykoukal J, Sater AHA, Ostrin E, Hanash S, Fahrmann JF. The kynurenine pathway presents multi-faceted metabolic vulnerabilities in cancer. Front Oncol 2023; 13:1256769. [PMID: 37876966 PMCID: PMC10591110 DOI: 10.3389/fonc.2023.1256769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/22/2023] [Indexed: 10/26/2023] Open
Abstract
The kynurenine pathway (KP) and associated catabolites play key roles in promoting tumor progression and modulating the host anti-tumor immune response. To date, considerable focus has been on the role of indoleamine 2,3-dioxygenase 1 (IDO1) and its catabolite, kynurenine (Kyn). However, increasing evidence has demonstrated that downstream KP enzymes and their associated metabolite products can also elicit tumor-microenvironment immune suppression. These advancements in our understanding of the tumor promotive role of the KP have led to the conception of novel therapeutic strategies to target the KP pathway for anti-cancer effects and reversal of immune escape. This review aims to 1) highlight the known biological functions of key enzymes in the KP, and 2) provide a comprehensive overview of existing and emerging therapies aimed at targeting discrete enzymes in the KP for anti-cancer treatment.
Collapse
Affiliation(s)
- Ricardo A. León-Letelier
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rongzhang Dou
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jody Vykoukal
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ali Hussein Abdel Sater
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Edwin Ostrin
- Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Samir Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Johannes F. Fahrmann
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
6
|
Stone TW, Williams RO. Modulation of T cells by tryptophan metabolites in the kynurenine pathway. Trends Pharmacol Sci 2023; 44:442-456. [PMID: 37248103 DOI: 10.1016/j.tips.2023.04.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/26/2023] [Accepted: 04/26/2023] [Indexed: 05/31/2023]
Abstract
Lymphocytes maturing in the thymus (T cells) are key factors in adaptive immunity and the regulation of inflammation. The kynurenine pathway of tryptophan metabolism includes several enzymes and compounds that can modulate T cell function, but manipulating these pharmacologically has not achieved the expected therapeutic activity for the treatment of autoimmune disorders and cancer. With increasing knowledge of other pathways interacting with kynurenines, the expansion of screening methods, and the application of virtual techniques to understanding enzyme structures and mechanisms, details of interactions between kynurenines and other pathways are being revealed. This review surveys some of these alternative approaches to influence T cell function indirectly via the kynurenine pathway and summarizes the most recent work on the development of compounds acting directly on the kynurenine pathway.
Collapse
Affiliation(s)
- Trevor W Stone
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Richard O Williams
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK.
| |
Collapse
|
7
|
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
|
8
|
Ahamad S, Bhat SA. The Emerging Landscape of Small-Molecule Therapeutics for the Treatment of Huntington's Disease. J Med Chem 2022; 65:15993-16032. [PMID: 36490325 DOI: 10.1021/acs.jmedchem.2c00799] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin gene (HTT). The new insights into HD's cellular and molecular pathways have led to the identification of numerous potent small-molecule therapeutics for HD therapy. The field of HD-targeting small-molecule therapeutics is accelerating, and the approval of these therapeutics to combat HD may be expected in the near future. For instance, preclinical candidates such as naphthyridine-azaquinolone, AN1, AN2, CHDI-00484077, PRE084, EVP4593, and LOC14 have shown promise for further optimization to enter into HD clinical trials. This perspective aims to summarize the advent of small-molecule therapeutics at various stages of clinical development for HD therapy, emphasizing their structure and design, therapeutic effects, and specific mechanisms of action. Further, we have highlighted the key drivers involved in HD pathogenesis to provide insights into the basic principle for designing promising anti-HD therapeutic leads.
Collapse
Affiliation(s)
- Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh, Uttar Pradesh202002, India
| | - Shahnawaz A Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh, Uttar Pradesh202002, India
| |
Collapse
|
9
|
Swaih AM, Breda C, Sathyasaikumar KV, Allcock N, Collier MEW, Mason RP, Feasby A, Herrera F, Outeiro TF, Schwarcz R, Repici M, Giorgini F. Kynurenine 3-Monooxygenase Interacts with Huntingtin at the Outer Mitochondrial Membrane. Biomedicines 2022; 10:2294. [PMID: 36140394 PMCID: PMC9496550 DOI: 10.3390/biomedicines10092294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
The flavoprotein kynurenine 3-monooxygenase (KMO) is localised to the outer mitochondrial membrane and catalyses the synthesis of 3-hydroxykynurenine from L-kynurenine, a key step in the kynurenine pathway (KP) of tryptophan degradation. Perturbation of KP metabolism due to inflammation has long been associated with the pathogenesis of several neurodegenerative disorders, including Huntington's disease (HD)-which is caused by the expansion of a polyglutamine stretch in the huntingtin (HTT) protein. While HTT is primarily localised to the cytoplasm, it also associates with mitochondria, where it may physically interact with KMO. In order to test this hypothesis, we employed bimolecular fluorescence complementation (BiFC) and found that KMO physically interacts with soluble HTT exon 1 protein fragment in living cells. Notably, expansion of the disease-causing polyglutamine tract in HTT leads to the formation of proteinaceous intracellular inclusions that disrupt this interaction with KMO, markedly decreasing BiFC efficiency. Using confocal microscopy and ultrastructural analysis, we determined KMO and HTT localisation within the cell and found that the KMO-HTT interaction is localized to the outer mitochondrial membrane. These data suggest that KMO may interact with a pool of HTT at the mitochondrial membrane, highlighting a possible physiological role for mitochondrial HTT. The KMO-HTT interaction is abrogated upon polyglutamine expansion, which may indicate a heretofore unrecognized relevance in the pathogenesis of this disorder.
