1
|
Malepathirana T, Senanayake D, Gautam V, Engel M, Balez R, Lovelace MD, Sundaram G, Heng B, Chow S, Marquis C, Guillemin GJ, Brew B, Jagadish C, Ooi L, Halgamuge S. Visualization of incrementally learned projection trajectories for longitudinal data. Sci Rep 2024; 14:13558. [PMID: 38866809 PMCID: PMC11169470 DOI: 10.1038/s41598-024-63511-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: 09/27/2023] [Accepted: 05/29/2024] [Indexed: 06/14/2024] Open
Abstract
Longitudinal studies that continuously generate data enable the capture of temporal variations in experimentally observed parameters, facilitating the interpretation of results in a time-aware manner. We propose IL-VIS (incrementally learned visualizer), a new machine learning pipeline that incrementally learns and visualizes a progression trajectory representing the longitudinal changes in longitudinal studies. At each sampling time point in an experiment, IL-VIS generates a snapshot of the longitudinal process on the data observed thus far, a new feature that is beyond the reach of classical static models. We first verify the utility and correctness of IL-VIS using simulated data, for which the true progression trajectories are known. We find that it accurately captures and visualizes the trends and (dis)similarities between high-dimensional progression trajectories. We then apply IL-VIS to longitudinal multi-electrode array data from brain cortical organoids when exposed to different levels of quinolinic acid, a metabolite contributing to many neuroinflammatory diseases including Alzheimer's disease, and its blocking antibody. We uncover valuable insights into the organoids' electrophysiological maturation and response patterns over time under these conditions.
Collapse
Affiliation(s)
- Tamasha Malepathirana
- Department of Mechanical Engineering, University of Melbourne, Melbourne, 3010, VIC, Australia
| | - Damith Senanayake
- Department of Mechanical Engineering, University of Melbourne, Melbourne, 3010, VIC, Australia
| | - Vini Gautam
- School of Chemical and Biomedical Engineering, University of Melbourne, Melbourne, 3010, VIC, Australia
- Centre for Nano Science and Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Martin Engel
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, 2522, NSW, Australia
| | - Rachelle Balez
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, 2522, NSW, Australia
| | - Michael D Lovelace
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St. Vincent's Centre for Applied Medical Research, 405 Liverpool St., Darlinghurst, Sydney, 2010, NSW, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St. Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, 2010, NSW, Australia
| | | | - Benjamin Heng
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia
| | - Sharron Chow
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia
| | - Christopher Marquis
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, 2052, NSW, Australia
| | - Gilles J Guillemin
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St. Vincent's Centre for Applied Medical Research, 405 Liverpool St., Darlinghurst, Sydney, 2010, NSW, Australia
- IPB University, Bogor, Indonesia
| | - Bruce Brew
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St. Vincent's Centre for Applied Medical Research, 405 Liverpool St., Darlinghurst, Sydney, 2010, NSW, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St. Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, 2010, NSW, Australia
- Departments of Neurology and Immunology, St. Vincent's Hospital, Sydney, 2010, NSW, Australia
| | - Chennupati Jagadish
- Research School of Physics, Australian National University, Canberra, 2601, ACT, Australia
| | - Lezanne Ooi
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, 2522, NSW, Australia.
| | - Saman Halgamuge
- Department of Mechanical Engineering, University of Melbourne, Melbourne, 3010, VIC, Australia.
| |
Collapse
|
2
|
Summers BS, Thomas Broome S, Pang TWR, Mundell HD, Koh Belic N, Tom NC, Ng ML, Yap M, Sen MK, Sedaghat S, Weible MW, Castorina A, Lim CK, Lovelace MD, Brew BJ. A Review of the Evidence for Tryptophan and the Kynurenine Pathway as a Regulator of Stem Cell Niches in Health and Disease. Int J Tryptophan Res 2024; 17:11786469241248287. [PMID: 38757094 PMCID: PMC11097742 DOI: 10.1177/11786469241248287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 04/03/2024] [Indexed: 05/18/2024] Open
Abstract
Stem cells are ubiquitously found in various tissues and organs in the body, and underpin the body's ability to repair itself following injury or disease initiation, though repair can sometimes be compromised. Understanding how stem cells are produced, and functional signaling systems between different niches is critical to understanding the potential use of stem cells in regenerative medicine. In this context, this review considers kynurenine pathway (KP) metabolism in multipotent adult progenitor cells, embryonic, haematopoietic, neural, cancer, cardiac and induced pluripotent stem cells, endothelial progenitor cells, and mesenchymal stromal cells. The KP is the major enzymatic pathway for sequentially catabolising the essential amino acid tryptophan (TRP), resulting in key metabolites including kynurenine, kynurenic acid, and quinolinic acid (QUIN). QUIN metabolism transitions into the adjoining de novo pathway for nicotinamide adenine dinucleotide (NAD) production, a critical cofactor in many fundamental cellular biochemical pathways. How stem cells uptake and utilise TRP varies between different species and stem cell types, because of their expression of transporters and responses to inflammatory cytokines. Several KP metabolites are physiologically active, with either beneficial or detrimental outcomes, and evidence of this is presented relating to several stem cell types, which is important as they may exert a significant impact on surrounding differentiated cells, particularly if they metabolise or secrete metabolites differently. Interferon-gamma (IFN-γ) in mesenchymal stromal cells, for instance, highly upregulates rate-limiting enzyme indoleamine-2,3-dioxygenase (IDO-1), initiating TRP depletion and production of metabolites including kynurenine/kynurenic acid, known agonists of the Aryl hydrocarbon receptor (AhR) transcription factor. AhR transcriptionally regulates an immunosuppressive phenotype, making them attractive for regenerative therapy. We also draw attention to important gaps in knowledge for future studies, which will underpin future application for stem cell-based cellular therapies or optimising drugs which can modulate the KP in innate stem cell populations, for disease treatment.
Collapse
Affiliation(s)
- Benjamin Sebastian Summers
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St. Vincent’s Centre for Applied Medical Research, Sydney, NSW, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, UNSW Sydney, NSW, Australia
| | - Sarah Thomas Broome
- Faculty of Science, Laboratory of Cellular and Molecular Neuroscience, School of Life Sciences, University of Technology Sydney, NSW, Australia
| | | | - Hamish D Mundell
- Faculty of Medicine and Health, New South Wales Brain Tissue Resource Centre, School of Medical Sciences, Charles Perkins Centre, University of Sydney, NSW, Australia
| | - Naomi Koh Belic
- School of Life Sciences, University of Technology, Sydney, NSW, Australia
| | - Nicole C Tom
- Formerly of the Department of Physiology, University of Sydney, NSW, Australia
| | - Mei Li Ng
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Maylin Yap
- Formerly of the Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Monokesh K Sen
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St. Vincent’s Centre for Applied Medical Research, Sydney, NSW, Australia
- School of Medicine, Western Sydney University, NSW, Australia
- Faculty of Medicine and Health, School of Medical Sciences, Charles Perkins Centre, The University of Sydney, NSW, Australia
| | - Sara Sedaghat
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Michael W Weible
- School of Environment and Science, Griffith University, Brisbane, QLD, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, Australia
| | - Alessandro Castorina
- Faculty of Science, Laboratory of Cellular and Molecular Neuroscience, School of Life Sciences, University of Technology Sydney, NSW, Australia
| | - Chai K Lim
- Faculty of Medicine, Macquarie University, Sydney, NSW, Australia
| | - Michael D Lovelace
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St. Vincent’s Centre for Applied Medical Research, Sydney, NSW, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, UNSW Sydney, NSW, Australia
| | - Bruce J Brew
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St. Vincent’s Centre for Applied Medical Research, Sydney, NSW, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, UNSW Sydney, NSW, Australia
- Departments of Neurology and Immunology, St. Vincent’s Hospital, Sydney, NSW, Australia
- University of Notre Dame, Darlinghurst, Sydney, NSW, Australia
| |
Collapse
|
3
|
Sharma VK, Singh TG, Prabhakar NK, Mannan A. Kynurenine Metabolism and Alzheimer's Disease: The Potential Targets and Approaches. Neurochem Res 2022; 47:1459-1476. [PMID: 35133568 DOI: 10.1007/s11064-022-03546-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 12/12/2022]
Abstract
L-tryptophan, an essential amino acid, regulates protein homeostasis and plays a role in neurotransmitter-mediated physiological events. It also influences age-associated neurological alterations and neurodegenerative changes. The metabolism of tryptophan is carried majorly through the kynurenine route, leading to the production of several pharmacologically active enzymes, substrates, and metabolites. These metabolites and enzymes influence a variety of physiological and pathological outcomes of the majority of systems, including endocrine, haemopoietic, gastrointestinal, immunomodulatory, inflammatory, bioenergetic metabolism, and neuronal functions. An extensive literature review of PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was carried out to understand the nature of the extensive work done on the kynurenine metabolites that influence cellular redox potential, immunoregulatory mechanisms, inflammatory pathways, cell survival channels, and cellular communication in close association with several neurodegenerative changes. The imbalanced state of kynurenine pathways has found a close association to several pathological disorders, including HIV infections, cancer, autoimmune disorders, neurodegenerative and neurological disorders including Parkinson's disease, epilepsy and has found special attention in Alzheimer's disease (AD). Kynurenine pathway (KP) is intricately linked to AD pathogenesis owing to the influence of kynurenine metabolites on excitotoxic neurotransmission, oxidative stress, uptake of neurotransmitters, and modulation of neuroinflammation, amyloid aggregation, microtubule disruption, and their ability to induce a state of dysbiosis. Pharmacological modulation of KP pathways has shown encouraging results, indicating that it may be a viable and explorable target for the therapy of AD.
Collapse
Affiliation(s)
- Vivek Kumar Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Govt. College of Pharmacy, Rohru, Shimla, Himachal Pradesh, 171207, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| | | | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| |
Collapse
|
4
|
Liang Y, Xie S, He Y, Xu M, Qiao X, Zhu Y, Wu W. Kynurenine Pathway Metabolites as Biomarkers in Alzheimer's Disease. DISEASE MARKERS 2022; 2022:9484217. [PMID: 35096208 PMCID: PMC8791723 DOI: 10.1155/2022/9484217] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/21/2021] [Accepted: 12/31/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that deteriorates cognitive function. Patients with AD generally exhibit neuroinflammation, elevated beta-amyloid (Aβ), tau phosphorylation (p-tau), and other pathological changes in the brain. The kynurenine pathway (KP) and several of its metabolites, especially quinolinic acid (QA), are considered to be involved in the neuropathogenesis of AD. The important metabolites and key enzymes show significant importance in neuroinflammation and AD. Meanwhile, the discovery of changed levels of KP metabolites in patients with AD suggests that KP metabolites may have a prominent role in the pathogenesis of AD. Further, some KP metabolites exhibit other effects on the brain, such as oxidative stress regulation and neurotoxicity. Both analogs of the neuroprotective and antineuroinflammation metabolites and small molecule enzyme inhibitors preventing the formation of neurotoxic and neuroinflammation compounds may have potential therapeutic significance. This review focused on the KP metabolites through the relationship of neuroinflammation in AD, significant KP metabolites, and associated molecular mechanisms as well as the utility of these metabolites as biomarkers and therapeutic targets for AD. The objective is to provide references to find biomarkers and therapeutic targets for patients with AD.
Collapse
Affiliation(s)
- Yuqing Liang
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, China
| | - Shan Xie
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, China
| | - Yanyun He
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, China
| | - Manru Xu
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, China
| | - Xi Qiao
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, China
| | - Yue Zhu
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, China
| | - Wenbin Wu
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, China
| |
Collapse
|
5
|
Sil S, Periyasamy P, Thangaraj A, Niu F, Chemparathy DT, Buch S. Advances in the Experimental Models of HIV-Associated Neurological Disorders. Curr HIV/AIDS Rep 2021; 18:459-474. [PMID: 34427869 DOI: 10.1007/s11904-021-00570-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Involvement of the central nervous system (CNS) in HIV-1 infection is commonly associated with neurological disorders and cognitive impairment, commonly referred to as HIV-associated neurocognitive disorders (HAND). Severe and progressive neurocognitive impairment is rarely observed in the post-cART era; however, asymptomatic and mild neurocognitive disorders still exist, despite viral suppression. Additionally, comorbid conditions can also contribute to the pathogenesis of HAND. RECENT FINDINGS In this review, we summarize the characterization of HAND, factors contributing, and the functional impairments in both preclinical and clinical models. Specifically, we also discuss recent advances in the animal models of HAND and in in vitro cultures and the potential role of drugs of abuse in this model system of HAND. Potential peripheral biomarkers associated with HAND are also discussed. Overall, this review identifies some of the recent advances in the field of HAND in cell culture studies, animal models, clinical findings, and the limitations of each model system, which can play a key role in developing novel therapeutics in the field.
