1
|
Jung ES, Choi H, Mook-Jung I. Decoding microglial immunometabolism: a new frontier in Alzheimer's disease research. Mol Neurodegener 2025; 20:37. [PMID: 40149001 PMCID: PMC11948825 DOI: 10.1186/s13024-025-00825-0] [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: 09/01/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Alzheimer's disease (AD) involves a dynamic interaction between neuroinflammation and metabolic dysregulation, where microglia play a central role. These immune cells undergo metabolic reprogramming in response to AD-related pathology, with key genes such as TREM2, APOE, and HIF-1α orchestrating these processes. Microglial metabolism adapts to environmental stimuli, shifting between oxidative phosphorylation and glycolysis. Hexokinase-2 facilitates glycolytic flux, while AMPK acts as an energy sensor, coordinating lipid and glucose metabolism. TREM2 and APOE regulate microglial lipid homeostasis, influencing Aβ clearance and immune responses. LPL and ABCA7, both associated with AD risk, modulate lipid processing and cholesterol transport, linking lipid metabolism to neurodegeneration. PPARG further supports lipid metabolism by regulating microglial inflammatory responses. Amino acid metabolism also contributes to microglial function. Indoleamine 2,3-dioxygenase controls the kynurenine pathway, producing neurotoxic metabolites linked to AD pathology. Additionally, glucose-6-phosphate dehydrogenase regulates the pentose phosphate pathway, maintaining redox balance and immune activation. Dysregulated glucose and lipid metabolism, influenced by genetic variants such as APOE4, impair microglial responses and exacerbate AD progression. Recent findings highlight the interplay between metabolic regulators like REV-ERBα, which modulates lipid metabolism and inflammation, and Syk, which influences immune responses and Aβ clearance. These insights offer promising therapeutic targets, including strategies aimed at HIF-1α modulation, which could restore microglial function depending on disease stage. By integrating metabolic, immune, and genetic factors, this review underscores the importance of microglial immunometabolism in AD. Targeting key metabolic pathways could provide novel therapeutic strategies for mitigating neuroinflammation and restoring microglial function, ultimately paving the way for innovative treatments in neurodegenerative diseases.
Collapse
Affiliation(s)
- Eun Sun Jung
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Hayoung Choi
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Inhee Mook-Jung
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.
- Korea Dementia Research Center, Seoul, South Korea.
| |
Collapse
|
2
|
Mie Y, Mikami C, Yasutake Y, Shigemura Y, Yamashita T, Tsujino H. Electrochemical Analysis and Inhibition Assay of Immune-Modulating Enzyme, Indoleamine 2,3-Dioxygenase. Pharmaceuticals (Basel) 2025; 18:352. [PMID: 40143129 PMCID: PMC11944389 DOI: 10.3390/ph18030352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Background: An accurate and rapid analysis of human indoleamine 2,3-dioxygenase (hIDO) is crucial for the development of anticancer pharmaceuticals because of the role of hIDO in promoting tumoral immune escape. However, the conventional assay of hIDO is limited by interference from reductants, which are used to reduce the heme iron to begin the hIDO catalytic reaction. Methods: A direct electrochemical method was applied to drive the hIDO reaction. Results: The nanostructured gold electrode enabled the electrochemical reduction of the heme iron of hIDO1. In the presence of substrates (tryptophan and oxygen), a bioelectrocatalytic current was observed, confirming an electrochemically driven hIDO reaction. A well-known inhibitor of hIDO, epacadostat, hindered this catalytic signal according to its concentration, demonstrating the rapid evaluation of its inhibition activity for the hIDO reaction. Through an in silico study using the proposed electrochemical assay system, we discovered a strong inhibitor candidate with a half-maximal inhibitory concentration of 10 nM. Conclusions: An accurate and rapid assay system in drug discovery for hIDO and kynureine pathway-targeted immunotherapy has been developed.
Collapse
Affiliation(s)
- Yasuhiro Mie
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Sapporo 062-8517, Japan (Y.Y.)
| | - Chitose Mikami
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Sapporo 062-8517, Japan (Y.Y.)
| | - Yoshiaki Yasutake
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Sapporo 062-8517, Japan (Y.Y.)
- Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), AIST, Tokyo 169-8555, Japan
| | - Yuki Shigemura
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan (H.T.)
| | - Taku Yamashita
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University, Nishinomiya 663-8179, Japan
| | - Hirofumi Tsujino
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan (H.T.)
- Museum Links, Osaka University, Osaka 560-0043, Japan
| |
Collapse
|
3
|
Baran H, Jan Pietryja M, Kepplinger B. Importance of Modulating Kynurenic Acid Metabolism-Approaches for the Treatment of Dementia. Biomolecules 2025; 15:74. [PMID: 39858468 PMCID: PMC11764436 DOI: 10.3390/biom15010074] [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: 09/11/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
In this article, we focus on kynurenic acid metabolism in neuropsychiatric disorders and the biochemical processes involved in memory and cognitive impairment, followed by different approaches in the fight against dementia. Kynurenic acid-a biochemical part of L-tryptophan catabolism-is synthesized from L-kynurenine by kynurenine aminotransferases. Experimental pharmacological studies have shown that elevated levels of kynurenic acid in the brain are associated with impaired learning and that lowering kynurenic acid levels can improve these symptoms. The discovery of new compounds with the ability to block kynurenine aminotransferases opens new therapeutic avenues for the treatment of memory impairment and dementia. The newly developed Helix pomatia snail model of memory can be used for the assessment of novel pharmacological approaches. Dietary supplementation with natural molecular/herbal extracts, exercise, and physical activity have significant impacts on endogenous pharmacology by reducing kynurenic acid synthesis, and these factors are likely to significantly modulate steady-state biological conditions and delay the negative consequences of aging, including the onset of pathological processes.
Collapse
Affiliation(s)
- Halina Baran
- Karl Landsteiner Research Institute for Neurochemistry, Neuropharmacology, Neurorehabilitation and Pain Therapy, 3362 Mauer-Amstetten, Austria;
- Neurophysiology Unit, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Marcelin Jan Pietryja
- St. Francis Herbarium, Monastery of the Franciscan Friars Minor, 40-760 Katowice, Poland;
| | - Berthold Kepplinger
- Karl Landsteiner Research Institute for Neurochemistry, Neuropharmacology, Neurorehabilitation and Pain Therapy, 3362 Mauer-Amstetten, Austria;
- Department of Neurology, Neuropsychiatric Hospital, 3362 Mauer-Amstetten, Austria
| |
Collapse
|
4
|
Liang Y, Yang Y, Jie Z, Kang X, Xu H, Zhang H, Wu X. Hippocampal GPR35 Participates in the Pathogenesis of Cognitive Deficits and Emotional Alterations Induced by Aβ 1-42 in Mice. Mol Neurobiol 2025; 62:557-582. [PMID: 38878116 DOI: 10.1007/s12035-024-04296-0] [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: 12/21/2023] [Accepted: 06/08/2024] [Indexed: 01/11/2025]
Abstract
The amyloid-beta (Aβ) aggregation in Alzheimer's disease (AD) triggers neuroinflammation, and neurodegeneration, which lead to cognitive deficits along with other neuropsychiatric symptoms, including depression and anxiety. G protein-coupled receptor 35 (GPR35) is expressed in the brain and is involved in metabolic stresses. However, the role of GPR35 in AD pathogenesis remains unknown. Herein, pharmacological blockade, shRNA-mediated knockdown or knockout of GPR35 was performed to investigate the role and mechanisms of GPR35 in Aβ1-42-induced cognitive impairment and emotional alterations in mice. A series of behavioral, histopathological, and biochemical tests were performed in mice. Our results showed that hippocampal GPR35 expression was significantly increased in Aβ1-42-induced and APP/PS1 AD mouse models. Pharmacological blockade or knockdown of GPR35 ameliorated cognitive impairment and emotional alterations induced by Aβ1-42 in mice. We also found that blockade or knockdown of GPR35 decreased the accumulation of Aβ, and improved neuroinflammation, cholinergic system deficiency, and neuronal apoptosis via the RhoA/ROCK2 pathway in Aβ1-42-treaed mice. However, activation of GPR35 aggravates Aβ1-42-induced cognitive deficits and emotional alterations in mice. In addition, genetic deletion of GPR35 protects against the Aβ1-42-induced cognitive deficits and emotional alterations in mice. Moreover, GPR35 could bind to TLR4. These results indicate that GPR35 participates in the pathogenesis of cognitive deficits and emotional alterations induced by Aβ1-42 in mice, suggesting that GPR35 could be a potential therapeutic target for AD.