Collapse
Affiliation(s)
- Aisha M. Swaih
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Carlo Breda
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
- Leicester School of Allied Health Sciences, Faculty of Health and Life Sciences, De Montfort University, Leicester LE1 9BH, UK
| | - Korrapati V. Sathyasaikumar
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Natalie Allcock
- Core Biotechnology Services, Adrian Building, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - Mary E. W. Collier
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Robert P. Mason
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Adam Feasby
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Federico Herrera
- Cell Structure and Dynamics Laboratory, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
- BioISI—Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Tiago F. Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany
- Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle NE2 4HH, UK
- Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mariaelena Repici
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| |
Collapse
|
10
|
Chen Y, Zhang J, Yang Y, Xiang K, Li H, Sun D, Chen L. Kynurenine‐3‐monooxygenase (KMO): From its biological functions to therapeutic effect in diseases progression. J Cell Physiol 2022; 237:4339-4355. [DOI: 10.1002/jcp.30876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/21/2022] [Accepted: 09/01/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Yanmei Chen
- Key Laboratory of Structure‐Based Drug Design & Discovery, Wuya College of Innovation, School of Traditional Chinese Materia Medica, Ministry of Education Shenyang Pharmaceutical University Shenyang China
| | - Jiahui Zhang
- Key Laboratory of Structure‐Based Drug Design & Discovery, Wuya College of Innovation, School of Traditional Chinese Materia Medica, Ministry of Education Shenyang Pharmaceutical University Shenyang China
| | - Yueying Yang
- Key Laboratory of Structure‐Based Drug Design & Discovery, Wuya College of Innovation, School of Traditional Chinese Materia Medica, Ministry of Education Shenyang Pharmaceutical University Shenyang China
| | - Ke Xiang
- Key Laboratory of Structure‐Based Drug Design & Discovery, Wuya College of Innovation, School of Traditional Chinese Materia Medica, Ministry of Education Shenyang Pharmaceutical University Shenyang China
| | - Hua Li
- Key Laboratory of Structure‐Based Drug Design & Discovery, Wuya College of Innovation, School of Traditional Chinese Materia Medica, Ministry of Education Shenyang Pharmaceutical University Shenyang China
- College of Pharmacy Fujian University of Traditional Chinese Medicine Fuzhou China
| | - Dejuan Sun
- Key Laboratory of Structure‐Based Drug Design & Discovery, Wuya College of Innovation, School of Traditional Chinese Materia Medica, Ministry of Education Shenyang Pharmaceutical University Shenyang China
| | - Lixia Chen
- Key Laboratory of Structure‐Based Drug Design & Discovery, Wuya College of Innovation, School of Traditional Chinese Materia Medica, Ministry of Education Shenyang Pharmaceutical University Shenyang China
| |
Collapse
|
11
|
Chen J, Vitetta L, Henson JD, Hall S. Intestinal Dysbiosis, the Tryptophan Pathway and Nonalcoholic Steatohepatitis. Int J Tryptophan Res 2022; 15:11786469211070533. [PMID: 35153490 PMCID: PMC8829707 DOI: 10.1177/11786469211070533] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/07/2021] [Indexed: 12/15/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) progresses from simple steatosis to steatohepatitis (NASH), which may then progress to the development of cirrhosis and hepatocarcinoma. NASH is characterized by both steatosis and inflammation. Control of inflammation in NASH is a key step for the prevention of disease progression to severe sequalae. Intestinal dysbiosis has been recognized to be an important causal factor in the pathogenesis of NASH, involving both the accumulation of lipids and aggravation of inflammation. The effects of gut dysbiosis are mediated by adverse shifts of various intestinal commensal bacterial genera and their associated metabolites such as butyrate, tryptophan, and bile acids. In this review, we focus on the roles of tryptophan and its metabolites in NASH in association with intestinal dysbiosis and discuss possible therapeutic implications.
Collapse
Affiliation(s)
- Jiezhong Chen
- Research Department, Medlab Clinical, Sydney, NSW, Australia
| | - Luis Vitetta
- Research Department, Medlab Clinical, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Jeremy D Henson
- Research Department, Medlab Clinical, Sydney, NSW, Australia
- Faculty of Medicine, Prince of Wales Clinical School, The University of New South Wales, Sydney, NSW, Australia
| | - Sean Hall
- Research Department, Medlab Clinical, Sydney, NSW, Australia
| |
Collapse
|
12
|
Curry A, White D, Cen Y. Small Molecule Regulators Targeting NAD + Biosynthetic Enzymes. Curr Med Chem 2022; 29:1718-1738. [PMID: 34060996 PMCID: PMC8630097 DOI: 10.2174/0929867328666210531144629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 01/03/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a key player in many metabolic pathways as an activated carrier of electrons. In addition to being the cofactor for redox reactions, NAD+ also serves as the substrate for various enzymatic transformations such as adenylation and ADP-ribosylation. Maintaining cellular NAD+ homeostasis has been suggested as an effective anti-aging strategy. Given the importance of NAD+ in regulating a broad spectrum of cellular events, small molecules targeting NAD+ metabolism have been pursued as therapeutic interventions for the treatment of mitochondrial disorders and agerelated diseases. In this article, small molecule regulators of NAD+ biosynthetic enzymes will be reviewed. The focus will be given to the discovery and development of these molecules, the mechanism of action as well as their therapeutic potentials.
Collapse
Affiliation(s)
- Alyson Curry
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA
| | - Dawanna White
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA
| | - Yana Cen
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA;,Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, USA,Address correspondence to this author at the Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, USA; Tel: 804-828-7405;
| |
Collapse
|
13
|
Ostapiuk A, Urbanska EM. Kynurenic acid in neurodegenerative disorders-unique neuroprotection or double-edged sword? CNS Neurosci Ther 2022; 28:19-35. [PMID: 34862742 PMCID: PMC8673711 DOI: 10.1111/cns.13768] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 12/24/2022] Open
Abstract
AIMS The family of kynurenine pathway (KP) metabolites includes compounds produced along two arms of the path and acting in clearly opposite ways. The equilibrium between neurotoxic kynurenines, such as 3-hydroxykynurenine (3-HK) or quinolinic acid (QUIN), and neuroprotective kynurenic acid (KYNA) profoundly impacts the function and survival of neurons. This comprehensive review summarizes accumulated evidence on the role of KYNA in Alzheimer's, Parkinson's and Huntington's diseases, and discusses future directions of potential pharmacological manipulations aimed to modulate brain KYNA. DISCUSSION The synthesis of specific KP metabolites is tightly regulated and may considerably vary under physiological and pathological conditions. Experimental data consistently imply that shift of the KP to neurotoxic branch producing 3-HK and QUIN formation, with a relative or absolute deficiency of KYNA, is an important factor contributing to neurodegeneration. Targeting specific brain regions to maintain adequate KYNA levels seems vital; however, it requires the development of precise pharmacological tools, allowing to avoid the potential cognitive adverse effects. CONCLUSIONS Boosting KYNA levels, through interference with the KP enzymes or through application of prodrugs/analogs with high bioavailability and potency, is a promising clinical approach. The use of KYNA, alone or in combination with other compounds precisely influencing specific populations of neurons, is awaiting to become a significant therapy for neurodegenerative disorders.
Collapse
Affiliation(s)
- Aleksandra Ostapiuk
- Laboratory of Cellular and Molecular PharmacologyDepartment of Experimental and Clinical PharmacologyMedical University of LublinLublinPoland
- Present address:
Department of Clinical Digestive OncologyKU LeuvenLeuvenBelgium
| | - Ewa M. Urbanska
- Laboratory of Cellular and Molecular PharmacologyDepartment of Experimental and Clinical PharmacologyMedical University of LublinLublinPoland
| |
Collapse
|
14
|
Huang Y, Zhao M, Chen X, Zhang R, Le A, Hong M, Zhang Y, Jia L, Zang W, Jiang C, Wang J, Fan X, Wang J. Tryptophan Metabolism in Central Nervous System Diseases: Pathophysiology and Potential Therapeutic Strategies. Aging Dis 2022; 14:858-878. [PMID: 37191427 DOI: 10.14336/ad.2022.0916] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 09/16/2022] [Indexed: 11/19/2022] Open
Abstract
The metabolism of L-tryptophan (TRP) regulates homeostasis, immunity, and neuronal function. Altered TRP metabolism has been implicated in the pathophysiology of various diseases of the central nervous system. TRP is metabolized through two main pathways, the kynurenine pathway and the methoxyindole pathway. First, TRP is metabolized to kynurenine, then kynurenic acid, quinolinic acid, anthranilic acid, 3-hydroxykynurenine, and finally 3-hydroxyanthranilic acid along the kynurenine pathway. Second, TRP is metabolized to serotonin and melatonin along the methoxyindole pathway. In this review, we summarize the biological properties of key metabolites and their pathogenic functions in 12 disorders of the central nervous system: schizophrenia, bipolar disorder, major depressive disorder, spinal cord injury, traumatic brain injury, ischemic stroke, intracerebral hemorrhage, multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease. Furthermore, we summarize preclinical and clinical studies, mainly since 2015, that investigated the metabolic pathway of TRP, focusing on changes in biomarkers of these neurologic disorders, their pathogenic implications, and potential therapeutic strategies targeting this metabolic pathway. This critical, comprehensive, and up-to-date review helps identify promising directions for future preclinical, clinical, and translational research on neuropsychiatric disorders.