Collapse
Affiliation(s)
- Susmita Sil
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA.
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA.
| | - Annadurai Thangaraj
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Fang Niu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Divya T Chemparathy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| |
Collapse
|
6
|
Pharmacokinetic Properties of the Novel Synthetic Cannabinoid 5F-APINAC and Its Influence on Metabolites Associated with Neurotransmission in Rabbit Plasma. Pharmaceuticals (Basel) 2021; 14:ph14070668. [PMID: 34358094 PMCID: PMC8308683 DOI: 10.3390/ph14070668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 11/23/2022] Open
Abstract
The strong psychoactive effects of synthetic cannabinoids raise the need for the deeper studying of their neurometabolic effects. The pharmacokinetic properties of 5F-APINAC and its influence on metabolomics profiles associated with neurotransmission were investigated in rabbit plasma. Twelve rabbits divided into three groups received 1-mL 5F-APINAC at 0.1, 1 and 2 mg/kg. The intervention groups were compared with the controls. Sampling was performed at nine time points (0–24 h). Ultra-high-performance liquid chromatography–tandem mass spectrometry was used. The pharmacokinetics were dose-dependent (higher curve at a higher dose) with a rapid biotransformation, followed by gradual elimination within 24 h. The tryptophan concentrations abruptly decreased (p < 0.05) in all tested groups, returning to the basal levels after 6 h. 5-hydroxylindole acetic acid increased (p < 0.05) in the controls, but this trend was absent in the treated groups. The aspartic acid concentrations were elevated (p < 0.001) in the treated groups. L-kynurenine was elevated (p < 0.01) in the intervention groups receiving 1 mg/kg to 2 mg/kg. Dose-dependent elevations (p < 0.01) were found for kynurenic acid, xanthurenic acid and quinolinic acid (p < 0.01), whereas the anthranilic acid trends were decreased (p < 0.01). The indole-3-propionic acid and indole-3-carboxaldehyde trends were elevated (p < 0.05), whereas the indole-3-lactic acid trajectories were decreased (p < 0.01) in the intervention groups. 5F-APINAC administration had a rapid biotransformation and gradual elimination. The metabolites related to the kynurenine and serotonergic system/serotonin pathways, aspartic acid innervation system and microbial tryptophan catabolism were altered.
Collapse
|
7
|
Sandi D, Fricska-Nagy Z, Bencsik K, Vécsei L. Neurodegeneration in Multiple Sclerosis: Symptoms of Silent Progression, Biomarkers and Neuroprotective Therapy-Kynurenines Are Important Players. Molecules 2021; 26:molecules26113423. [PMID: 34198750 PMCID: PMC8201043 DOI: 10.3390/molecules26113423] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 12/17/2022] Open
Abstract
Neurodegeneration is one of the driving forces behind the pathogenesis of multiple sclerosis (MS). Progression without activity, pathopsychological disturbances (cognitive impairment, depression, fatigue) and even optic neuropathy seems to be mainly routed in this mechanism. In this article, we aim to give a comprehensive review of the clinical aspects and symptomology, radiological and molecular markers and potential therapeutic targets of neurodegeneration in connection with MS. As the kynurenine pathway (KP) was evidenced to play an important role in the pathogenesis of other neurodegenerative conditions (even implied to have a causative role in some of these diseases) and more and more recent evidence suggest the same central role in the neurodegenerative processes of MS as well, we pay special attention to the KP. Metabolites of the pathway are researched as biomarkers of the disease and new, promising data arising from clinical evaluations show the possible therapeutic capability of KP metabolites as neuroprotective drugs in MS. Our conclusion is that the kynurenine pathway is a highly important route of research both for diagnostic and for therapeutic values and is expected to yield concrete results for everyday medicine in the future.
Collapse
Affiliation(s)
- Dániel Sandi
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, H-6725 Szeged, Hungary; (D.S.); (Z.F.-N.); (K.B.)
| | - Zsanett Fricska-Nagy
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, H-6725 Szeged, Hungary; (D.S.); (Z.F.-N.); (K.B.)
| | - Krisztina Bencsik
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, H-6725 Szeged, Hungary; (D.S.); (Z.F.-N.); (K.B.)
| | - László Vécsei
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, H-6725 Szeged, Hungary; (D.S.); (Z.F.-N.); (K.B.)
- MTA-SZTE Neuroscience Research Group, University of Szeged, H-6725 Szeged, Hungary
- Interdisciplinary Excellence Centre, University of Szeged, H-6725 Szeged, Hungary
- Correspondence: ; Tel.: +36-62-545-384; Fax: +36-62-545-597
| |
Collapse
|
8
|
Collier ME, Zhang S, Scrutton NS, Giorgini F. Inflammation control and improvement of cognitive function in COVID-19 infections: is there a role for kynurenine 3-monooxygenase inhibition? Drug Discov Today 2021; 26:1473-1481. [PMID: 33609782 PMCID: PMC7889466 DOI: 10.1016/j.drudis.2021.02.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/26/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
The novel respiratory virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), emerged during late 2019 and spread rapidly across the world. It is now recognised that the nervous system can be affected in COVID-19, with several studies reporting long-term cognitive problems in patients. The metabolic pathway of tryptophan degradation, known as the kynurenine pathway (KP), is significantly activated in patients with COVID-19. KP metabolites have roles in regulating both inflammatory/immune responses and neurological functions. In this review, we speculate on the effects of KP activation in patients with COVID-19, and how modulation of this pathway might impact inflammation and reduce neurological symptoms.
Collapse
Affiliation(s)
- Mary Ew Collier
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK.
| | - Shaowei Zhang
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, The University of Manchester, 131 Princess Street, Manchester M1 7DN, 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
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| |
Collapse
|
9
|
Pukoli D, Polyák H, Rajda C, Vécsei L. Kynurenines and Neurofilament Light Chain in Multiple Sclerosis. Front Neurosci 2021; 15:658202. [PMID: 34113231 PMCID: PMC8185147 DOI: 10.3389/fnins.2021.658202] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/29/2021] [Indexed: 12/30/2022] Open
Abstract
Multiple sclerosis is an autoimmune, demyelinating, and neurodegenerative disease of the central nervous system. In recent years, it has been proven that the kynurenine system plays a significant role in the development of several nervous system disorders, including multiple sclerosis. Kynurenine pathway metabolites have both neurotoxic and neuroprotective effects. Moreover, the enzymes of the kynurenine pathway play an important role in immunomodulation processes, among others, as well as interacting with neuronal energy balance and various redox reactions. Dysregulation of many of the enzymatic steps in kynurenine pathway and upregulated levels of these metabolites locally in the central nervous system, contribute to the progression of multiple sclerosis pathology. This process can initiate a pathogenic cascade, including microglia activation, glutamate excitotoxicity, chronic oxidative stress or accumulated mitochondrial damage in the axons, that finally disrupt the homeostasis of neurons, leads to destabilization of neuronal cell cytoskeleton, contributes to neuro-axonal damage and neurodegeneration. Neurofilaments are good biomarkers of the neuro-axonal damage and their level reliably indicates the severity of multiple sclerosis and the treatment response. There is increasing evidence that connections exist between the molecules generated in the kynurenine metabolic pathway and the change in neurofilament concentrations. Thus the alterations in the kynurenine pathway may be an important biomarker of the course of multiple sclerosis. In our present review, we report the possible relationship and connection between neurofilaments and the kynurenine system in multiple sclerosis based on the available evidences.
Collapse
Affiliation(s)
- Dániel Pukoli
- Department of Neurology, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Department of Neurology, Vaszary Kolos Hospital, Esztergom, Hungary
| | - Helga Polyák
- Department of Neurology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Cecilia Rajda
- Department of Neurology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Neuroscience Research Group, Department of Neurology, Faculty of Medicine, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| |
Collapse
|
10
|
Hunt C, Macedo E Cordeiro T, Suchting R, de Dios C, Cuellar Leal VA, Soares JC, Dantzer R, Teixeira AL, Selvaraj S. Effect of immune activation on the kynurenine pathway and depression symptoms - A systematic review and meta-analysis. Neurosci Biobehav Rev 2020; 118:514-523. [PMID: 32853625 DOI: 10.1016/j.neubiorev.2020.08.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/30/2020] [Accepted: 08/14/2020] [Indexed: 12/28/2022]
Abstract
Dysregulated kynurenine (KYN) pathway has been implicated in the pathophysiology of depression. In this systematic review, we examined the relationship between kynurenine pathway metabolites (KYN, kynurenic acid KYNA, tryptophan TRP, quinolinic acid QUIN, KYN/TRP ratio) and depression symptoms in the context of pro-inflammatory activation and immune response. Out of 5,082 articles, fifteen studies were suitable; ten studies (N = 315 medically ill patients treated with interferon-alpha IFN-α) reported baseline and post-intervention plasma KYN, TRP and KYN/TRP ratios which were included in quantitative meta-analysis. Data from five studies were summarized (IFN-α, interferon-beta IFN-β, and lipopolysaccharide LPS). We found that IFN-α treatment in patients with chronic illnesses was associated with decreased TRP, increased levels of KYN and KYN/TRP ratio and depression scores from baseline to follow-up at both 4 and 24 weeks. Our findings suggest that increased risk of depression observed after immune-activating agents in patients with chronic medical illnesses is likely mediated by the kynurenine pathway. Further prospective studies are required to investigate the exact pathophysiology of the KYN pathway in depression.
Collapse
Affiliation(s)
- Charlotte Hunt
- The University of Texas Health Science Center at Houston, McGovern Medical School, 6431 Fannin St, Houston, TX, 77030, USA
| | - Thiago Macedo E Cordeiro
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), 1941 East Rd, Houston, TX, 77054, USA
| | - Robert Suchting
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), 1941 East Rd, Houston, TX, 77054, USA
| | - Constanza de Dios
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), 1941 East Rd, Houston, TX, 77054, USA
| | - Valeria A Cuellar Leal
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), 1941 East Rd, Houston, TX, 77054, USA
| | - Jair C Soares
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), 1941 East Rd, Houston, TX, 77054, USA
| | - Robert Dantzer
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Antonio L Teixeira
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), 1941 East Rd, Houston, TX, 77054, USA
| | - Sudhakar Selvaraj
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), 1941 East Rd, Houston, TX, 77054, USA; Houston Methodist Research Institute, Institute for Academic Medicine, 6670 Bertner St., Houston, TX, 77030, USA.
| |
Collapse
|
11
|
Biernacki T, Sandi D, Bencsik K, Vécsei L. Kynurenines in the Pathogenesis of Multiple Sclerosis: Therapeutic Perspectives. Cells 2020; 9:cells9061564. [PMID: 32604956 PMCID: PMC7349747 DOI: 10.3390/cells9061564] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
Over the past years, an increasing amount of evidence has emerged in support of the kynurenine pathway’s (KP) pivotal role in the pathogenesis of several neurodegenerative, psychiatric, vascular and autoimmune diseases. Different neuroactive metabolites of the KP are known to exert opposite effects on neurons, some being neuroprotective (e.g., picolinic acid, kynurenic acid, and the cofactor nicotinamide adenine dinucleotide), while others are toxic to neurons (e.g., 3-hydroxykynurenine, quinolinic acid). Not only the alterations in the levels of the metabolites but also disturbances in their ratio (quinolinic acid/kynurenic acid) have been reported in several diseases. In addition to the metabolites, the enzymes participating in the KP have been unearthed to be involved in modulation of the immune system, the energetic upkeep of neurons and have been shown to influence redox processes and inflammatory cascades, revealing a sophisticated, intertwined system. This review considers various methods through which enzymes and metabolites of the kynurenine pathway influence the immune system, the roles they play in the pathogenesis of neuroinflammatory diseases based on current evidence with a focus on their involvement in multiple sclerosis, as well as therapeutic approaches.