Collapse
Affiliation(s)
- YuSheng Liang
- School of Pharmacy, The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Yan Yang
- School of Pharmacy, The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Zhi Jie
- School of Pharmacy, The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Xu Kang
- School of Pharmacy, The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Heng Xu
- School of Pharmacy, The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - HaiWang Zhang
- School of Pharmacy, The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Xian Wu
- School of Pharmacy, The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Medical University, Hefei, 230032, China.
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
5
|
Geeraerts Z, Ishigami I, Gao Y, Yeh SR. Heme-based dioxygenases: Structure, function and dynamics. J Inorg Biochem 2024; 261:112707. [PMID: 39217822 PMCID: PMC11590650 DOI: 10.1016/j.jinorgbio.2024.112707] [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: 07/31/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Tryptophan dioxygenase (TDO) and indoleamine 2,3 dioxygenase (IDO) belong to a unique class of heme-based enzymes that insert dioxygen into the essential amino acid, L-tryptophan (Trp), to generate N-formylkynurenine (NFK), a critical metabolite in the kynurenine pathway. Recently, the two dioxygenases were recognized as pivotal cancer immunotherapeutic drug targets, which triggered a great deal of drug discovery targeting them. The advancement of the field is however hampered by the poor understanding of the structural properties of the two enzymes and the mechanisms by which the structures dictate their functions. In this review, we summarize recent findings centered on the structure, function, and dynamics of the human isoforms of the two enzymes.
Collapse
Affiliation(s)
- Zachary Geeraerts
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Izumi Ishigami
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Yuan Gao
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Syun-Ru Yeh
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| |
Collapse
|
6
|
Frost ED, Shi SX, Byroju VV, Pitton Rissardo J, Donlon J, Vigilante N, Murray BP, Walker IM, McGarry A, Ferraro TN, Hanafy KA, Echeverria V, Mitrev L, Kling MA, Krishnaiah B, Lovejoy DB, Rahman S, Stone TW, Koola MM. Galantamine-Memantine Combination in the Treatment of Parkinson's Disease Dementia. Brain Sci 2024; 14:1163. [PMID: 39766362 PMCID: PMC11674513 DOI: 10.3390/brainsci14121163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/16/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that affects over 1% of population over age 60. It is defined by motor and nonmotor symptoms including a spectrum of cognitive impairments known as Parkinson's disease dementia (PDD). Currently, the only US Food and Drug Administration-approved treatment for PDD is rivastigmine, which inhibits acetylcholinesterase and butyrylcholinesterase increasing the level of acetylcholine in the brain. Due to its limited efficacy and side effect profile, rivastigmine is often not prescribed, leaving patients with no treatment options. PD has several derangements in neurotransmitter pathways (dopaminergic neurons in the nigrostriatal pathway, kynurenine pathway (KP), acetylcholine, α7 nicotinic receptor, and N-methyl-D-aspartate (NMDA) receptors) and rivastigmine is only partially effective as it only targets one pathway. Kynurenic acid (KYNA), a metabolite of tryptophan metabolism, affects the pathophysiology of PDD in multiple ways. Both galantamine (α7 nicotinic receptor) and memantine (antagonist of the NMDA subtype of the glutamate receptor) are KYNA modulators. When used in combination, they target multiple pathways. While randomized controlled trials (RCTs) with each drug alone for PD have failed, the combination of galantamine and memantine has demonstrated a synergistic effect on cognitive enhancement in animal models. It has therapeutic potential that has not been adequately assessed, warranting future randomized controlled trials. In this review, we summarize the KYNA-centric model for PD pathophysiology and discuss how this treatment combination is promising in improving cognitive function in patients with PDD through its action on KYNA.