Collapse
|
15
|
Khetarpal V, Herbst T, Shefchek D, Ash S, Fitzsimmons M, Gohdes M, Munoz-Sanjuan I, Dominguez C. Pharmacokinetics and metabolic disposition of a potent and selective kynurenine monooxygenase inhibitor, CHDI-340246, in laboratory animals. Xenobiotica 2021; 51:1155-1180. [PMID: 34496722 DOI: 10.1080/00498254.2021.1977868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The disposition of a novel kynurenine monooxygenase inhibitor, CHDI-340246, was investigated in vitro and in animals.In vitro, there was minimal metabolic turnover of CHDI-340246 in all species. The protein binding was higher in human plasma (99.7%) relative to other species.In all species, blood clearance was low (<20% of liver blood flow) and volume of distribution was small (<0.5 L/kg). The terminal half-life was longer in monkeys (9 hr) than in mice, rats, or dogs (1-2 hr). CHDI-340246 was orally bioavailable (>60%) in all species.In rats, [14C]CHDI-340246 showed wide distribution of radioactivity in all tissues except brain and testes. In rats, the parent drug was the major circulating moiety with minor amounts of a sulphate conjugate of an O-dealkylated metabolite. The elimination occurred via the urinary route and to a lesser extent by biliary route, but mostly as metabolites. In cynomolgus monkeys, the parent drug predominated in plasma with only trace amounts of metabolites detected.Acyl glucuronide conjugate of CHDI-340246 was not detected in plasma of rats or monkeys.Overall, the ADME profile of CHDI-340246 was favourable in rats and monkeys for potential evaluation of KMO inhibition in humans.
Collapse
Affiliation(s)
| | - Todd Herbst
- CHDI Management/CHDI Foundation, Princeton, NJ, USA
| | | | - Steven Ash
- Covance Laboratories Inc, Madison, WI, USA
| | | | | | | | | |
Collapse
|
16
|
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
|
17
|
Rodrigues FB, Byrne LM, Lowe AJ, Tortelli R, Heins M, Flik G, Johnson EB, De Vita E, Scahill RI, Giorgini F, Wild EJ. Kynurenine pathway metabolites in cerebrospinal fluid and blood as potential biomarkers in Huntington's disease. J Neurochem 2021; 158:539-553. [PMID: 33797782 PMCID: PMC8375100 DOI: 10.1111/jnc.15360] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/18/2021] [Accepted: 03/26/2021] [Indexed: 01/31/2023]
Abstract
Converging lines of evidence from several models, and post-mortem human brain tissue studies, support the involvement of the kynurenine pathway (KP) in Huntington's disease (HD) pathogenesis. Quantifying KP metabolites in HD biofluids is desirable, both to study pathobiology and as a potential source of biomarkers to quantify pathway dysfunction and evaluate the biochemical impact of therapeutic interventions targeting its components. In a prospective single-site controlled cohort study with standardised collection of cerebrospinal fluid (CSF), blood, phenotypic and imaging data, we used high-performance liquid-chromatography to measure the levels of KP metabolites-tryptophan, kynurenine, kynurenic acid, 3-hydroxykynurenine, anthranilic acid and quinolinic acid-in CSF and plasma of 80 participants (20 healthy controls, 20 premanifest HD and 40 manifest HD). We investigated short-term stability, intergroup differences, associations with clinical and imaging measures and derived sample-size calculation for future studies. Overall, KP metabolites in CSF and plasma were stable over 6 weeks, displayed no significant group differences and were not associated with clinical or imaging measures. We conclude that the studied metabolites are readily and reliably quantifiable in both biofluids in controls and HD gene expansion carriers. However, we found little evidence to support a substantial derangement of the KP in HD, at least to the extent that it is reflected by the levels of the metabolites in patient-derived biofluids.
Collapse
Affiliation(s)
- Filipe B. Rodrigues
- UCL Huntington's Disease CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Lauren M. Byrne
- UCL Huntington's Disease CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Alexander J. Lowe
- UCL Huntington's Disease CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Rosanna Tortelli
- UCL Huntington's Disease CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | | | - Gunnar Flik
- Charles River LaboratoriesGroningenThe Netherlands
| | - Eileanoir B. Johnson
- UCL Huntington's Disease CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Enrico De Vita
- Lysholm Department of NeuroradiologyNational Hospital for Neurology & NeurosurgeryLondonUK
- Department of Biomedical EngineeringSchool of Biomedical Engineering and Imaging SciencesKing's College LondonLondonUK
| | - Rachael I. Scahill
- UCL Huntington's Disease CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Flaviano Giorgini
- Department of Genetics and Genome BiologyUniversity of LeicesterLeicesterUK
| | - Edward J. Wild
- UCL Huntington's Disease CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| |
Collapse
|
18
|
Role of Kynurenine Pathway in Oxidative Stress during Neurodegenerative Disorders. Cells 2021; 10:cells10071603. [PMID: 34206739 PMCID: PMC8306609 DOI: 10.3390/cells10071603] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/18/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative disorders are chronic and life-threatening conditions negatively affecting the quality of patients’ lives. They often have a genetic background, but oxidative stress and mitochondrial damage seem to be at least partly responsible for their development. Recent reports indicate that the activation of the kynurenine pathway (KP), caused by an activation of proinflammatory factors accompanying neurodegenerative processes, leads to the accumulation of its neuroactive and pro-oxidative metabolites. This leads to an increase in the oxidative stress level, which increases mitochondrial damage, and disrupts the cellular energy metabolism. This significantly reduces viability and impairs the proper functioning of central nervous system cells and may aggravate symptoms of many psychiatric and neurodegenerative disorders. This suggests that the modulation of KP activity could be effective in alleviating these symptoms. Numerous reports indicate that tryptophan supplementation, inhibition of KP enzymes, and administration or analogs of KP metabolites show promising results in the management of neurodegenerative disorders in animal models. This review gathers and systematizes the knowledge concerning the role of metabolites and enzymes of the KP in the development of oxidative damage within brain cells during neurodegenerative disorders and potential strategies that could reduce the severity of this process.