Collapse
Affiliation(s)
- Tamás Biernacki
- Department of Neurology, Faculty of General Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, H-6725 Szeged, Hungary; (T.B.); (D.S.); (K.B.)
| | - Dániel Sandi
- Department of Neurology, Faculty of General Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, H-6725 Szeged, Hungary; (T.B.); (D.S.); (K.B.)
| | - Krisztina Bencsik
- Department of Neurology, Faculty of General Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, H-6725 Szeged, Hungary; (T.B.); (D.S.); (K.B.)
| | - László Vécsei
- Department of Neurology, Faculty of General Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, H-6725 Szeged, Hungary; (T.B.); (D.S.); (K.B.)
- MTA—SZTE Neuroscience Research Group, H-6725 Szeged, Hungary
- Interdisciplinary Excellence Center, University of Szeged, H-6720 Szeged, Hungary
- Correspondence: ; Tel.: +36-62-545-356; Fax: +36-62-545-597
| |
Collapse
|
12
|
Williams ME, Zulu SS, Stein DJ, Joska JA, Naudé PJW. Signatures of HIV-1 subtype B and C Tat proteins and their effects in the neuropathogenesis of HIV-associated neurocognitive impairments. Neurobiol Dis 2019; 136:104701. [PMID: 31837421 DOI: 10.1016/j.nbd.2019.104701] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/18/2019] [Accepted: 12/08/2019] [Indexed: 11/16/2022] Open
Abstract
HIV-associated neurocognitive impairments (HANI) are a spectrum of neurological disorders due to the effects of HIV-1 on the central nervous system (CNS). The HIV-1 subtypes; HIV-1 subtype B (HIV-1B) and HIV-1 subtype C (HIV-1C) are responsible for the highest prevalence of HANI and HIV infections respectively. The HIV transactivator of transcription (Tat) protein is a major contributor to the neuropathogenesis of HIV. The effects of the Tat protein on cells of the CNS is determined by the subtype-associated amino acid sequence variations. The extent to which the sequence variation between Tat-subtypes contribute to underlying mechanisms and neurological outcomes are not clear. In this review of the literature, we discuss how amino acid variations between HIV-1B Tat (TatB) and HIV-1C Tat (TatC) proteins contribute to the potential underlying neurobiological mechanisms of HANI. Tat-C is considered to be a more effective transactivator, whereas Tat-B may exert increased neurovirulence, including neuronal apoptosis, monocyte infiltration into the brain, (neuro)inflammation, oxidative stress and blood-brain barrier damage. These findings support the premise that Tat variants from different HIV-1 subtypes may direct neurovirulence and neurological outcomes in HANI.
Collapse
Affiliation(s)
- Monray E Williams
- Department of Psychiatry and Mental Health and Neuroscience Institute, Brain Behaviour Unit, University of Cape Town, Cape Town, South Africa.
| | - Simo S Zulu
- Department of Psychiatry and Mental Health and Neuroscience Institute, Brain Behaviour Unit, University of Cape Town, Cape Town, South Africa
| | - Dan J Stein
- Department of Psychiatry and Mental Health and Neuroscience Institute, Brain Behaviour Unit, University of Cape Town, Cape Town, South Africa; SAMRC Unit on Risk and Resilience in Mental Disorders and Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - John A Joska
- Division of Neuropsychiatry, Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Petrus J W Naudé
- Department of Psychiatry and Mental Health and Neuroscience Institute, Brain Behaviour Unit, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
13
|
Rahman A, Al-Qenaie S, Rao MS, Khan KM, Guillemin GJ. Memantine Is Protective against Cytotoxicity Caused by Lead and Quinolinic Acid in Cultured Rat Embryonic Hippocampal Cells. Chem Res Toxicol 2019; 32:1134-1143. [PMID: 30950269 DOI: 10.1021/acs.chemrestox.8b00421] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Quinolinic acid (QA) is an excitotoxic metabolite of the kynurenine pathway of tryptophan metabolism produced in response to inflammation and oxidative stress. Lead (Pb) causes oxidative stress and thus may produce neurotoxicity by increasing QA production. We investigated the in vitro cytotoxic effects of Pb and QA and the protective effects of the NMDA receptor antagonist memantine. Primary cultures of embryonic hippocampal cells from Wistar rats were treated with different concentrations of Pb, QA, and Pb + QA with and without memantine. Cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). Apoptosis was analyzed by flow cytometry after Annexin-V/propidium iodide staining. The numbers of immunostained neurons (with β3-Tubulin; Tuj1) and astrocytes (with glial fibrillary acidic protein) were counted. Pb at 20 μg/dL (0.97 μM) and QA at 500 nM concentrations showed significant cytotoxic effects, as evidenced by decreased cell viability, increased apoptosis, and a decrease in the number of both astrocytes and neurons. The combination of Pb and QA showed significant synergistic apoptotic effects at lower doses. Memantine (500 nM) was largely protective against the cytotoxic effects of both Pb and QA, suggesting that Pb's and QA's cytotoxicity involves NMDA receptor activation. Whereas the neuroprotection by memantine from QA-induced toxicity has been previously reported, this is the first study reporting the protection by memantine against Pb-induced cytotoxicity in cultured hippocampal cells. Protection by memantine against these neurotoxicants in vivo needs to be investigated.
Collapse
Affiliation(s)
- Abdur Rahman
- Department of Food Science and Nutrition, College of Life Sciences , Kuwait University , 13060 Kuwait City , Kuwait
| | - Sara Al-Qenaie
- Department of Food Science and Nutrition, College of Life Sciences , Kuwait University , 13060 Kuwait City , Kuwait.,Kuwait Oil Company Hospital , 61008 Ahmadi , Kuwait
| | - Muddanna S Rao
- Department of Anatomy, Faculty of Medicine , Kuwait University , 13060 Kuwait City , Kuwait
| | - Khalid M Khan
- Department of Anatomy, Faculty of Medicine , Kuwait University , 13060 Kuwait City , Kuwait
| | - Gilles J Guillemin
- Neuroinflammation Group, Faculty of Medicine and Health Sciences , Macquarie University , Macquarie Park , New South Wales 2109 , Australia
| |
Collapse
|
14
|
Rahman A, Rao MS, Khan KM. Intraventricular infusion of quinolinic acid impairs spatial learning and memory in young rats: a novel mechanism of lead-induced neurotoxicity. J Neuroinflammation 2018; 15:263. [PMID: 30217162 PMCID: PMC6137743 DOI: 10.1186/s12974-018-1306-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/04/2018] [Indexed: 12/20/2022] Open
Abstract
Background Lead (Pb), a heavy metal, and quinolinic acid (QA), a metabolite of the kynurenine pathway of tryptophan metabolism, are known neurotoxicants. Both Pb and QA impair spatial learning and memory. Pb activates astrocytes and microglia, which in turn induce the synthesis of QA. We hypothesized increased QA production in response to Pb exposure as a novel mechanism of Pb-neurotoxicity. Methods Two experimental paradigms were used. In experiment one, Wistar rat pups were exposed to Pb via their dams’ drinking water from postnatal day 1 to 21. Control group was given regular water. In the second protocol, QA (9 mM) or normal saline (as Vehicle Control) was infused into right lateral ventricle of 21-day old rats for 7 days using osmotic pumps. Learning and memory were assessed by Morris water maze test on postnatal day 30 or 45 in both Pb- and QA-exposed rats. QA levels in the Pb exposed rats were measured in blood by ELISA and in the brain by immunohistochemistry on postnatal days 45 and 60. Expression of various molecules involved in learning and memory was analyzed by Western blot. Means of control and experimental groups were compared with two-way repeated measure ANOVA (learning) and t test (all other variables). Results Pb exposure increased QA level in the blood (by ~ 58%) and increased (p < 0.05) the number of QA-immunoreactive cells in the cortex, and CA1, CA3 and dentate gyrus regions of the hippocampus, compared to control rats. In separate experiments, QA infusion impaired learning and short-term memory similar to Pb. PSD-95, PP1, and PP2A were decreased (p < 0.05) in the QA-infused rats, whereas tau phosphorylation was increased, compared to vehicle infused rats. Conclusion Putting together the results of the two experimental paradigms, we propose that increased QA production in response to Pb exposure is a novel mechanism of Pb-induced neurotoxicity.
Collapse
Affiliation(s)
- Abdur Rahman
- Department of Food Science and Nutrition, College of Life Sciences, Kuwait University, Kuwait City, Kuwait.
| | - Muddanna S Rao
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Khalid M Khan
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
15
|
Aguilera-Portillo G, Rangel-López E, Villeda-Hernández J, Chavarría A, Castellanos P, Elmazoglu Z, Karasu Ç, Túnez I, Pedraza G, Königsberg M, Santamaría A. The Pharmacological Inhibition of Fatty Acid Amide Hydrolase Prevents Excitotoxic Damage in the Rat Striatum: Possible Involvement of CB1 Receptors Regulation. Mol Neurobiol 2018; 56:844-856. [DOI: 10.1007/s12035-018-1129-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023]
|
16
|
Feng W, Wang Y, Liu ZQ, Zhang X, Han R, Miao YZ, Qin ZH. Microglia activation contributes to quinolinic acid-induced neuronal excitotoxicity through TNF-α. Apoptosis 2018; 22:696-709. [PMID: 28315174 DOI: 10.1007/s10495-017-1363-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
It has been reported that activation of NF-κB is involved in excitotoxicity; however, it is not fully understood how NF-κB contributes to excitotoxicity. The aim of this study is to investigate if NF-κB contributes to quinolinic acid (QA)-mediated excitotoxicity through activation of microglia. In the cultured primary cortical neurons and microglia BV-2 cells, the effects of QA on cell survival, NF-κB expression and cytokines production were investigated. The effects of BV-2-conditioned medium (BCM) on primary cortical neurons were examined. The effects of pyrrolidine dithiocarbamate (PDTC), an inhibitor of NF-κB, and minocycline (MC), an inhibitor of microglia activation, on QA-induced excitotoxicity were assessed. QA-induced NF-κB activation and TNF-α secretion, and the roles of TNF-α in excitotoxicity were studied. QA at the concentration below 1 mM had no apparent toxic effects on cultured primary neurons or BV-2 cells. However, addition of QA-primed BCM to primary neurons did aggravate QA-induced excitotoxicity. The exacerbation of QA-induced excitotoxicity by BCM was partially ameliorated by inhibiting NF-κB and microglia activation. QA induced activation of NF-κB and upregulation of TNF-α in BV-2 cells. Addition of recombinant TNF-α mimicked QA-induced excitotoxic effects on neurons, and neutralizing TNF-α with specific antibodies partially abolished exacerbation of QA-induced excitotoxicity by BCM. These studies suggested that QA activated microglia and upregulated TNF-α through NF-κB pathway in microglia. The microglia-mediated inflammatory pathway contributed, at least in part, to QA-induced excitotoxicity.
Collapse
Affiliation(s)
- Wei Feng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases (SZS0703), College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Yan Wang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases (SZS0703), College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Zi-Qi Liu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases (SZS0703), College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Xuan Zhang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases (SZS0703), College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China.,Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Rong Han
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases (SZS0703), College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - You-Zhu Miao
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases (SZS0703), College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases (SZS0703), College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
17
|
de Paula Martins R, Ghisoni K, Lim CK, Aguiar AS, Guillemin GJ, Latini A. Neopterin preconditioning prevents inflammasome activation in mammalian astrocytes. Free Radic Biol Med 2018; 115:371-382. [PMID: 29198726 DOI: 10.1016/j.freeradbiomed.2017.11.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 11/23/2017] [Accepted: 11/27/2017] [Indexed: 12/13/2022]
Abstract
Neopterin, a well-established biomarker for immune system activation, is found at increased levels in the cerebrospinal fluid of individuals affected by neurological/neurodegenerative diseases. Here, neopterin synthesis was investigated in different nerve cells (rodent and human) and in the mouse hippocampus under inflammatory stimuli. We also aimed to investigate whether neopterin preconditioning could modulate the inflammasome activation, a component of the innate immune system. Increased neopterin was detected in human nerve cells supernatants (highest secretion in astrocytes) exposed to lipopolysaccharide (LPS) and interferon-gamma (INF-γ) and in the hippocampus of mice receiving LPS (0.33mg/kg; intraperitoneal). In parallel to the hippocampal-increased neopterin, it was observed a significant increase in the expression of the rate-limiting enzyme of its biosynthetic pathway, and both phenomena occurred before the inflammasome activation. Moreover, a significant inhibition of the inflammasome activation was observed in neopterin pre-conditioned human astrocytes, when challenged with LPS, by reducing IL-1β, caspase-1 and ASC expression or content, components of the NLRP3 inflammasome. Mechanistically, neopterin might induce eletrophilic stress and consequently the nuclear translocation of the transcription factor Nrf-2, and the anti-inflammatory cytokines IL-10 and IL-1ra release, which would induce the inhibition of the inflammasome activation. Altogether, this strongly suggests an essential role of neopterin during inflammatory processes.