Collapse
Affiliation(s)
- Emma D. Frost
- Cooper Neurological Institute, Cooper University Health Care, Camden, NJ 08103, USA
| | - Swanny X. Shi
- Department of Neurology, Montefiore Medical Center, Bronx, NY 10467, USA
| | - Vishnu V. Byroju
- Cooper Neurological Institute, Cooper University Health Care, Camden, NJ 08103, USA
| | | | - Jack Donlon
- Cooper Medical School, Rowan University, Camden, NJ 08103, USA
| | | | | | - Ian M. Walker
- Cooper Neurological Institute, Cooper University Health Care, Camden, NJ 08103, USA
- Cooper Medical School, Rowan University, Camden, NJ 08103, USA
| | - Andrew McGarry
- Cooper Neurological Institute, Cooper University Health Care, Camden, NJ 08103, USA
- Cooper Medical School, Rowan University, Camden, NJ 08103, USA
| | - Thomas N. Ferraro
- Department of Biomedical Sciences, Cooper Medical School, Rowan University, Camden, NJ 08103, USA
| | - Khalid A. Hanafy
- Cooper Neurological Institute, Cooper University Health Care, Camden, NJ 08103, USA
- Cooper Medical School, Rowan University, Camden, NJ 08103, USA
| | - Valentina Echeverria
- Research and Development Department, Bay Pines VAHCS, Bay Pines, FL 33744, USA
- Medicine Department, Universidad San Sebastián, Concepción 4081339, Bío Bío, Chile
| | - Ludmil Mitrev
- Cooper Medical School, Rowan University, Camden, NJ 08103, USA
| | - Mitchel A. Kling
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Stratford, NJ 08084, USA
| | - Balaji Krishnaiah
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - David B. Lovejoy
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2113, Australia
| | - Shafiqur Rahman
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, SD 57007, USA
| | - Trevor W. Stone
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford OX3 7LD, UK
| | - Maju Mathew Koola
- Department of Public Safety and Correctional Services, Baltimore, MD 21215, USA
| |
Collapse
|
7
|
Shastak Y, Pelletier W. Exploring the role of riboflavin in swine well-being: a literature review. Porcine Health Manag 2024; 10:46. [PMID: 39482748 PMCID: PMC11526614 DOI: 10.1186/s40813-024-00399-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/20/2024] [Indexed: 11/03/2024] Open
Abstract
Riboflavin (vitamin B2) is an essential B-vitamin crucial for the metabolism, development, and overall well-being of porcine species. As pig production intensifies, understanding the micronutrient needs of swine, particularly riboflavin, becomes increasingly vital. Riboflavin acts as a precursor for coenzymes involved in key redox reactions essential for energy production, growth, and immune regulation. Ariboflavinosis can disrupt metabolic functions, leading to impaired growth, reproductive issues, decreased feed efficiency, compromised immune function, ocular problems, and liver dysfunction. To ensure optimal growth and health, pig diets are consistently supplemented with riboflavin-enriched supplements. This review explores the diverse functions of riboflavin in swine metabolism, focusing on biochemical basics, metabolic pathways, riboflavin uptake and distribution, consequences of deficiency, and benefits of adequate intake. It emphasizes the need for optimized riboflavin supplementation strategies tailored to different production stages and environmental conditions. According to recommendations from four major breeding companies, the dietary riboflavin levels for swine are advised to range between 7.5 and 15 mg/kg for piglets, 3.5 to 8.0 mg/kg for finishing gilts and barrows, 4 to 10 mg/kg for gestating sows, and 5 to 10 mg/kg for lactating sows. Advances in precision nutrition, microbial production of riboflavin, and the development of functional feed additives are potential innovations to enhance swine health, growth performance, and sustainability. Comprehensive studies on the long-term effects of subclinical riboflavin deficiency and the broader health and welfare implications of supplementation are also needed. Addressing knowledge gaps and embracing future trends and innovations will be key to optimizing riboflavin supplementation and advancing the swine industry.