Collapse
|
19
|
Mithaiwala MN, Santana-Coelho D, Porter GA, O’Connor JC. Neuroinflammation and the Kynurenine Pathway in CNS Disease: Molecular Mechanisms and Therapeutic Implications. Cells 2021; 10:1548. [PMID: 34205235 PMCID: PMC8235708 DOI: 10.3390/cells10061548] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/13/2021] [Accepted: 06/15/2021] [Indexed: 12/16/2022] Open
Abstract
Diseases of the central nervous system (CNS) remain a significant health, social and economic problem around the globe. The development of therapeutic strategies for CNS conditions has suffered due to a poor understanding of the underlying pathologies that manifest them. Understanding common etiological origins at the cellular and molecular level is essential to enhance the development of efficacious and targeted treatment options. Over the years, neuroinflammation has been posited as a common link between multiple neurological, neurodegenerative and neuropsychiatric disorders. Processes that precipitate neuroinflammatory conditions including genetics, infections, physical injury and psychosocial factors, like stress and trauma, closely link dysregulation in kynurenine pathway (KP) of tryptophan metabolism as a possible pathophysiological factor that 'fuel the fire' in CNS diseases. In this study, we aim to review emerging evidence that provide mechanistic insights between different CNS disorders, neuroinflammation and the KP. We provide a thorough overview of the different branches of the KP pertinent to CNS disease pathology that have therapeutic implications for the development of selected and efficacious treatment strategies.
Collapse
Affiliation(s)
- Mustafa N. Mithaiwala
- Integrated Biomedical Sciences Program, Graduate School of Biomedical Sciences, UT Health San Antonio, San Antonio, TX 78229, USA; (M.N.M.); (D.S.-C.); (G.A.P.)
- Department of Pharmacology, Long School of Medicine, UT Health San Antonio, Mail Code 8864, San Antonio, TX 78229, USA
| | - Danielle Santana-Coelho
- Integrated Biomedical Sciences Program, Graduate School of Biomedical Sciences, UT Health San Antonio, San Antonio, TX 78229, USA; (M.N.M.); (D.S.-C.); (G.A.P.)
- Department of Pharmacology, Long School of Medicine, UT Health San Antonio, Mail Code 8864, San Antonio, TX 78229, USA
| | - Grace A. Porter
- Integrated Biomedical Sciences Program, Graduate School of Biomedical Sciences, UT Health San Antonio, San Antonio, TX 78229, USA; (M.N.M.); (D.S.-C.); (G.A.P.)
- Department of Pharmacology, Long School of Medicine, UT Health San Antonio, Mail Code 8864, San Antonio, TX 78229, USA
| | - Jason C. O’Connor
- Integrated Biomedical Sciences Program, Graduate School of Biomedical Sciences, UT Health San Antonio, San Antonio, TX 78229, USA; (M.N.M.); (D.S.-C.); (G.A.P.)
- Department of Pharmacology, Long School of Medicine, UT Health San Antonio, Mail Code 8864, San Antonio, TX 78229, USA
- Department of Research, Audie L. Murphy VA Hospital, South Texas Veterans Heath System, San Antonio, TX 78229, USA
| |
Collapse
|
20
|
Bondulich MK, Fan Y, Song Y, Giorgini F, Bates GP. Ablation of kynurenine 3-monooxygenase rescues plasma inflammatory cytokine levels in the R6/2 mouse model of Huntington's disease. Sci Rep 2021; 11:5484. [PMID: 33750843 PMCID: PMC7943810 DOI: 10.1038/s41598-021-84858-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 02/18/2021] [Indexed: 12/31/2022] Open
Abstract
Kynurenine 3-monooxygenase (KMO) regulates the levels of neuroactive metabolites in the kynurenine pathway (KP), dysregulation of which is associated with Huntington's disease (HD) pathogenesis. KMO inhibition leads to increased levels of neuroprotective relative to neurotoxic metabolites, and has been found to ameliorate disease-relevant phenotypes in several HD models. Here, we crossed KMO knockout mice to R6/2 HD mice to examine the effect of KMO depletion in the brain and periphery. KP genes were dysregulated in peripheral tissues from R6/2 mice and KMO ablation normalised levels of a subset of these. KP metabolites were also assessed, and KMO depletion led to increased levels of neuroprotective kynurenic acid in brain and periphery, and dramatically reduced neurotoxic 3-hydroxykunurenine levels in striatum and cortex. Notably, the increased levels of pro-inflammatory cytokines TNFa, IL1β, IL4 and IL6 found in R6/2 plasma were normalised upon KMO deletion. Despite these improvements in KP dysregulation and peripheral inflammation, KMO ablation had no effect upon several behavioural phenotypes. Therefore, although genetic inhibition of KMO in R6/2 mice modulates several metabolic and inflammatory parameters, these do not translate to improvements in primary disease indicators-observations which will likely be relevant for other interventions targeted at peripheral inflammation in HD.
Collapse
Affiliation(s)
- Marie Katrin Bondulich
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute At UCL, Queen Square Institute of Neurology, UCL, Queen Square, WC1N 3BG, UK
| | - Yilan Fan
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute At UCL, Queen Square Institute of Neurology, UCL, Queen Square, WC1N 3BG, UK
| | - Yeojin Song
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute At UCL, Queen Square Institute of Neurology, UCL, Queen Square, WC1N 3BG, UK
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, UK.
| | - Gillian P Bates
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute At UCL, Queen Square Institute of Neurology, UCL, Queen Square, WC1N 3BG, UK
| |
Collapse
|
21
|
Kimura H, Suda H, Kassai M, Endo M, Deai Y, Yahata M, Miyajima M, Isobe Y. N-(6-phenylpyridazin-3-yl)benzenesulfonamides as highly potent, brain-permeable, and orally active kynurenine monooxygenase inhibitors. Bioorg Med Chem Lett 2021; 33:127753. [PMID: 33359168 DOI: 10.1016/j.bmcl.2020.127753] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/07/2020] [Accepted: 12/12/2020] [Indexed: 10/22/2022]
Abstract
Huntington's disease (HD) is one of the serious neurodegenerative diseases and no disease modifiers are available to date. The correction of unbalanced kynurenine pathway metabolites may be useful to treat disease progression and kynurenine monooxygenase (KMO) is considered an ideal drug target. A couple of KMO inhibitors have been reported, but their brain permeability was very poor. We found pyridazinylsulfonamide as a novel lead compound, and it was optimized to the brain-permeable and highly potent KMO inhibitor 12, which was equipotent with CHDI-340246 and superior to CHDI-340246 in terms of brain penetration. Compound 12 was effective in R6/2 mice (HD model mice), i.e. neuroprotective kynurenic acid was increased, whereas neurotoxic 3-hydroxykynurenine was suppressed. In addition, impaired cognitive function was improved. Therefore, the brain-permeable KMO inhibitor was considered to be a disease modifier for HD treatment.