Collapse
Affiliation(s)
- Roberta de Paula Martins
- Laboratório de Bioenergética e Estresse Oxidativo - LABOX, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Campus Universitário, Córrego Grande, Florianópolis, SC 88040-900, Brazil; Faculty of Medicine and Health Sciences, Macquarie University, North Ryde, New South Wales 2109, Australia
| | - Karina Ghisoni
- Laboratório de Bioenergética e Estresse Oxidativo - LABOX, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Campus Universitário, Córrego Grande, Florianópolis, SC 88040-900, Brazil
| | - Chai K Lim
- Faculty of Medicine and Health Sciences, Macquarie University, North Ryde, New South Wales 2109, Australia
| | - Aderbal Silva Aguiar
- Laboratório de Bioenergética e Estresse Oxidativo - LABOX, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Campus Universitário, Córrego Grande, Florianópolis, SC 88040-900, Brazil
| | - Gilles J Guillemin
- Faculty of Medicine and Health Sciences, Macquarie University, North Ryde, New South Wales 2109, Australia
| | - Alexandra Latini
- Laboratório de Bioenergética e Estresse Oxidativo - LABOX, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Campus Universitário, Córrego Grande, Florianópolis, SC 88040-900, Brazil.
| |
Collapse
|
18
|
Loganathan C, Thayumanavan P. Asiatic acid prevents the quinolinic acid-induced oxidative stress and cognitive impairment. Metab Brain Dis 2018; 33:151-159. [PMID: 29086235 DOI: 10.1007/s11011-017-0143-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 10/23/2017] [Indexed: 02/08/2023]
Abstract
Increased accumulation of endogenous neurotoxin quinolinic acid has been found in various neurodegenerative diseases. Oxidative stress caused by quinolinic acid is considered as imperative factor for its toxicity. Asiatic acid, a natural triterpene is widely studied for its various medicinal values. In the present study the effects of asiatic acid in preventing the cognitive impairment and oxidative stress caused by quinolinic acid was investigated. Male Spraque-Dawley rats were orally administered asiatic acid (30 mg/kg/day) for 28 days, while quinolinic acid toxicity-induced animals received quinolinic acid (1.5 mmol/kg/day) from day 15 to day 28 for 14 days. Asiatic acid administration prevented the loss of spatial memory caused due to quinolinic acid-induced toxicity as determined using the novel object location test. In addition, asiatic acid administration alleviated the deleterious effect of quinolinic acid in brain such as increased oxidative stress, decreased antioxidant status and mitochondrial oxidative phosphorylation dysfunction. These data demonstrate that asiatic acid through its potent antioxidant and cognition enhancement property prevented the neuronal impairments caused by quinolinic acid.
Collapse
Affiliation(s)
- Chitra Loganathan
- Department of Biochemistry, Periyar University, Salem, Tamil Nadu, 636011, India
| | | |
Collapse
|
19
|
Fu Y, Hsiao JHT, Paxinos G, Halliday GM, Kim WS. ABCA7 Mediates Phagocytic Clearance of Amyloid-β in the Brain. J Alzheimers Dis 2018; 54:569-84. [PMID: 27472885 DOI: 10.3233/jad-160456] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by dementia and abnormal deposits of aggregated amyloid-β in the brain. Recent genome-wide association studies have revealed that ABCA7 is strongly associated with AD. In vitro evidence suggests that the role of ABCA7 is related to phagocytic activity. Deletion of ABCA7 in a mouse model of AD exacerbates cerebral amyloid-β plaque load. However, the biological role of ABCA7 in AD brain pathogenesis is unknown. We show that ABCA7 is highly expressed in microglia and when monocytes are differentiated into macrophages. We hypothesized that ABCA7 plays a protective role in the brain that is related to phagocytic clearance of amyloid-β. We isolated microglia and macrophages from Abca7-/- and wild type mice and tested them for their capacity to phagocytose amyloid-β oligomers. We found that the phagocytic clearance of amyloid-β was substantially reduced in both microglia and macrophages from Abca7-/- mice compared to wild type mice. Consistent with these results, in vivo phagocytic clearance of amyloid-β oligomers in the hippocampus was reduced in Abca7-/- mice. Furthermore, ABCA7 transcription was upregulated in AD brains and in amyloidogenic mouse brains specifically in the hippocampus as a response to the amyloid-β pathogenic state. Together these results indicate that ABCA7 mediates phagocytic clearance of amyloid-β in the brain, and reveal a mechanism by which loss of function of ABCA7 increases the susceptibility to AD.
Collapse
Affiliation(s)
- YuHong Fu
- Neuroscience Research Australia, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Jen-Hsiang T Hsiao
- Neuroscience Research Australia, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - George Paxinos
- Neuroscience Research Australia, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Glenda M Halliday
- Neuroscience Research Australia, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Woojin Scott Kim
- Neuroscience Research Australia, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
20
|
Lee JM, Tan V, Lovejoy D, Braidy N, Rowe DB, Brew BJ, Guillemin GJ. Involvement of quinolinic acid in the neuropathogenesis of amyotrophic lateral sclerosis. Neuropharmacology 2017; 112:346-364. [DOI: 10.1016/j.neuropharm.2016.05.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 05/13/2016] [Accepted: 05/17/2016] [Indexed: 10/21/2022]
|
21
|
Lovelace MD, Varney B, Sundaram G, Lennon MJ, Lim CK, Jacobs K, Guillemin GJ, Brew BJ. Recent evidence for an expanded role of the kynurenine pathway of tryptophan metabolism in neurological diseases. Neuropharmacology 2017; 112:373-388. [DOI: 10.1016/j.neuropharm.2016.03.024] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 03/10/2016] [Accepted: 03/12/2016] [Indexed: 12/13/2022]
|
22
|
Lovelace MD, Varney B, Sundaram G, Franco NF, Ng ML, Pai S, Lim CK, Guillemin GJ, Brew BJ. Current Evidence for a Role of the Kynurenine Pathway of Tryptophan Metabolism in Multiple Sclerosis. Front Immunol 2016; 7:246. [PMID: 27540379 PMCID: PMC4972824 DOI: 10.3389/fimmu.2016.00246] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 06/10/2016] [Indexed: 12/13/2022] Open
Abstract
The kynurenine pathway (KP) is the major metabolic pathway of the essential amino acid tryptophan (TRP). Stimulation by inflammatory molecules, such as interferon-γ (IFN-γ), is the trigger for induction of the KP, driving a complex cascade of production of both neuroprotective and neurotoxic metabolites, and in turn, regulation of the immune response and responses of brain cells to the KP metabolites. Consequently, substantial evidence has accumulated over the past couple of decades that dysregulation of the KP and the production of neurotoxic metabolites are associated with many neuroinflammatory and neurodegenerative diseases, including Parkinson’s disease, AIDS-related dementia, motor neurone disease, schizophrenia, Huntington’s disease, and brain cancers. In the past decade, evidence of the link between the KP and multiple sclerosis (MS) has rapidly grown and has implicated the KP in MS pathogenesis. KP enzymes, indoleamine 2,3-dioxygenase (IDO-1) and tryptophan dioxygenase (highest expression in hepatic cells), are the principal enzymes triggering activation of the KP to produce kynurenine from TRP. This is in preference to other routes such as serotonin and melatonin production. In neurological disease, degradation of the blood–brain barrier, even if transient, allows the entry of blood monocytes into the brain parenchyma. Similar to microglia and macrophages, these cells are highly responsive to IFN-γ, which upregulates the expression of enzymes, including IDO-1, producing neurotoxic KP metabolites such as quinolinic acid. These metabolites circulate systemically or are released locally in the brain and can contribute to the excitotoxic death of oligodendrocytes and neurons in neurological disease principally by virtue of their agonist activity at N-methyl-d-aspartic acid receptors. The latest evidence is presented and discussed. The enzymes that control the checkpoints in the KP represent an attractive therapeutic target, and consequently several KP inhibitors are currently in clinical trials for other neurological diseases, and hence may make suitable candidates for MS patients. Underpinning these drug discovery endeavors, in recent years, several advances have been made in how KP metabolites are assayed in various biological fluids, and tremendous advancements have been made in how specimens are imaged to determine disease progression and involvement of various cell types and molecules in MS.
Collapse
Affiliation(s)
- Michael D Lovelace
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Bianca Varney
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research , Sydney, NSW , Australia
| | - Gayathri Sundaram
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research , Sydney, NSW , Australia
| | - Nunzio F Franco
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research , Sydney, NSW , Australia
| | - Mei Li Ng
- Faculty of Medicine, Sydney Medical School, University of Sydney , Sydney, NSW , Australia
| | - Saparna Pai
- Sydney Medical School, University of Sydney , Sydney, NSW , Australia
| | - Chai K Lim
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University , Sydney, NSW , Australia
| | - Gilles J Guillemin
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University , Sydney, NSW , Australia
| | - Bruce J Brew
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia; Department of Neurology, St Vincent's Hospital, Sydney, NSW, Australia
| |
Collapse
|
23
|
Schwarcz R. Kynurenines and Glutamate: Multiple Links and Therapeutic Implications. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 76:13-37. [PMID: 27288072 PMCID: PMC5803753 DOI: 10.1016/bs.apha.2016.01.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glutamate is firmly established as the major excitatory neurotransmitter in the mammalian brain and is actively involved in most aspects of neurophysiology. Moreover, glutamatergic impairments are associated with a wide variety of dysfunctional states, and both hypo- and hyperfunction of glutamate have been plausibly linked to the pathophysiology of neurological and psychiatric diseases. Metabolites of the kynurenine pathway (KP), the major catabolic route of the essential amino acid tryptophan, influence glutamatergic activity in several distinct ways. This includes direct effects of these "kynurenines" on ionotropic and metabotropic glutamate receptors or vesicular glutamate transport, and indirect effects, which are initiated by actions at various other recognition sites. In addition, some KP metabolites affect glutamatergic functions by generating or scavenging highly reactive free radicals. This review summarizes these phenomena and discusses implications for brain physiology and pathology.
Collapse
Affiliation(s)
- R Schwarcz
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
24
|
The Janus-faced nature of IDO1 in infectious diseases: challenges and therapeutic opportunities. Future Med Chem 2015; 8:39-54. [PMID: 26692277 DOI: 10.4155/fmc.15.165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Inhibition of IDO1 is a strategy pursued to develop novel therapeutic treatments for cancer. Recent years have witnessed growing evidence that the enzyme plays a pivotal role in viral, bacterial and fungal infections. These studies have underscored the Janus-faced nature of IDO1 in the regulation of host-pathogen interactions and commensalism. Starting with an outlook on the advances in the structural features of IDO1, herein we report recent findings that pinpoint the involvement of IDO1 in infectious diseases. Then, we present an overview of IDO1 inhibitors that have been enrolled in clinical trials as well as other distinct modulators of the enzyme that may enable further investigations of IDO1 and its role in infectious disease.
Collapse
|
25
|
O'Farrell K, Harkin A. Stress-related regulation of the kynurenine pathway: Relevance to neuropsychiatric and degenerative disorders. Neuropharmacology 2015; 112:307-323. [PMID: 26690895 DOI: 10.1016/j.neuropharm.2015.12.004] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/02/2015] [Accepted: 12/08/2015] [Indexed: 02/08/2023]
Abstract
The kynurenine pathway (KP), which is activated in times of stress and infection has been implicated in the pathophysiology of neurodegenerative and psychiatric disorders. Activation of this tryptophan metabolising pathway results in the production of neuroactive metabolites which have the potential to interfere with normal neuronal functioning which may contribute to altered neuronal transmission and the emergence of symptoms of these brain disorders. This review investigates the involvement of the KP in a range of neurological disorders, examining recent in vitro, in vivo and clinical discoveries highlights evidence to indicate that the KP is a potential therapeutic target in both neurodegenerative and stress-related neuropsychiatric disorders. Furthermore, this review identifies gaps in our knowledge with regard to this field which are yet to be examined to lead to a more comprehensive understanding of the role of KP activation in brain health and disease. This article is part of the Special Issue entitled 'The Kynurenine Pathway in Health and Disease'.