Collapse
Affiliation(s)
- Yauheni Shastak
- BASF SE, Nutrition & Health Division, 67063, Ludwigshafen am Rhein, Germany.
| | - Wolf Pelletier
- BASF SE, Nutrition & Health Division, 67063, Ludwigshafen am Rhein, Germany
| |
Collapse
|
8
|
Kearns R. Gut-Brain Axis and Neuroinflammation: The Role of Gut Permeability and the Kynurenine Pathway in Neurological Disorders. Cell Mol Neurobiol 2024; 44:64. [PMID: 39377830 PMCID: PMC11461658 DOI: 10.1007/s10571-024-01496-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: 05/17/2024] [Accepted: 09/13/2024] [Indexed: 10/09/2024]
Abstract
The increasing prevalence of neurological disorders such as Alzheimer's, Parkinson's, and multiple sclerosis presents a significant global health challenge. Despite extensive research, the precise mechanisms underlying these conditions remain elusive, with current treatments primarily addressing symptoms rather than root causes. Emerging evidence suggests that gut permeability and the kynurenine pathway are involved in the pathogenesis of these neurological conditions, offering promising targets for novel therapeutic and preventive strategies. Gut permeability refers to the intestinal lining's ability to selectively allow essential nutrients into the bloodstream while blocking harmful substances. Various factors, including poor diet, stress, infections, and genetic predispositions, can compromise gut integrity, leading to increased permeability. This condition facilitates the translocation of toxins and bacteria into systemic circulation, triggering widespread inflammation that impacts neurological health via the gut-brain axis. The gut-brain axis (GBA) is a complex communication network between the gut and the central nervous system. Dysbiosis, an imbalance in the gut microbiota, can increase gut permeability and systemic inflammation, exacerbating neuroinflammation-a key factor in neurological disorders. The kynurenine pathway, the primary route for tryptophan metabolism, is significantly implicated in this process. Dysregulation of the kynurenine pathway in the context of inflammation leads to the production of neurotoxic metabolites, such as quinolinic acid, which contribute to neuronal damage and the progression of neurological disorders. This narrative review highlights the potential and progress in understanding these mechanisms. Interventions targeting the kynurenine pathway and maintaining a balanced gut microbiota through diet, probiotics, and lifestyle modifications show promise in reducing neuroinflammation and supporting brain health. In addition, pharmacological approaches aimed at modulating the kynurenine pathway directly, such as inhibitors of indoleamine 2,3-dioxygenase, offer potential avenues for new treatments. Understanding and targeting these interconnected pathways are crucial for developing effective strategies to prevent and manage neurological disorders.
Collapse
Affiliation(s)
- Rowan Kearns
- Ulster University, Life and Health Sciences, Belfast, UK.
| |
Collapse
|
9
|
Zhang S, Lu J, Jin Z, Xu H, Zhang D, Chen J, Wang J. Gut microbiota metabolites: potential therapeutic targets for Alzheimer's disease? Front Pharmacol 2024; 15:1459655. [PMID: 39355779 PMCID: PMC11442227 DOI: 10.3389/fphar.2024.1459655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/05/2024] [Indexed: 10/03/2024] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive decline in cognitive function, which significantly increases pain and social burden. However, few therapeutic interventions are effective in preventing or mitigating the progression of AD. An increasing number of recent studies support the hypothesis that the gut microbiome and its metabolites may be associated with upstream regulators of AD pathology. Methods In this review, we comprehensively explore the potential mechanisms and currently available interventions targeting the microbiome for the improvement of AD. Our discussion is structured around modern research advancements in AD, the bidirectional communication between the gut and brain, the multi-target regulatory effects of microbial metabolites on AD, and therapeutic strategies aimed at modulating gut microbiota to manage AD. Results The gut microbiota plays a crucial role in the pathogenesis of AD through continuous bidirectional communication via the microbiota-gut-brain axis. Among these, microbial metabolites such as lipids, amino acids, bile acids and neurotransmitters, especially sphingolipids and phospholipids, may serve as central components of the gut-brain axis, regulating AD-related pathogenic mechanisms including β-amyloid metabolism, Tau protein phosphorylation, and neuroinflammation. Additionally, interventions such as probiotic administration, fecal microbiota transplantation, and antibiotic use have also provided evidence supporting the association between gut microbiota and AD. At the same time, we propose an innovative strategy for treating AD: a healthy lifestyle combined with targeted probiotics and other potential therapeutic interventions, aiming to restore intestinal ecology and microbiota balance. Conclusion Despite previous efforts, the molecular mechanisms by which gut microbes act on AD have yet to be fully described. However, intestinal microorganisms may become an essential target for connecting the gut-brain axis and improving the symptoms of AD. At the same time, it requires joint exploration by multiple centers and multiple disciplines.