Collapse
Affiliation(s)
- Hidenori Kimura
- Drug Research Division, Sumitomo Dainippon Pharma. Co., Ltd., 3-1-98, Kasugade-naka, Konohana-ku, Osaka 554-0022, Japan
| | - Hitoshi Suda
- Drug Research Division, Sumitomo Dainippon Pharma. Co., Ltd., 3-1-98, Kasugade-naka, Konohana-ku, Osaka 554-0022, Japan
| | - Momoe Kassai
- Drug Research Division, Sumitomo Dainippon Pharma. Co., Ltd., 3-1-98, Kasugade-naka, Konohana-ku, Osaka 554-0022, Japan
| | - Mika Endo
- Drug Research Division, Sumitomo Dainippon Pharma. Co., Ltd., 3-1-98, Kasugade-naka, Konohana-ku, Osaka 554-0022, Japan
| | - Yoko Deai
- Drug Research Division, Sumitomo Dainippon Pharma. Co., Ltd., 3-1-98, Kasugade-naka, Konohana-ku, Osaka 554-0022, Japan
| | - Masahiro Yahata
- Drug Research Division, Sumitomo Dainippon Pharma. Co., Ltd., 3-1-98, Kasugade-naka, Konohana-ku, Osaka 554-0022, Japan
| | - Mari Miyajima
- Drug Research Division, Sumitomo Dainippon Pharma. Co., Ltd., 3-1-98, Kasugade-naka, Konohana-ku, Osaka 554-0022, Japan
| | - Yoshiaki Isobe
- Drug Research Division, Sumitomo Dainippon Pharma. Co., Ltd., 3-1-98, Kasugade-naka, Konohana-ku, Osaka 554-0022, Japan.
| |
Collapse
|
22
|
Zhang S, Collier MEW, Heyes DJ, Giorgini F, Scrutton NS. Advantages of brain penetrating inhibitors of kynurenine-3-monooxygenase for treatment of neurodegenerative diseases. Arch Biochem Biophys 2020; 697:108702. [PMID: 33275878 DOI: 10.1016/j.abb.2020.108702] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/18/2020] [Accepted: 11/24/2020] [Indexed: 01/16/2023]
Abstract
Kynurenine-3-monooxygenase (KMO) is an important therapeutic target for several brain disorders that has been extensively studied in recent years. Potent inhibitors towards KMO have been developed and tested within different disease models, showing great therapeutic potential, especially in models of neurodegenerative disease. The inhibition of KMO reduces the production of downstream toxic kynurenine pathway metabolites and shifts the flux to the formation of the neuroprotectant kynurenic acid. However, the efficacy of KMO inhibitors in neurodegenerative disease has been limited by their poor brain permeability. Combined with virtual screening and prodrug strategies, a novel brain penetrating KMO inhibitor has been developed which dramatically decreases neurotoxic metabolites. This review highlights the importance of KMO as a drug target in neurological disease and the benefits of brain permeable inhibitors in modulating kynurenine pathway metabolites in the central nervous system.
Collapse
Affiliation(s)
- Shaowei Zhang
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - Mary E W Collier
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, UK
| | - Derren J Heyes
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, UK
| | - Nigel S Scrutton
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK.
| |
Collapse
|
23
|
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
|
24
|
Maddison DC, Alfonso-Núñez M, Swaih AM, Breda C, Campesan S, Allcock N, Straatman-Iwanowska A, Kyriacou CP, Giorgini F. A novel role for kynurenine 3-monooxygenase in mitochondrial dynamics. PLoS Genet 2020; 16:e1009129. [PMID: 33170836 PMCID: PMC7654755 DOI: 10.1371/journal.pgen.1009129] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/22/2020] [Indexed: 11/19/2022] Open
Abstract
The enzyme kynurenine 3-monooxygenase (KMO) operates at a critical branch-point in the kynurenine pathway (KP), the major route of tryptophan metabolism. As the KP has been implicated in the pathogenesis of several human diseases, KMO and other enzymes that control metabolic flux through the pathway are potential therapeutic targets for these disorders. While KMO is localized to the outer mitochondrial membrane in eukaryotic organisms, no mitochondrial role for KMO has been described. In this study, KMO deficient Drosophila melanogaster were investigated for mitochondrial phenotypes in vitro and in vivo. We find that a loss of function allele or RNAi knockdown of the Drosophila KMO ortholog (cinnabar) causes a range of morphological and functional alterations to mitochondria, which are independent of changes to levels of KP metabolites. Notably, cinnabar genetically interacts with the Parkinson's disease associated genes Pink1 and parkin, as well as the mitochondrial fission gene Drp1, implicating KMO in mitochondrial dynamics and mitophagy, mechanisms which govern the maintenance of a healthy mitochondrial network. Overexpression of human KMO in mammalian cells finds that KMO plays a role in the post-translational regulation of DRP1. These findings reveal a novel mitochondrial role for KMO, independent from its enzymatic role in the kynurenine pathway.
Collapse
Affiliation(s)
- Daniel C. Maddison
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Mónica Alfonso-Núñez
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Aisha M. Swaih
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Carlo Breda
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, United Kingdom
- Leicester School of Allied Health Sciences, Faculty of Health and Life Sciences, De Montfort University, Leicester, LE1 9BH, United Kingdom
| | - Susanna Campesan
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Natalie Allcock
- Core Biotechnology Services, Adrian Building, University of Leicester, University Road, Leicester, LE1 7RH, Leicestershire, United Kingdom
| | - Anna Straatman-Iwanowska
- Core Biotechnology Services, Adrian Building, University of Leicester, University Road, Leicester, LE1 7RH, Leicestershire, United Kingdom
| | - Charalambos P. Kyriacou
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, United Kingdom
- * E-mail:
| |
Collapse
|
25
|
Mor A, Kalaska B, Pawlak D. Kynurenine Pathway in Chronic Kidney Disease: What’s Old, What’s New, and What’s Next? Int J Tryptophan Res 2020; 13:1178646920954882. [PMID: 35210786 PMCID: PMC8862190 DOI: 10.1177/1178646920954882] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/06/2020] [Indexed: 11/25/2022] Open
Abstract
Impaired kidney function and increased inflammatory process occurring in the course of Chronic Kidney Disease (CKD) contribute to the development of complex amino-acid alterations. The essential amino-acid tryptophan (TRP) undergoes extensive metabolism along several pathways, resulting in the production of many biologically active compounds. The results of many studies have shown that its metabolism via the kynurenine pathway is potently increased in the course of CKD. Metabolites of this pathway exhibit differential, sometimes opposite, roles in several biological processes. Their accumulation in the course of CKD may induce oxidative cell damage which stimulates inflammatory processes. They can also modulate the activity of numerous cellular signaling pathways through activation of the aryl hydrocarbon receptor, leading to the disruption of homeostasis of various organs. As a result, they can contribute to the development of the systemic disorders accompanying the course of chronic renal failure. This review gathers and systematizes reports concerning the knowledge connecting the kynurenine pathway metabolites to systemic disorders accompanying the development of CKD.
Collapse
Affiliation(s)
- Adrian Mor
- Department of Pharmacodynamics, Medical University of Bialystok, Bialystok, Poland
| | - Bartlomiej Kalaska
- Department of Pharmacodynamics, Medical University of Bialystok, Bialystok, Poland
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
26
|
Lu Y, Shao M, Wu T. Kynurenine-3-monooxygenase: A new direction for the treatment in different diseases. Food Sci Nutr 2020; 8:711-719. [PMID: 32148781 PMCID: PMC7020307 DOI: 10.1002/fsn3.1418] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 11/24/2019] [Accepted: 12/21/2019] [Indexed: 12/14/2022] Open
Abstract
Kynurenine-3-monooxygenase (KMO) is an enzyme that relies on nicotinamide adenine dinucleotide phosphate (NADP), a key site in the kynurenine pathway (KP), which has great effects on neurological diseases, cancer, and peripheral inflammation. This review mainly pay attention to the research of KMO mechanism for the treatment of different diseases, and hopes to provide assistance for clinical and drug use. KMO controlling the chief division of the KP, which directly controls downstream product quinolinic acid (QUIN) and indirectly controls kynurenic acid (KYNA), plays an important role in many diseases, especially neurological diseases.