Collapse
Affiliation(s)
- Katherine O'Farrell
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences & Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland
| | - Andrew Harkin
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences & Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland; Neuroimmunology Research Group, Department of Physiology, School of Medicine & Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland.
| |
Collapse
|
26
|
Kynurenines and Multiple Sclerosis: The Dialogue between the Immune System and the Central Nervous System. Int J Mol Sci 2015; 16:18270-82. [PMID: 26287161 PMCID: PMC4581244 DOI: 10.3390/ijms160818270] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 07/22/2015] [Accepted: 07/23/2015] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis is an inflammatory disease of the central nervous system, in which axonal transection takes place in parallel with acute inflammation to various, individual extents. The importance of the kynurenine pathway in the physiological functions and pathological processes of the nervous system has been extensively investigated, but it has additionally been implicated as having a regulatory function in the immune system. Alterations in the kynurenine pathway have been described in both preclinical and clinical investigations of multiple sclerosis. These observations led to the identification of potential therapeutic targets in multiple sclerosis, such as synthetic tryptophan analogs, endogenous tryptophan metabolites (e.g., cinnabarinic acid), structural analogs (laquinimod, teriflunomid, leflunomid and tranilast), indoleamine-2,3-dioxygenase inhibitors (1MT and berberine) and kynurenine-3-monooxygenase inhibitors (nicotinylalanine and Ro 61-8048). The kynurenine pathway is a promising novel target via which to influence the immune system and to achieve neuroprotection, and further research is therefore needed with the aim of developing novel drugs for the treatment of multiple sclerosis and other autoimmune diseases.
Collapse
|
27
|
Jones SP, Franco NF, Varney B, Sundaram G, Brown DA, de Bie J, Lim CK, Guillemin GJ, Brew BJ. Expression of the Kynurenine Pathway in Human Peripheral Blood Mononuclear Cells: Implications for Inflammatory and Neurodegenerative Disease. PLoS One 2015; 10:e0131389. [PMID: 26114426 PMCID: PMC4482723 DOI: 10.1371/journal.pone.0131389] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 06/02/2015] [Indexed: 12/14/2022] Open
Abstract
The kynurenine pathway is a fundamental mechanism of immunosuppression and peripheral tolerance. It is increasingly recognized as playing a major role in the pathogenesis of a wide variety of inflammatory, neurodegenerative and malignant disorders. However, the temporal dynamics of kynurenine pathway activation and metabolite production in human immune cells is currently unknown. Here we report the novel use of flow cytometry, combined with ultra high-performance liquid chromatography and gas chromatography-mass spectrometry, to sensitively quantify the intracellular expression of three key kynurenine pathway enzymes and the main kynurenine pathway metabolites in a time-course study. This is the first study to show that up-regulation of indoleamine 2,3-dioxygenase (IDO-1), kynurenine 3-monoxygenase (KMO) and quinolinate phosphoribosyltransferase (QPRT) is lacking in lymphocytes treated with interferon gamma. In contrast, peripheral monocytes showed a significant elevation of kynurenine pathway enzymes and metabolites when treated with interferon gamma. Expression of IDO-1, KMO and QPRT correlated significantly with activation of the kynurenine pathway (kynurenine:tryptophan ratio), quinolinic acid concentration and production of the monocyte derived, pro-inflammatory immune response marker: neopterin. Our results also describe an original and sensitive methodological approach to quantify kynurenine pathway enzyme expression in cells. This has revealed further insights into the potential role of these enzymes in disease processes.
Collapse
Affiliation(s)
- Simon P. Jones
- Peter Duncan Neurosciences Research Unit, St Vincent’s Centre for Applied Medical Research, Sydney, Australia
- St Vincent’s Clinical School, Faculty of Medicine, UNSW, Sydney, Australia
- * E-mail:
| | - Nunzio F. Franco
- Peter Duncan Neurosciences Research Unit, St Vincent’s Centre for Applied Medical Research, Sydney, Australia
| | - Bianca Varney
- Peter Duncan Neurosciences Research Unit, St Vincent’s Centre for Applied Medical Research, Sydney, Australia
| | - Gayathri Sundaram
- Peter Duncan Neurosciences Research Unit, St Vincent’s Centre for Applied Medical Research, Sydney, Australia
| | - David A. Brown
- Peter Duncan Neurosciences Research Unit, St Vincent’s Centre for Applied Medical Research, Sydney, Australia
- St Vincent’s Clinical School, Faculty of Medicine, UNSW, Sydney, Australia
| | - Josien de Bie
- Neuroinflammation group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Chai K. Lim
- Neuroinflammation group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Gilles J. Guillemin
- Peter Duncan Neurosciences Research Unit, St Vincent’s Centre for Applied Medical Research, Sydney, Australia
- Neuroinflammation group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Bruce J. Brew
- Peter Duncan Neurosciences Research Unit, St Vincent’s Centre for Applied Medical Research, Sydney, Australia
- St Vincent’s Clinical School, Faculty of Medicine, UNSW, Sydney, Australia
- Department of Neurology, St Vincent’s Hospital, Sydney, Australia
| |
Collapse
|
28
|
Yan EB, Frugier T, Lim CK, Heng B, Sundaram G, Tan M, Rosenfeld JV, Walker DW, Guillemin GJ, Morganti-Kossmann MC. Activation of the kynurenine pathway and increased production of the excitotoxin quinolinic acid following traumatic brain injury in humans. J Neuroinflammation 2015; 12:110. [PMID: 26025142 PMCID: PMC4457980 DOI: 10.1186/s12974-015-0328-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 05/20/2015] [Indexed: 12/14/2022] Open
Abstract
Abstract During inflammation, the kynurenine pathway (KP) metabolises the essential amino acid tryptophan (TRP) potentially contributing to excitotoxicity via the release of quinolinic acid (QUIN) and 3-hydroxykynurenine (3HK). Despite the importance of excitotoxicity in the development of secondary brain damage, investigations on the KP in TBI are scarce. In this study, we comprehensively characterised changes in KP activation by measuring numerous metabolites in cerebrospinal fluid (CSF) from TBI patients and assessing the expression of key KP enzymes in brain tissue from TBI victims. Acute QUIN levels were further correlated with outcome scores to explore its prognostic value in TBI recovery. Methods Twenty-eight patients with severe TBI (GCS ≤ 8, three patients had initial GCS = 9–10, but rapidly deteriorated to ≤8) were recruited. CSF was collected from admission to day 5 post-injury. TRP, kynurenine (KYN), kynurenic acid (KYNA), QUIN, anthranilic acid (AA) and 3-hydroxyanthranilic acid (3HAA) were measured in CSF. The Glasgow Outcome Scale Extended (GOSE) score was assessed at 6 months post-TBI. Post-mortem brains were obtained from the Australian Neurotrauma Tissue and Fluid Bank and used in qPCR for quantitating expression of KP enzymes (indoleamine 2,3-dioxygenase-1 (IDO1), kynurenase (KYNase), kynurenine amino transferase-II (KAT-II), kynurenine 3-monooxygenase (KMO), 3-hydroxyanthranilic acid oxygenase (3HAO) and quinolinic acid phosphoribosyl transferase (QPRTase) and IDO1 immunohistochemistry. Results In CSF, KYN, KYNA and QUIN were elevated whereas TRP, AA and 3HAA remained unchanged. The ratios of QUIN:KYN, QUIN:KYNA, KYNA:KYN and 3HAA:AA revealed that QUIN levels were significantly higher than KYN and KYNA, supporting increased neurotoxicity. Amplified IDO1 and KYNase mRNA expression was demonstrated on post-mortem brains, and enhanced IDO1 protein coincided with overt tissue damage. QUIN levels in CSF were significantly higher in patients with unfavourable outcome and inversely correlated with GOSE scores. Conclusion TBI induced a striking activation of the KP pathway with sustained increase of QUIN. The exceeding production of QUIN together with increased IDO1 activation and mRNA expression in brain-injured areas suggests that TBI selectively induces a robust stimulation of the neurotoxic branch of the KP pathway. QUIN’s detrimental roles are supported by its association to adverse outcome potentially becoming an early prognostic factor post-TBI.
Collapse
Affiliation(s)
- Edwin B Yan
- Department of Physiology, Monash University, Clayton, VIC, 3800, Australia.
| | - Tony Frugier
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia
| | - Chai K Lim
- Neuroinflammation group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Benjamin Heng
- Neuroinflammation group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Gayathri Sundaram
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, Australia
| | - May Tan
- Hospital Queen Elizabeth, Karung Berkunci No. 2029, 88586, Kota Kinabalu, Sabah, Malaysia
| | - Jeffrey V Rosenfeld
- Department of Neurosurgery, The Alfred Hospital, Melbourne, Australia.,Department of Surgery, Central Clinical School and Monash Institute of Medical Engineering, Monash University, Melbourne, Australia
| | - David W Walker
- The Ritchie Centre, Hudson Institute of Medical Research, Monash Medical Centre, Melbourne, Australia
| | - Gilles J Guillemin
- Neuroinflammation group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Maria Cristina Morganti-Kossmann
- Australian New Zealand Intensive Care Research Centre, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia.,Department of Child Health, Barrow Neurological Institute, University of Arizona, Phoenix, AZ, USA
| |
Collapse
|
29
|
Quinolinic acid/tryptophan ratios predict neurological disease in SIV-infected macaques and remain elevated in the brain under cART. J Neurovirol 2015; 21:449-63. [PMID: 25776527 DOI: 10.1007/s13365-015-0334-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/16/2015] [Accepted: 02/24/2015] [Indexed: 01/28/2023]
Abstract
Activation of the kynurenine pathway (KP) of tryptophan catabolism likely contributes to HIV-associated neurological disorders. However, KP activation in brain tissue during HIV infection has been understudied, and the effect of combination antiretroviral therapy (cART) on KP induction in the brain is unknown. To examine these questions, tryptophan, kynurenine, 3-hydroxykynurenine, quinolinic acid, and serotonin levels were measured longitudinally during SIV infection in the striatum and CSF from untreated and cART-treated pigtailed macaques. Messenger RNA (mRNA) levels of KP enzymes also were measured in the striatum. In untreated macaques, elevations in KP metabolites coincided with transcriptional induction of upstream enzymes in the KP. Striatal KP induction was also temporally associated-but did not directly correlate-with serotonin losses in the brain. CSF quinolinic acid/tryptophan ratios were found to be the earliest predictor of neurological disease in untreated SIV-infected macaques, outperforming other KP metabolites as well as the putative biomarkers interleukin-6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1). Finally, cART did not restore KP metabolites to control levels in the striatum despite the control of the virus, though CSF metabolite levels were normalized in most animals. Overall, these results demonstrate that cerebral KP activation is only partially resolved with cART and that CSF QUIN/TRP ratios are an early, predictive biomarker of CNS disease.
Collapse
|
30
|
Huo L, Davis I, Liu F, Andi B, Esaki S, Iwaki H, Hasegawa Y, Orville AM, Liu A. Crystallographic and spectroscopic snapshots reveal a dehydrogenase in action. Nat Commun 2015; 6:5935. [PMID: 25565451 PMCID: PMC4286809 DOI: 10.1038/ncomms6935] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/21/2014] [Indexed: 11/25/2022] Open
Abstract
Aldehydes are ubiquitous intermediates in metabolic pathways and their innate reactivity can often make them quite unstable. There are several aldehydic intermediates in the metabolic pathway for tryptophan degradation that can decay into neuroactive compounds that have been associated with numerous neurological diseases. An enzyme of this pathway, 2-aminomuconate-6-semialdehyde dehydrogenase, is responsible for ‘disarming’ the final aldehydic intermediate. Here we show the crystal structures of a bacterial analogue enzyme in five catalytically relevant forms: resting state, one binary and two ternary complexes, and a covalent, thioacyl intermediate. We also report the crystal structures of a tetrahedral, thiohemiacetal intermediate, a thioacyl intermediate and an NAD+-bound complex from an active site mutant. These covalent intermediates are characterized by single-crystal and solution-state electronic absorption spectroscopy. The crystal structures reveal that the substrate undergoes an E/Z isomerization at the enzyme active site before an sp3-to-sp2 transition during enzyme-mediated oxidation. Aldehydes are common intermediates in enzymatic pathways, but their high reactivity can make them difficult to observe. Here, the authors study the mechanism of aldehyde deactivation in a dehydrogenase, showing a key E/Z isomerization and observing a thiohemiacetal intermediate by crystal structure analysis.