Collapse
Affiliation(s)
- Yifei Lu
- Institute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Mingmei Shao
- Institute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
27
|
Kim BJ, Lee SH, Koh JM. Clinical insights into the kynurenine pathway in age-related diseases. Exp Gerontol 2019; 130:110793. [PMID: 31765740 DOI: 10.1016/j.exger.2019.110793] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/18/2019] [Accepted: 11/21/2019] [Indexed: 01/02/2023]
Abstract
Accumulating evidence from diverse experiments, including heterochronic parabiosis-the surgical joining of two animals of different ages-has highlighted the importance of systemic factors in the progressive functional decline of various organs and tissues during aging. The major metabolic pathway of tryptophan, an essential amino acid in humans, is the kynurenine pathway (KP) in which indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) catalyze the conversion of tryptophan into kynurenine. Importantly, circulating kynurenine produced by this enzymatic breakdown, as a primary driver of the aging process, has been linked to higher mortality in humans. This review discusses the potential roles of tryptophan derivatives as biomarkers for the risk of frailty in the elderly, based on human observational studies as well as the KP as a therapeutic target for age-related diseases.
Collapse
Affiliation(s)
- Beom-Jun Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Seung Hun Lee
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jung-Min Koh
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
28
|
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: 861] [Impact Index Per Article: 143.5] [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
|
29
|
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
|
30
|
Swainson LA, Ahn H, Pajanirassa P, Khetarpal V, Deleage C, Estes JD, Hunt PW, Munoz-Sanjuan I, McCune JM. Kynurenine 3-Monooxygenase Inhibition during Acute Simian Immunodeficiency Virus Infection Lowers PD-1 Expression and Improves Post-Combination Antiretroviral Therapy CD4 + T Cell Counts and Body Weight. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:899-910. [PMID: 31285277 PMCID: PMC6684450 DOI: 10.4049/jimmunol.1801649] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/17/2019] [Indexed: 01/31/2023]
Abstract
The kynurenine pathway (KP) is a key regulator of many important physiological processes and plays a harmful role in cancer, many neurologic conditions, and chronic viral infections. In HIV infection, KP activity is consistently associated with reduced CD4 T cell counts and elevated levels of T cell activation and viral load; it also independently predicts mortality and morbidity from non-AIDS events. Kynurenine 3-monooxygenase (KMO) is a therapeutically important target in the KP. Using the nonhuman primate model of SIV infection in rhesus macaques, we investigated whether KMO inhibition could slow the course of disease progression. We used a KMO inhibitor, CHDI-340246, to perturb the KP during early acute infection and followed the animals for 1 y to assess clinical outcomes and immune phenotype and function during pre-combination antiretroviral therapy acute infection and combination antiretroviral therapy-treated chronic infection. Inhibition of KMO in acute SIV infection disrupted the KP and prevented SIV-induced increases in downstream metabolites, improving clinical outcome as measured by both increased CD4+ T cell counts and body weight. KMO inhibition increased naive T cell frequency and lowered PD-1 expression in naive and memory T cell subsets. Importantly, early PD-1 expression during acute SIV infection predicted clinical outcomes of body weight and CD4+ T cell counts. Our data indicate that KMO inhibition in early acute SIV infection provides clinical benefit and suggest a rationale for testing KMO inhibition as an adjunctive treatment in SIV/HIV infection to slow the progression of the disease and improve immune reconstitution.
Collapse
Affiliation(s)
- Louise A Swainson
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110;
| | - Haelee Ahn
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110
| | - Priya Pajanirassa
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110
| | | | - Claire Deleage
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21701
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21701
| | - Peter W Hunt
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110
| | | | - Joseph M McCune
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110
| |
Collapse
|
31
|
Zhang S, Sakuma M, Deora GS, Levy CW, Klausing A, Breda C, Read KD, Edlin CD, Ross BP, Wright Muelas M, Day PJ, O’Hagan S, Kell DB, Schwarcz R, Leys D, Heyes DJ, Giorgini F, Scrutton NS. A brain-permeable inhibitor of the neurodegenerative disease target kynurenine 3-monooxygenase prevents accumulation of neurotoxic metabolites. Commun Biol 2019; 2:271. [PMID: 31372510 PMCID: PMC6656724 DOI: 10.1038/s42003-019-0520-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 06/28/2019] [Indexed: 12/16/2022] Open
Abstract
Dysregulation of the kynurenine pathway (KP) leads to imbalances in neuroactive metabolites associated with the pathogenesis of several neurodegenerative disorders, including Huntington's disease (HD). Inhibition of the enzyme kynurenine 3-monooxygenase (KMO) in the KP normalises these metabolic imbalances and ameliorates neurodegeneration and related phenotypes in several neurodegenerative disease models. KMO is thus a promising candidate drug target for these disorders, but known inhibitors are not brain permeable. Here, 19 new KMO inhibitors have been identified. One of these (1) is neuroprotective in a Drosophila HD model but is minimally brain penetrant in mice. The prodrug variant (1b) crosses the blood-brain barrier, releases 1 in the brain, thereby lowering levels of 3-hydroxykynurenine, a toxic KP metabolite linked to neurodegeneration. Prodrug 1b will advance development of targeted therapies against multiple neurodegenerative and neuroinflammatory diseases in which KP likely plays a role, including HD, Alzheimer's disease, and Parkinson's disease.
Collapse
Affiliation(s)
- Shaowei Zhang
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Michiyo Sakuma
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Girdhar S. Deora
- School of Pharmacy, The University of Queensland, Brisbane, Queensland 4072 Australia
| | - Colin W. Levy
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Alex Klausing
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228 USA
| | - Carlo Breda
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH UK
| | - Kevin D. Read
- Drug Discovery Unit, School of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH UK
| | | | - Benjamin P. Ross
- School of Pharmacy, The University of Queensland, Brisbane, Queensland 4072 Australia
| | - Marina Wright Muelas
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Philip J. Day
- Manchester Institute of Biotechnology and Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL UK
| | - Stephen O’Hagan
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Douglas B. Kell
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228 USA
| | - David Leys
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Derren J. Heyes
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH UK
| | - Nigel S. Scrutton
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| |
Collapse
|
32
|
The ‘Yin’ and the ‘Yang’ of the kynurenine pathway: excitotoxicity and neuroprotection imbalance in stress-induced disorders. Behav Pharmacol 2019; 30:163-186. [DOI: 10.1097/fbp.0000000000000477] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
33
|
Di Marco A, Gonzalez Paz O, Fini I, Vignone D, Cellucci A, Battista MR, Auciello G, Orsatti L, Zini M, Monteagudo E, Khetarpal V, Rose M, Dominguez C, Herbst T, Toledo-Sherman L, Summa V, Muñoz-Sanjuán I. Application of an in Vitro Blood–Brain Barrier Model in the Selection of Experimental Drug Candidates for the Treatment of Huntington’s Disease. Mol Pharm 2019; 16:2069-2082. [DOI: 10.1021/acs.molpharmaceut.9b00042] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Vinod Khetarpal
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| | - Mark Rose
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| | - Celia Dominguez
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| | - Todd Herbst
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| | - Leticia Toledo-Sherman
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| | | | - Ignacio Muñoz-Sanjuán
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| |
Collapse
|
34
|
Abstract
The 25 years since the identification of the gene responsible for Huntington disease (HD) have stood witness to profound discoveries about the nature of the disease and its pathogenesis. Despite this progress, however, the development of disease-modifying therapies has thus far been slow. Preclinical validation of the therapeutic potential of disrupted pathways in HD has led to the advancement of pharmacological agents, both novel and repurposed, for clinical evaluation. The most promising therapeutic approaches include huntingtin (HTT) lowering and modification as well as modulation of neuroinflammation and synaptic transmission. With clinical trials for many of these approaches imminent or currently ongoing, the coming years are promising not only for HD but also for more prevalent neurodegenerative disorders, such as Alzheimer and Parkinson disease, in which many of these pathways have been similarly implicated.
Collapse
|
35
|
Kim HT, Na BK, Chung J, Kim S, Kwon SK, Cha H, Son J, Cho JM, Hwang KY. Structural Basis for Inhibitor-Induced Hydrogen Peroxide Production by Kynurenine 3-Monooxygenase. Cell Chem Biol 2018; 25:426-438.e4. [PMID: 29429898 DOI: 10.1016/j.chembiol.2018.01.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/17/2017] [Accepted: 01/08/2018] [Indexed: 11/18/2022]
Abstract
Kynurenine 3-monooxygenase (KMO) inhibitors have been developed for the treatment of neurodegenerative disorders. The mechanisms of flavin reduction and hydrogen peroxide production by KMO inhibitors are unknown. Herein, we report the structure of human KMO and crystal structures of Saccharomyces cerevisiae (sc) and Pseudomonas fluorescens (pf) KMO with Ro 61-8048. Proton transfer in the hydrogen bond network triggers flavin reduction in p-hydroxybenzoate hydroxylase, but the mechanism triggering flavin reduction in KMO is different. Conformational changes via π-π interactions between the loop above the flavin and substrate or non-substrate effectors lead to disorder of the C-terminal α helix in scKMO and shifts of domain III in pfKMO, stimulating flavin reduction. Interestingly, Ro 61-8048 has two different binding modes. It acts as a competitive inhibitor in scKMO and as a non-substrate effector in pfKMO. These findings provide understanding of the catalytic cycle of KMO and insight for structure-based drug design of KMO inhibitors.
Collapse
Affiliation(s)
- Hyun Tae Kim
- Crystalgenomics, Inc., 5F, Tower A, Korea Bio Park 700, Daewangpangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13524, Korea; Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea
| | - Byeong Kwan Na
- Crystalgenomics, Inc., 5F, Tower A, Korea Bio Park 700, Daewangpangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13524, Korea
| | - Jiwoung Chung
- Crystalgenomics, Inc., 5F, Tower A, Korea Bio Park 700, Daewangpangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13524, Korea
| | - Sulhee Kim
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea
| | - Sool Ki Kwon
- Crystalgenomics, Inc., 5F, Tower A, Korea Bio Park 700, Daewangpangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13524, Korea
| | - Hyunju Cha
- Crystalgenomics, Inc., 5F, Tower A, Korea Bio Park 700, Daewangpangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13524, Korea
| | - Jonghyeon Son
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea
| | - Joong Myung Cho
- Crystalgenomics, Inc., 5F, Tower A, Korea Bio Park 700, Daewangpangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13524, Korea.
| | - Kwang Yeon Hwang
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea.
| |
Collapse
|
36
|
Gao J, Yao L, Xia T, Liao X, Zhu D, Xiang Y. Biochemistry and structural studies of kynurenine 3-monooxygenase reveal allosteric inhibition by Ro 61-8048. FASEB J 2018; 32:2036-2045. [PMID: 29208702 DOI: 10.1096/fj.201700397rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The human kynurenine 3-monooxygenase (hKMO) is a potential therapeutic target for neurodegenerative and neurologic disorders. Inhibition of KMO by Ro 61-8048, a potent, selective, and the most widely used inhibitor of KMO, was shown effective in various models of neurodegenerative or neurologic disorders. However, the molecular basis of hKMO inhibition by Ro 61-8048 is not clearly understood. Here, we report biochemistry studies on hKMO and crystal structures of an hKMO homolog, pfKMO from Pseudomonas fluorescens, in complex with the substrate l-kynurenine and Ro 61-8048. We found that the C-terminal ∼110 aa are essential for the enzymatic activity of hKMO and the homologous C-terminal region of pfKMO folds into a distinct, all-α-helical domain, which associates with the N-terminal catalytic domain to form a unique tunnel in proximity to the substrate-binding pocket. The tunnel binds the Ro 61-8048 molecule, which fills most of the tunnel, and Ro 61-8048 is hydrogen bonded with several completely conserved residues, including an essential catalytic residue. Modification of Ro 61-8048 and biochemical studies of the modified Ro 61-8048 derivatives suggested that Ro 61-8048 inhibits the enzyme in an allosteric manner by affecting the conformation of the essential catalytic residue and by blocking entry of the substrate or product release. The unique binding sites distinguish Ro 61-8048 as a noncompetitive and highly selective inhibitor from other competitive inhibitors, which should facilitate further optimization of Ro 61-8048 and the development of new inhibitory drugs to hKMO.-Gao, J., Yao, L., Xia, T., Liao, X., Zhu, D., Xiang, Y. Biochemistry and structural studies of kynurenine 3-monooxygenase reveal allosteric inhibition by Ro 61-8048.
Collapse
Affiliation(s)
- Jingjing Gao
- School of Medicine, Center for Infectious Disease Research, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Advanced Innovation Center for Structural Biology
| | - Licheng Yao
- School of Pharmaceutical Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, China; and
| | - Tingting Xia
- School of Pharmaceutical Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, China; and
| | - Xuebin Liao
- School of Pharmaceutical Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, China; and
| | - Deyu Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Ye Xiang
- School of Medicine, Center for Infectious Disease Research, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Advanced Innovation Center for Structural Biology
| |
Collapse
|
37
|
Ghosh R, Tabrizi SJ. Clinical Features of Huntington's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1049:1-28. [PMID: 29427096 DOI: 10.1007/978-3-319-71779-1_1] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Huntington's disease (HD) is the most common monogenic neurodegenerative disease and the commonest genetic dementia in the developed world. With autosomal dominant inheritance, typically mid-life onset, and unrelenting progressive motor, cognitive and psychiatric symptoms over 15-20 years, its impact on patients and their families is devastating. The causative genetic mutation is an expanded CAG trinucleotide repeat in the gene encoding the Huntingtin protein, which leads to a prolonged polyglutamine stretch at the N-terminus of the protein. Since the discovery of the gene over 20 years ago much progress has been made in HD research, and although there are currently no disease-modifying treatments available, there are a number of exciting potential therapeutic developments in the pipeline. In this chapter we discuss the epidemiology, genetics and pathogenesis of HD as well as the clinical presentation and management of HD, which is currently focused on symptomatic treatment. The principles of genetic testing for HD are also explained. Recent developments in therapeutics research, including gene silencing and targeted small molecule approaches are also discussed, as well as the search for HD biomarkers that will assist the validation of these potentially new treatments.