Collapse
Affiliation(s)
- Lu Huo
- 1] Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, USA [2] Molecular Basis of Disease Area of Focus Program, Georgia State University, Atlanta, Georgia 30303, USA
| | - Ian Davis
- 1] Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, USA [2] Molecular Basis of Disease Area of Focus Program, Georgia State University, Atlanta, Georgia 30303, USA
| | - Fange Liu
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, USA
| | - Babak Andi
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, New York 11973, USA
| | - Shingo Esaki
- 1] Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, USA [2] Molecular Basis of Disease Area of Focus Program, Georgia State University, Atlanta, Georgia 30303, USA
| | - Hiroaki Iwaki
- Department of Life Science and Biotechnology and ORDIST, Kansai University, Suita, Osaka 564-8680, Japan
| | - Yoshie Hasegawa
- Department of Life Science and Biotechnology and ORDIST, Kansai University, Suita, Osaka 564-8680, Japan
| | - Allen M Orville
- 1] Photon Sciences Directorate, Brookhaven National Laboratory, Upton, New York 11973, USA [2] Biosciences Department, Brookhaven National Laboratory, Upton, New York 11973, USA
| | - Aimin Liu
- 1] Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, USA [2] Molecular Basis of Disease Area of Focus Program, Georgia State University, Atlanta, Georgia 30303, USA
| |
Collapse
|
31
|
Bay-Richter C, Linderholm KR, Lim CK, Samuelsson M, Träskman-Bendz L, Guillemin GJ, Erhardt S, Brundin L. A role for inflammatory metabolites as modulators of the glutamate N-methyl-D-aspartate receptor in depression and suicidality. Brain Behav Immun 2015; 43:110-7. [PMID: 25124710 DOI: 10.1016/j.bbi.2014.07.012] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 07/16/2014] [Accepted: 07/25/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Patients with depression and suicidality suffer from low-grade neuroinflammation. Pro-inflammatory cytokines activate indoleamine 2,3-dioxygenase, an initial enzyme of the kynurenine pathway. This pathway produces neuroactive metabolites, including quinolinic- and kynurenic acid, binding to the glutamate N-methyl-d-aspartate-receptor, which is hypothesized to be part of the neural mechanisms underlying symptoms of depression. We therefore hypothesized that symptoms of depression and suicidality would fluctuate over time in patients prone to suicidal behavior, depending on the degree of inflammation and kynurenine metabolite levels in the cerebrospinal fluid (CSF). METHODS We measured cytokines and kynurenine metabolites in CSF, collected from suicide attempters at repeated occasions over 2 years (total patient samples n=143, individuals n=30) and healthy controls (n=36). The association between the markers and psychiatric symptoms was assessed using the Montgomery Asberg Depression Rating Scale and the Suicide Assessment Scale. RESULTS Quinolinic acid was increased and kynurenic acid decreased over time in suicidal patients versus healthy controls. Furthermore, we found a significant association between low kynurenic acid and severe depressive symptoms, as well as between high interleukin-6 levels and more severe suicidal symptoms. CONCLUSIONS We demonstrate a long-term dysregulation of the kynurenine pathway in the central nervous system of suicide attempters. An increased load of inflammatory cytokines was coupled to more severe symptoms. We therefore suggest that patients with a dysregulated kynurenine pathway are vulnerable to develop depressive symptoms upon inflammatory conditions, as a result the excess production of the NMDA-receptor agonist quinolinic acid. This study provides a neurobiological framework supporting the use of NMDA-receptor antagonists in the treatment of suicidality and depression.
Collapse
Affiliation(s)
- Cecilie Bay-Richter
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark.
| | - Klas R Linderholm
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Chai K Lim
- Neuroinflammation Group, Australian School of Advanced Medicine, Macquarie University, NSW, Australia
| | - Martin Samuelsson
- Faculty of Health Sciences, Department of Clinical and Experimental Medicine, Division of Psychiatry, Linköping University, Linköping, Sweden
| | - Lil Träskman-Bendz
- Department of Clinical Sciences, Section of Psychiatry, Lund University, Lund, Sweden
| | - Gilles J Guillemin
- Neuroinflammation Group, Australian School of Advanced Medicine, Macquarie University, NSW, Australia
| | - Sophie Erhardt
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Lena Brundin
- Division of Psychiatry and Behavioral Medicine, Michigan State University, Grand Rapids, MI, USA; Laboratory of Behavioral Medicine, Van Andel Research Institute, Grand Rapids, MI, USA
| |
Collapse
|
32
|
Sundaram G, Brew BJ, Jones SP, Adams S, Lim CK, Guillemin GJ. Quinolinic acid toxicity on oligodendroglial cells: relevance for multiple sclerosis and therapeutic strategies. J Neuroinflammation 2014; 11:204. [PMID: 25498310 PMCID: PMC4302518 DOI: 10.1186/s12974-014-0204-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 11/17/2014] [Indexed: 11/10/2022] Open
Abstract
The excitotoxin quinolinic acid, a by-product of the kynurenine pathway, is known to be involved in several neurological diseases including multiple sclerosis (MS). Quinolinic acid levels are elevated in experimental autoimmune encephalomyelitis rodents, the widely used animal model of MS. Our group has also found pathophysiological concentrations of quinolinic acid in MS patients. This led us to investigate the effect of quinolinic acid on oligodendrocytes; the main cell type targeted by the autoimmune response in MS. We have examined the kynurenine pathway (KP) profile of two oligodendrocyte cell lines and show that these cells have a limited threshold to catabolize exogenous quinolinic acid. We further propose and demonstrate two strategies to limit quinolinic acid gliotoxicity: 1) by neutralizing quinolinic acid’s effects with anti-quinolinic acid monoclonal antibodies and 2) directly inhibiting quinolinic acid production from activated monocytic cells using specific KP enzyme inhibitors. The outcome of this study provides a new insight into therapeutic strategies for limiting quinolinic acid-induced neurodegeneration, especially in neurological disorders that target oligodendrocytes, such as MS.
Collapse
Affiliation(s)
- Gayathri Sundaram
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, Australia. .,School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia.
| | - Bruce J Brew
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, Australia. .,Department of Neurology, St Vincent's Hospital, Sydney, Australia.
| | - Simon P Jones
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, Australia.
| | - Seray Adams
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia. .,Neurodegenerative diseases Research Group, Australian School of Advanced Medicine, Faculty of Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia.
| | - Chai K Lim
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, Australia. .,School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia. .,Neurodegenerative diseases Research Group, Australian School of Advanced Medicine, Faculty of Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia.
| | - Gilles J Guillemin
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, Australia. .,School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia. .,Neurodegenerative diseases Research Group, Australian School of Advanced Medicine, Faculty of Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
33
|
Pierozan P, Ferreira F, de Lima BO, Pessoa-Pureur R. Quinolinic acid induces disrupts cytoskeletal homeostasis in striatal neurons. Protective role of astrocyte-neuron interaction. J Neurosci Res 2014; 93:268-84. [DOI: 10.1002/jnr.23494] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 08/29/2014] [Accepted: 09/14/2014] [Indexed: 12/18/2022]
Affiliation(s)
- Paula Pierozan
- Departamento de Bioquímica; Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul; Porto Alegre Brazil
| | - Fernanda Ferreira
- Departamento de Bioquímica; Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul; Porto Alegre Brazil
| | - Bárbara Ortiz de Lima
- Departamento de Bioquímica; Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul; Porto Alegre Brazil
| | - Regina Pessoa-Pureur
- Departamento de Bioquímica; Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul; Porto Alegre Brazil
| |
Collapse
|
34
|
Braidy N, Brew BJ, Inestrosa NC, Chung R, Sachdev P, Guillemin GJ. Changes in Cathepsin D and Beclin-1 mRNA and protein expression by the excitotoxin quinolinic acid in human astrocytes and neurons. Metab Brain Dis 2014; 29:873-83. [PMID: 24833554 DOI: 10.1007/s11011-014-9557-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 04/28/2014] [Indexed: 01/18/2023]
Abstract
Quinolinic acid (QUIN) is an excitotoxin that has been implicated in the pathogenesis of several neurodegenerative diseases including Alzheimer's disease (AD). While QUIN has been shown to induce neuronal and astrocytic apoptosis as well as excitotoxic cell death, other mechanisms such as autophagy remain unexplored. We investigated the role of Cathepsin D (CatD) and Beclin-1 (Bc1) in QUIN-treated primary human astrocytes and neurons. We demonstrated that the expression patterns of CatD, a lysosomal aspartic protease associated with autophagy, are increased at 24 h after QUIN treatment. However, unlike CatD, the expression patterns of Bc1, a tumour suppressor protein, are significantly reduced at 24 h after QUIN treatment in both brain cell types. Furthermore, we showed that the NMDA ion channel blockers, MK801, can attenuate QUIN-induced changes CatD and Bc1 expression in both astrocytes and neurons. Taken together, these results suggest that induction of deficits in CatD and Bc1 is a significant mechanism for QUIN toxicity in glial and neuronal cells. Maintenance of autophagy may play a crucial role in neuroprotection in the setting of AD.
Collapse
Affiliation(s)
- Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| | | | | | | | | | | |
Collapse
|
35
|
Rao VR, Ruiz AP, Prasad VR. Viral and cellular factors underlying neuropathogenesis in HIV associated neurocognitive disorders (HAND). AIDS Res Ther 2014; 11:13. [PMID: 24894206 PMCID: PMC4043700 DOI: 10.1186/1742-6405-11-13] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 05/08/2014] [Indexed: 11/11/2022] Open
Abstract
As the HIV-1 epidemic enters its fourth decade, HIV-1 associated neurological disorders (HAND) continue to be a major concern in the infected population, despite the widespread use of anti-retroviral therapy. Advancing age and increased life expectancy of the HIV-1 infected population have been shown to increase the risk of cognitive dysfunction. Over the past 10 years, there has been a significant progress in our understanding of the mechanisms and the risk factors involved in the development of HAND. Key events that lead up to neuronal damage in HIV-1 infected individuals can be categorized based on the interaction of HIV-1 with the various cell types, including but not limited to macrophages, brain endothelial cells, microglia, astrocytes and the neurons. This review attempts to decipher these interactions, beginning with HIV-1 infection of macrophages and ultimately resulting in the release of neurotoxic viral and host products. These include: interaction with endothelial cells, resulting in the impairment of the blood brain barrier; interaction with the astrocytes, leading to metabolic and neurotransmitter imbalance; interactions with resident immune cells in the brain, leading to release of toxic cytokines and chemokines. We also review the mechanisms underlying neuronal damage caused by the factors mentioned above. We have attempted to bring together recent findings in these areas to help appreciate the viral and host factors that bring about neurological dysfunction. In addition, we review host factors and viral genotypic differences that affect phenotypic pathological outcomes, as well as recent advances in treatment options to specifically address the neurotoxic mechanisms in play.
Collapse
|
36
|
Barichello T, Generoso JS, Simões LR, Elias SG, Tashiro MH, Dominguini D, Comim CM, Vilela MC, Teixeira AL, Quevedo J. Inhibition of indoleamine 2,3-dioxygenase prevented cognitive impairment in adult Wistar rats subjected to pneumococcal meningitis. Transl Res 2013; 162:390-7. [PMID: 23994082 DOI: 10.1016/j.trsl.2013.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 07/30/2013] [Accepted: 08/05/2013] [Indexed: 10/26/2022]
Abstract
Streptococcus pneumoniae is a common cause of forms of bacterial meningitis that have a high mortality rate and cause long-term neurologic sequelae. We evaluated the effects of an indoleamine 2,3-dioxygenase (IDO) inhibitor on proinflammatory mediators and memory in Wistar rats subjected to pneumococcal meningitis. The animals were divided into 4 groups: sham, sham treated with IDO inhibitor, meningitis, and meningitis treated with IDO inhibitor. During the first experiment, the animals were killed 24 hours later, and the hippocampus was isolated for the analysis of tumor necrosis factor (TNF)-α, interleukin (IL)-4, IL-6, IL-10, and cytokine-induced neutrophil chemoattractant 1 (CINC-1) levels. The survival rate was 56.296% in the meningitis group and 29.616% in the meningitis group with IDO inhibitor. In the control group, we found a mean of 14.29 white blood cells/mL cerebrospinal fluid, whereas the mean was 80.00 white blood cells/mL cerebrospinal fluid in the sham IDO inhibitor group, 1167.00 white blood cells/mL cerebrospinal fluid in the meningitis group, and 286.70 white blood cells/mL cerebrospinal fluid in the meningitis IDO inhibitor group. In the meningitis group with IDO inhibitor, the levels of TNF-α and CINC-1 were reduced. In the second experiment, animals were subjected to a behavioral task and cytokine analysis 10 days after meningitis induction. In the meningitis group, there was an impairment of aversive memory. However, in the meningitis group that received adjuvant treatment with the IDO inhibitor, animals demonstrated preservation of aversive memory. These findings showed dual effects of the IDO inhibitor on a pneumococcal meningitis animal model because the inhibitor impaired survival but also produced beneficial effects, including anti-inflammatory activity and neuroprotection against the latter behavioral deficits.
Collapse
Affiliation(s)
- Tatiana Barichello
- Laboratório de Microbiologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil; Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil; Núcleo de Excelência em Neurociências Aplicadas de Santa Catarina (NENASC), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Quinolinic acid: an endogenous neurotoxin with multiple targets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:104024. [PMID: 24089628 PMCID: PMC3780648 DOI: 10.1155/2013/104024] [Citation(s) in RCA: 411] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 07/23/2013] [Accepted: 08/01/2013] [Indexed: 11/21/2022]
Abstract
Quinolinic acid (QUIN), a neuroactive metabolite of the kynurenine pathway, is normally presented in nanomolar concentrations in human brain and cerebrospinal fluid (CSF) and is often implicated in the pathogenesis of a variety of human neurological diseases. QUIN is an agonist of N-methyl-D-aspartate (NMDA) receptor, and it has a high in vivo potency as an excitotoxin. In fact, although QUIN has an uptake system, its neuronal degradation enzyme is rapidly saturated, and the rest of extracellular QUIN can continue stimulating the NMDA receptor. However, its toxicity cannot be fully explained by its activation of NMDA receptors it is likely that additional mechanisms may also be involved. In this review we describe some of the most relevant targets of QUIN neurotoxicity which involves presynaptic receptors, energetic dysfunction, oxidative stress, transcription factors, cytoskeletal disruption, behavior alterations, and cell death.
Collapse
|
38
|
Lim CK, Yap MMC, Kent SJ, Gras G, Samah B, Batten JC, De Rose R, Heng B, Brew BJ, Guillemin GJ. Characterization of the kynurenine pathway and quinolinic Acid production in macaque macrophages. Int J Tryptophan Res 2013; 6:7-19. [PMID: 23761975 PMCID: PMC3662399 DOI: 10.4137/ijtr.s11789] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The kynurenine pathway (KP) and one of its end-products, the excitotoxin quinolinic acid (QUIN), are involved in the pathogenesis of several major neuroinflammatory brain diseases. A relevant animal model to study KP metabolism is now needed to assess whether intervention in this pathway may improve the outcome of such diseases. Humans and macaques share a very similar genetic makeup. In this study, we characterized the KP metabolism in macaque primary macrophages of three different species in comparison to human cells. We found that the KP profiles in simian macrophages were very similar to those in humans when challenged with inflammatory cytokines. Further, we found that macaque macrophages are capable of producing a pathophysiological concentration of QUIN. Our data validate the simian model as a relevant model to study the human cellular KP metabolism in the context of inflammation.
Collapse
Affiliation(s)
- Chai K Lim
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, Australia. ; MND and Neurodegenerative disease Research Group, Australian School of Advance Medicine, Macquarie University, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Schwarcz R, Bruno JP, Muchowski PJ, Wu HQ. Kynurenines in the mammalian brain: when physiology meets pathology. Nat Rev Neurosci 2012; 13:465-77. [PMID: 22678511 DOI: 10.1038/nrn3257] [Citation(s) in RCA: 1033] [Impact Index Per Article: 86.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The essential amino acid tryptophan is not only a precursor of serotonin but is also degraded to several other neuroactive compounds, including kynurenic acid, 3-hydroxykynurenine and quinolinic acid. The synthesis of these metabolites is regulated by an enzymatic cascade, known as the kynurenine pathway, that is tightly controlled by the immune system. Dysregulation of this pathway, resulting in hyper-or hypofunction of active metabolites, is associated with neurodegenerative and other neurological disorders, as well as with psychiatric diseases such as depression and schizophrenia. With recently developed pharmacological agents, it is now possible to restore metabolic equilibrium and envisage novel therapeutic interventions.
Collapse
Affiliation(s)
- Robert Schwarcz
- University of Maryland School of Medicine, Baltimore, Maryland 21228, USA. rschwarc@mprc. umaryland.edu
| | | | | | | |
Collapse
|
40
|
Kandanearatchi A, Brew BJ. The kynurenine pathway and quinolinic acid: pivotal roles in HIV associated neurocognitive disorders. FEBS J 2012; 279:1366-74. [PMID: 22260426 DOI: 10.1111/j.1742-4658.2012.08500.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This brief review will first consider HIV associated neurocognitive disorder followed by the current understanding of its neuropathogenesis. Against this background the role of the kynurenine pathway will be detailed. Evidence both direct and indirect will be discussed for involvement of the kynurenine pathway at each step in the neuropathogenesis of HIV associated neurocognitive disorder.
Collapse
Affiliation(s)
- Apsara Kandanearatchi
- St Vincent's Centre for Applied Medical Research, Sydney, University of New South Wales, Sydney, Australia.
| | | |
Collapse
|
41
|
Maes M, Leonard BE, Myint AM, Kubera M, Verkerk R. The new '5-HT' hypothesis of depression: cell-mediated immune activation induces indoleamine 2,3-dioxygenase, which leads to lower plasma tryptophan and an increased synthesis of detrimental tryptophan catabolites (TRYCATs), both of which contribute to the onset of depression. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:702-21. [PMID: 21185346 DOI: 10.1016/j.pnpbp.2010.12.017] [Citation(s) in RCA: 475] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 12/07/2010] [Accepted: 12/16/2010] [Indexed: 02/07/2023]
Abstract
This paper reviews the body of evidence that not only tryptophan and consequent 5-HT depletion, but also induction of indoleamine 2,3-dioxygenase (IDO) and the detrimental effects of tryptophan catabolites (TRYCATs) play a role in the pathophysiology of depression. IDO is induced by interferon (IFN)γ, interleukin-6 and tumor necrosis factor-α, lipopolysaccharides and oxidative stress, factors that play a role in the pathophysiology of depression. TRYCATs, like kynurenine and quinolinic acid, are depressogenic and anxiogenic; activate oxidative pathways; cause mitochondrial dysfunctions; and have neuroexcitatory and neurotoxic effects that may lead to neurodegeneration. The TRYCAT pathway is also activated following induction of tryptophan 2,3-dioxygenase (TDO) by glucocorticoids, which are elevated in depression. There is evidence that activation of IDO reduces plasma tryptophan and increases TRYCAT synthesis in depressive states and that TDO activation may play a role as well. The development of depressive symptoms during IFNα-based immunotherapy is strongly associated with IDO activation, increased production of detrimental TRYCATs and lowered levels of tryptophan. Women show greater IDO activation and TRYCAT production following immune challenge than men. In the early puerperium, IDO activation and TRYCAT production are associated with the development of affective symptoms. Clinical depression is accompanied by lowered levels of neuroprotective TRYCATs or increased levels or neurotoxic TRYCATs, and lowered plasma tryptophan, which is associated with indices of immune activation and glucocorticoid hypersecretion. Lowered tryptophan and increased TRYCATs induce T cell unresponsiveness and therefore may exert a negative feedback on the primary inflammatory response in depression. It is concluded that activation of the TRYCAT pathway by IDO and TDO may be associated with the development of depressive symptoms through tryptophan depletion and the detrimental effects of TRYCATs. Therefore, the TRYCAT pathway should be a new drug target in depression. Direct inhibitors of IDO are less likely to be useful drugs than agents, such as kynurenine hydroxylase inhibitors; drugs which block the primary immune response; compounds that increase the protective effects of kynurenic acid; and specific antioxidants that target IDO activation, the immune and oxidative pathways, and 5-HT as well.
Collapse
Affiliation(s)
- M Maes
- Maes Clinics @ TRIA, Piyavate Hospital, 998 Rimklongsamsen Road, Bangkok 10310, Thailand.
| | | | | | | | | |
Collapse
|
42
|
Lee MC, Ting KK, Adams S, Brew BJ, Chung R, Guillemin GJ. Characterisation of the expression of NMDA receptors in human astrocytes. PLoS One 2010; 5:e14123. [PMID: 21152063 PMCID: PMC2994931 DOI: 10.1371/journal.pone.0014123] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 10/26/2010] [Indexed: 12/14/2022] Open
Abstract
Astrocytes have long been perceived only as structural and supporting cells within the central nervous system (CNS). However, the discovery that these glial cells may potentially express receptors capable of responding to endogenous neurotransmitters has resulted in the need to reassess astrocytic physiology. The aim of the current study was to characterise the expression of NMDA receptors (NMDARs) in primary human astrocytes, and investigate their response to physiological and excitotoxic concentrations of the known endogenous NMDAR agonists, glutamate and quinolinic acid (QUIN). Primary cultures of human astrocytes were used to examine expression of these receptors at the mRNA level using RT-PCR and qPCR, and at the protein level using immunocytochemistry. The functionality role of the receptors was assessed using intracellular calcium influx experiments and measuring extracellular lactate dehydrogenase (LDH) activity in primary cultures of human astrocytes treated with glutamate and QUIN. We found that all seven currently known NMDAR subunits (NR1, NR2A, NR2B, NR2C, NR2D, NR3A and NR3B) are expressed in astrocytes, but at different levels. Calcium influx studies revealed that both glutamate and QUIN could activate astrocytic NMDARs, which stimulates Ca2+ influx into the cell and can result in dysfunction and death of astrocytes. Our data also show that the NMDAR ion channel blockers, MK801, and memantine can attenuate glutamate and QUIN mediated cell excitotoxicity. This suggests that the mechanism of glutamate and QUIN gliotoxicity is at least partially mediated by excessive stimulation of NMDARs. The present study is the first to provide definitive evidence for the existence of functional NMDAR expression in human primary astrocytes. This discovery has significant implications for redefining the cellular interaction between glia and neurons in both physiological processes and pathological conditions.
Collapse
Affiliation(s)
- Ming-Chak Lee
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, Australia
- St. Vincent's Centre for Applied Medical Research, St. Vincent's Hospital, Sydney, Australia
| | - Ka Ka Ting
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, Australia
- St. Vincent's Centre for Applied Medical Research, St. Vincent's Hospital, Sydney, Australia
| | - Seray Adams
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, Australia
- St. Vincent's Centre for Applied Medical Research, St. Vincent's Hospital, Sydney, Australia
| | - Bruce J. Brew
- St. Vincent's Centre for Applied Medical Research, St. Vincent's Hospital, Sydney, Australia
- Departments of Neurology and HIV Medicine, St. Vincent's Hospital, Sydney, Australia
| | - Roger Chung
- NeuroRepair Group, Menzies Research Institute, University of Tasmania, Hobart, Tasmania, Australia
| | - Gilles J. Guillemin
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, Australia
- St. Vincent's Centre for Applied Medical Research, St. Vincent's Hospital, Sydney, Australia
- * E-mail:
| |
Collapse
|
43
|
Lim CK, Brew BJ, Sundaram G, Guillemin GJ. Understanding the roles of the kynurenine pathway in multiple sclerosis progression. Int J Tryptophan Res 2010; 3:157-67. [PMID: 22084596 PMCID: PMC3195238 DOI: 10.4137/ijtr.s4294] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The kynurenine pathway (KP) is a major degradative pathway of tryptophan ultimately leading to the production of nicotinamide adenine dinucleotide (NAD+) and is also one of the major regulatory mechanisms of the immune response. The KP is known to be involved in several neuroinflammatory disorders including Alzheimer’s disease, amyotrophic lateral sclerosis, AIDS dementia complex, Parkinson’s disease, schizophrenia, Huntington’s disease and brain tumours. However, the KP remains a relatively new topic for the field of multiple sclerosis (MS). Over the last 2–3 years, some evidence has progressively emerged suggesting that the KP is likely to be involved in the pathogenesis of autoimmune diseases especially MS. Some KP modulators are already in clinical trials for other inflammatory diseases and would potentially provide a new and important therapeutic strategy for MS patients. This review summarizes the known relationships between the KP and MS.