Collapse
Affiliation(s)
- Rhia Ghosh
- UCL Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Sarah J Tabrizi
- UCL Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1N 3BG, UK.
| |
Collapse
|
38
|
Sathyasaikumar KV, Breda C, Schwarcz R, Giorgini F. Assessing and Modulating Kynurenine Pathway Dynamics in Huntington's Disease: Focus on Kynurenine 3-Monooxygenase. Methods Mol Biol 2018; 1780:397-413. [PMID: 29856028 DOI: 10.1007/978-1-4939-7825-0_18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The link between disturbances in kynurenine pathway (KP) metabolism and Huntington's disease (HD) pathogenesis has been explored for a number of years. Several novel genetic and pharmacological tools have recently been developed to modulate key regulatory steps in the KP such as the reaction catalyzed by the enzyme kynurenine 3-monooxygenase (KMO). This insight has offered new options for exploring the mechanistic link between this metabolic pathway and HD, and provided novel opportunities for the development of candidate drug-like compounds. Here, we present an overview of the field, focusing on some novel approaches for interrogating the pathway experimentally.
Collapse
Affiliation(s)
- Korrapati V Sathyasaikumar
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Carlo Breda
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK.
| |
Collapse
|
39
|
Dickey AS, La Spada AR. Therapy development in Huntington disease: From current strategies to emerging opportunities. Am J Med Genet A 2017; 176:842-861. [PMID: 29218782 DOI: 10.1002/ajmg.a.38494] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 09/08/2017] [Indexed: 12/13/2022]
Abstract
Huntington disease (HD) is a progressive autosomal dominant neurodegenerative disorder in which patients typically present with uncontrolled involuntary movements and subsequent cognitive decline. In 1993, a CAG trinucleotide repeat expansion in the coding region of the huntingtin (HTT) gene was identified as the cause of this disorder. This extended CAG repeat results in production of HTT protein with an expanded polyglutamine tract, leading to pathogenic HTT protein conformers that are resistant to protein turnover, culminating in cellular toxicity and neurodegeneration. Research into the mechanistic basis of HD has highlighted a role for bioenergetics abnormalities stemming from mitochondrial dysfunction, and for synaptic defects, including impaired neurotransmission and excitotoxicity. Interference with transcription regulation may underlie the mitochondrial dysfunction. Current therapies for HD are directed at treating symptoms, as there are no disease-modifying therapies. Commonly prescribed drugs for involuntary movement control include tetrabenazine, a potent and selective inhibitor of vesicular monoamine transporter 2 that depletes synaptic monoamines, and olanzapine, an atypical neuroleptic that blocks the dopamine D2 receptor. Various drugs are used to treat non-motor features. The HD therapeutic pipeline is robust, as numerous efforts are underway to identify disease-modifying treatments, with some small compounds and biological agents moving into clinical trials. Especially encouraging are dosage reduction strategies, including antisense oligonucleotides, and molecules directed at transcription dysregulation. Given the depth and breadth of current HD drug development efforts, there is reason to believe that disease-modifying therapies for HD will emerge, and this achievement will have profound implications for the entire neurotherapeutics field.
Collapse
Affiliation(s)
- Audrey S Dickey
- Departments of Neurology, Neurobiology, and Cell Biology, Duke Center for Neurodegeneration & Neurotherapeutics, Duke University Medical Center, Durham, North Carolina
| | - Albert R La Spada
- Departments of Neurology, Neurobiology, and Cell Biology, Duke Center for Neurodegeneration & Neurotherapeutics, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
40
|
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
|
41
|
Hutchinson JP, Rowland P, Taylor MRD, Christodoulou EM, Haslam C, Hobbs CI, Holmes DS, Homes P, Liddle J, Mole DJ, Uings I, Walker AL, Webster SP, Mowat CG, Chung CW. Structural and mechanistic basis of differentiated inhibitors of the acute pancreatitis target kynurenine-3-monooxygenase. Nat Commun 2017; 8:15827. [PMID: 28604669 PMCID: PMC5477544 DOI: 10.1038/ncomms15827] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 04/28/2017] [Indexed: 12/31/2022] Open
Abstract
Kynurenine-3-monooxygenase (KMO) is a key FAD-dependent enzyme of tryptophan metabolism. In animal models, KMO inhibition has shown benefit in neurodegenerative diseases such as Huntington's and Alzheimer's. Most recently it has been identified as a target for acute pancreatitis multiple organ dysfunction syndrome (AP-MODS); a devastating inflammatory condition with a mortality rate in excess of 20%. Here we report and dissect the molecular mechanism of action of three classes of KMO inhibitors with differentiated binding modes and kinetics. Two novel inhibitor classes trap the catalytic flavin in a previously unobserved tilting conformation. This correlates with picomolar affinities, increased residence times and an absence of the peroxide production seen with previous substrate site inhibitors. These structural and mechanistic insights culminated in GSK065(C1) and GSK366(C2), molecules suitable for preclinical evaluation. Moreover, revising the repertoire of flavin dynamics in this enzyme class offers exciting new opportunities for inhibitor design. Kynurenine-3-monooxygenase (KMO) is an emerging clinical target for treatment of neurodegenerative diseases and acute pancreatitis. Here, the authors report potent inhibitors that bind KMO in an unexpected conformation, offering structural and mechanistic insights for future drug discovery ventures.
Collapse
Affiliation(s)
| | - Paul Rowland
- Platform Technologies and Science, GlaxoSmithKline, Stevenage SG1 2NY, UK
| | - Mark R D Taylor
- EastChem School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, UK
| | | | - Carl Haslam
- Platform Technologies and Science, GlaxoSmithKline, Stevenage SG1 2NY, UK
| | - Clare I Hobbs
- Platform Technologies and Science, GlaxoSmithKline, Stevenage SG1 2NY, UK
| | - Duncan S Holmes
- Discovery Partnerships with Academia, GlaxoSmithKline, Stevenage, UK
| | - Paul Homes
- Platform Technologies and Science, GlaxoSmithKline, Stevenage SG1 2NY, UK
| | - John Liddle
- Discovery Partnerships with Academia, GlaxoSmithKline, Stevenage, UK
| | - Damian J Mole
- Medical Research Council Centre for Inflammation Research, Edinburgh EH16 4TJ, UK.,Clinical Surgery, University of Edinburgh, Edinburgh EH16 4SA, UK
| | - Iain Uings
- Discovery Partnerships with Academia, GlaxoSmithKline, Stevenage, UK
| | - Ann L Walker
- Discovery Partnerships with Academia, GlaxoSmithKline, Stevenage, UK
| | - Scott P Webster
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Christopher G Mowat
- EastChem School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, UK
| | - Chun-Wa Chung
- Platform Technologies and Science, GlaxoSmithKline, Stevenage SG1 2NY, UK
| |
Collapse
|