Collapse
Affiliation(s)
- Chai K Lim
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney 2052, Australia
| | | | | | | |
Collapse
|
44
|
Davies NW, Guillemin G, Brew BJ. Tryptophan, Neurodegeneration and HIV-Associated Neurocognitive Disorder. Int J Tryptophan Res 2010; 3:121-40. [PMID: 22084594 PMCID: PMC3195234 DOI: 10.4137/ijtr.s4321] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This review presents an up-to-date assessment of the role of the tryptophan metabolic and catabolic pathways in neurodegenerative disease and HIV-associated neurocognitive disorder. The kynurenine pathway and the effects of each of its enzymes and products are reviewed. The differential expression of the kynurenine pathway in cells within the brain, including inflammatory cells, is explored given the increasing recognition of the importance of inflammation in neurodegenerative disease. An overview of common mechanisms of neurodegeneration is presented before a review and discussion of the evidence for a pathogenetic role of the kynurenine pathway in Alzheimer's disease, HIV-associated neurocognitive disorder, Huntington's disease, motor neurone disease, and Parkinson's disease.
Collapse
Affiliation(s)
- Nicholas W.S. Davies
- Department of Neurology, and
- St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Darlinghurst, Sydney, Australia
| | - Gilles Guillemin
- St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Darlinghurst, Sydney, Australia
| | - Bruce J. Brew
- Department of Neurology, and
- St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Darlinghurst, Sydney, Australia
| |
Collapse
|
45
|
Pierozan P, Zamoner A, Soska AK, Silvestrin RB, Loureiro SO, Heimfarth L, Mello e Souza T, Wajner M, Pessoa-Pureur R. Acute intrastriatal administration of quinolinic acid provokes hyperphosphorylation of cytoskeletal intermediate filament proteins in astrocytes and neurons of rats. Exp Neurol 2010; 224:188-96. [PMID: 20303347 DOI: 10.1016/j.expneurol.2010.03.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Revised: 03/05/2010] [Accepted: 03/10/2010] [Indexed: 01/06/2023]
Abstract
In the present study we investigated the effect of in vivo intrastriatal injection of quinolinic acid (QA) on cytoskeletal proteins in astrocytes and neurons of young rats at early stage (30 min) after infusion. QA (150 nmoles/0.5 microL) significantly increased the in vitro phosphorylation of the low molecular weight neurofilament subunit (NFL) and the glial fibrillary acidic protein (GFAP) of neurons and astrocytes, respectively. This effect was mediated by cAMP-dependent protein kinase A (PKA), protein kinase C (PKC) and Ca(2+)/calmodulin-dependent protein kinase II (PKCaMII). In contrast, mitogen activated protein kinases were not activated by QA infusion. Furthermore, the specific N-methyl-D-aspartate (NMDA) antagonist MK-801 (0.25 mg/kg i.p), the antioxidant L-NAME (60 mg\kg\day), and diphenyldisselenide (PheSe)(2) (0.625 mg\kg\day) injected prior to QA infusion totally prevented QA-induced cytoskeletal hyperphosphorylation. We also observed that QA-induced hyperphosphorylation was targeted at the Ser55 phosphorylating site on NFL head domain, described as a regulatory site for NF assembly in vivo. This effect was fully prevented by MK801, by the PKA inhibitor H89 and by (PheSe)(2), whereas staurosporine (PKC inhibitor) only partially prevented Ser55 phosphorylation. The PKCaMII inhibitor (KN93) and the antioxidant L-NAME failed to prevent the hyperphosphorylation of Ser55 by QA infusion. Therefore, we presume that QA-elicited hyperphosphorylation of the neural cytoskeleton, and specially of NFLSer55, achieved by intrastriatal QA injection could represent an early step in the pathophysiological cascade of deleterious events exerted by QA in rat striatum. Our observations also indicate that NMDA-mediated Ca(2+) events and oxidative stress may be related to the altered protein cytoskeleton hyperphosphorylation observed with important implications for brain function.
Collapse
Affiliation(s)
- Paula Pierozan
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brasil
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Braidy N, Grant R, Adams S, Brew BJ, Guillemin GJ. Mechanism for Quinolinic Acid Cytotoxicity in Human Astrocytes and Neurons. Neurotox Res 2009; 16:77-86. [DOI: 10.1007/s12640-009-9051-z] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Revised: 03/31/2009] [Accepted: 04/02/2009] [Indexed: 12/28/2022]
|
47
|
Maes M, Yirmyia R, Noraberg J, Brene S, Hibbeln J, Perini G, Kubera M, Bob P, Lerer B, Maj M. The inflammatory & neurodegenerative (I&ND) hypothesis of depression: leads for future research and new drug developments in depression. Metab Brain Dis 2009; 24:27-53. [PMID: 19085093 DOI: 10.1007/s11011-008-9118-1] [Citation(s) in RCA: 646] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Accepted: 10/28/2008] [Indexed: 12/27/2022]
Abstract
Despite extensive research, the current theories on serotonergic dysfunctions and cortisol hypersecretion do not provide sufficient explanations for the nature of depression. Rational treatments aimed at causal factors of depression are not available yet. With the currently available antidepressant drugs, which mainly target serotonin, less than two thirds of depressed patients achieve remission. There is now evidence that inflammatory and neurodegenerative (I&ND) processes play an important role in depression and that enhanced neurodegeneration in depression may-at least partly-be caused by inflammatory processes. Multiple inflammatory-cytokines, oxygen radical damage, tryptophan catabolites-and neurodegenerative biomarkers have been established in patients with depression and these findings are corroborated by animal models of depression. A number of vulnerability factors may predispose towards depression by enhancing inflammatory reactions, e.g. lower peptidase activities (dipeptidyl-peptidase IV, DPP IV), lower omega-3 polyunsaturated levels and an increased gut permeability (leaky gut). The cytokine hypothesis considers that external, e.g. psychosocial stressors, and internal stressors, e.g. organic inflammatory disorders or conditions, such as the postpartum period, may trigger depression via inflammatory processes. Most if not all antidepressants have specific anti-inflammatory effects, while restoration of decreased neurogenesis, which may be induced by inflammatory processes, may be related to the therapeutic efficacy of antidepressant treatments. Future research to disentangle the complex etiology of depression calls for a powerful paradigm shift, i.e. by means of a high throughput-high quality screening, including functional genetics and genotyping microarrays; established and novel animal and ex vivo-in vitro models for depression, such as new transgenic mouse models and endophenotype-based animal models, specific cell lines, in vivo and ex vivo electroporation, and organotypic brain slice culture models. This screening will allow to: 1) discover new I&ND biomarkers, both at the level of gene expression and the phenotype; and elucidate the underlying molecular I&ND pathways causing depression; and 2) identify new therapeutic targets in the I&ND pathways; develop new anti-I&ND drugs for these targets; select existing anti-I&ND drugs or substances that could augment the efficacy of antidepressants; and predict therapeutic response by genetic I&ND profiles.
Collapse
Affiliation(s)
- Michael Maes
- Clinical Research Center for Mental Health, Olmenlaan 9, Antwerp Wilrijk 2610, Belgium.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Promotion of cellular NAD(+) anabolism: therapeutic potential for oxidative stress in ageing and Alzheimer's disease. Neurotox Res 2008; 13:173-84. [PMID: 18522897 DOI: 10.1007/bf03033501] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Oxidative imbalance is a prominent feature in Alzheimer's disease and ageing. Increased levels of reactive oxygen species (ROS) can result in disordered cellular metabolism due to lipid peroxdation, protein-cross linking, DNA damage and the depletion of nicotinamide adenine dinucleotide (NAD(+)). NAD(+) is a ubiquitous pyridine nucleotide that plays an essential role in important biological reactions., from ATP production and secondary messenger signaling, to transcriptional regulation and DNA repair. Chronic oxidative stress may be associated with NAD(+) depletion and a subsequent decrease in metabolic regulation and cell viability. Hence, therapies targeted toward maintaining intracellular NAD(+) pools may prove efficacious in the protection of age-dependent cellular damage, in general, and neurodegeneration in chronic central nervous system inflammatory diseases such as Alzheimer's disease, in particular.
Collapse
|
49
|
Gätjens J, Mullins CS, Kampf JW, Thuéry P, Pecoraro VL. Corroborative cobalt and zinc model compounds of alpha-amino-beta-carboxymuconic-epsilon-semialdehyde decarboxylase (ACMSD). Dalton Trans 2008:51-62. [PMID: 19081971 DOI: 10.1039/b809453e] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
We have synthesised and characterised a series of new Co(II) complexes (1-4, 6, 7) and one new Zn(II) complex (5) employing N(3)- and N(3)O-donor ligands [biap: N,N-bis(2-ethyl-5-methyl-imidazol-4-ylmethyl)amino-propane, KBPZG: potassium N,N-bis(3,5-dimethylpyrazolylmethyl) glycinate, KBPZA: potassium N,N-bis(3,5-dimethylpyrazolylmethyl) alaninate, KB(i)PrPZG: potassium N,N-bis(3,5-di-iso-propylpyrazolylmethyl) glycinate, and KB((t)BuM)PZG: potassium N,N-bis(3-methyl-5-tert-butyl-pyrazolylmethyl)glycinate] as structural models of the metalloenzyme alpha-amino-beta-carboxymuconic-epsilon-semialdehyde decarboxylase (ACMSD). These complexes were characterised by several techniques including X-ray crystallographic analysis, X-band EPR, and mass spectrometry (ESI-MS). The crystal structures of 1, 2, 6,7 revealed that they exist as mononuclear Co(II) complexes with trigonal-bipyramidal geometry in the solid state. Compounds 3 and 5 form infinite polymeric chains of Co(II) or Zn(II) complexes, respectively, linked by the pendant carboxylate arms of the BPZG(-) ligand. By comparing the degree of distortion in the penta-coordinate complexes, defined by the Addison-parameter tau, with the value determined for the five-coordinate centres found in the active site of ACMSD, it could be seen that complexes 5 and 7 are very good matches for the geometry of the zinc(II) centre in monomer A of the native enzyme. All complexes could be seen as model compounds for the active site of the enzyme ACMSD, where the Co(II) complexes reflected the structural flexibility found in case of two histidine (His177 and His228) residues found in the active site of the enzyme.
Collapse
Affiliation(s)
- Jessica Gätjens
- University of Michigan, Department of Chemistry, Willard H. Dow Laboratories, 930 North University Ave, Ann Arbor, MI 48109, USA
| | | | | | | | | |
Collapse
|
50
|
Owe-Young R, Webster NL, Mukhtar M, Pomerantz RJ, Smythe G, Walker D, Armati PJ, Crowe SM, Brew BJ. Kynurenine pathway metabolism in human blood-brain-barrier cells: implications for immune tolerance and neurotoxicity. J Neurochem 2008; 105:1346-57. [PMID: 18221377 DOI: 10.1111/j.1471-4159.2008.05241.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The catabolic pathway of l-tryptophan (l-trp), known as the kynurenine pathway (KP), has been implicated in the pathogenesis of a wide range of brain diseases through its ability to lead to immune tolerance and neurotoxicity. As endothelial cells (ECs) and pericytes of the blood-brain-barrier (BBB) are among the first brain-associated cells that a blood-borne pathogen would encounter, we sought to determine their expression of the KP. Using RT-PCR and HPLC/GC-MS, we show that BBB ECs and pericytes constitutively express components of the KP. BBB ECs constitutively synthesized kynurenic acid, and after immune activation, kynurenine (KYN), which is secreted basolaterally. BBB pericytes produced small amounts of picolinic acid and after immune activation, KYN. These results have significant implications for the pathogenesis of inflammatory brain diseases in general, particularly human immunodeficiency virus (HIV)-related brain disease. Kynurenine pathway activation at the BBB results in local immune tolerance and neurotoxicity: the basolateral secretion of excess KYN can be further metabolized by perivascular macrophages and microglia with synthesis of quinolinic acid. The results point to a mechanism whereby a systemic inflammatory signal can be transduced across an intact BBB to cause local neurotoxicity.
Collapse
Affiliation(s)
- Robert Owe-Young
- University of New South Wales Centre for Immunology, St. Vincent's Hospital, Darlinghurst, New South Wales, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|