Collapse
Affiliation(s)
- Shanshan Zhang
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Jing Lu
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Ziqi Jin
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Hanying Xu
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Dongmei Zhang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Jianan Chen
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Jian Wang
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
10
|
Kearns R. The Kynurenine Pathway in Gut Permeability and Inflammation. Inflammation 2024:10.1007/s10753-024-02135-x. [PMID: 39256304 DOI: 10.1007/s10753-024-02135-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/09/2024] [Accepted: 08/21/2024] [Indexed: 09/12/2024]
Abstract
The gut-brain axis (GBA) is a crucial communication network linking the gastrointestinal (GI) tract and the central nervous system (CNS). The gut microbiota significantly influences metabolic, immune, and neural functions by generating a diverse array of bioactive compounds that modulate brain function and maintain homeostasis. A pivotal mechanism in this communication is the kynurenine pathway, which metabolises tryptophan into various derivatives, including neuroactive and neurotoxic compounds. Alterations in gut microbiota composition can increase gut permeability, triggering inflammation and neuroinflammation, and contributing to neuropsychiatric disorders. This review elucidates the mechanisms by which changes in gut permeability may lead to systemic inflammation and neuroinflammation, with a focus on the kynurenine pathway. We explore how probiotics can modulate the kynurenine pathway and reduce neuroinflammation, highlighting their potential as therapeutic interventions for neuropsychiatric disorders. The review integrates experimental data, discusses the balance between neurotoxic and neuroprotective kynurenine metabolites, and examines the role of probiotics in regulating inflammation, cognitive development, and gut-brain axis functions. The insights provided aim to guide future research and therapeutic strategies for mitigating GI complaints and their neurological consequences.
Collapse
Affiliation(s)
- Rowan Kearns
- Ulster University, Life and Health Sciences, Newry, Northern Ireland, United Kingdom.
| |
Collapse
|
11
|
Coretti L, Buommino E, Lembo F. The aryl hydrocarbon receptor pathway: a linking bridge between the gut microbiome and neurodegenerative diseases. Front Cell Neurosci 2024; 18:1433747. [PMID: 39175504 PMCID: PMC11338779 DOI: 10.3389/fncel.2024.1433747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/31/2024] [Indexed: 08/24/2024] Open
Abstract
The Aryl hydrocarbon receptor (AHR) is a cytosolic receptor and ligand-activated transcription factor widely expressed across various cell types in the body. Its signaling is vital for host responses at barrier sites, regulating epithelial renewal, barrier integrity, and the activities of several types of immune cells. This makes AHR essential for various cellular responses during aging, especially those governing inflammation and immunity. In this review, we provided an overview of the mechanisms by which the AHR mediates inflammatory response at gut and brain level through signals from intestinal microbes. The age-related reduction of gut microbiota functions is perceived as a trigger of aberrant immune responses linking gut and brain inflammation to neurodegeneration. Thus, we explored gut microbiome impact on the nature and availability of AHR ligands and outcomes for several signaling pathways involved in neurodegenerative diseases and age-associated decline of brain functions, with an insight on Parkinson's and Alzheimer's diseases, the most common neurodegenerative diseases in the elderly. Specifically, we focused on microbial tryptophan catabolism responsible for the production of several AHR ligands. Perspectives for the development of microbiota-based interventions targeting AHR activity are presented for a healthy aging.
Collapse
Affiliation(s)
- Lorena Coretti
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | | | - Francesca Lembo
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| |
Collapse
|
12
|
Sobczuk J, Paczkowska K, Andrusiów S, Bolanowski M, Daroszewski J. Are Women with Polycystic Ovary Syndrome at Increased Risk of Alzheimer Disease? Lessons from Insulin Resistance, Tryptophan and Gonadotropin Disturbances and Their Link with Amyloid-Beta Aggregation. Biomolecules 2024; 14:918. [PMID: 39199306 PMCID: PMC11352735 DOI: 10.3390/biom14080918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
Alzheimer disease, the leading cause of dementia, and polycystic ovary syndrome, one of the most prevalent female endocrine disorders, appear to be unrelated conditions. However, studies show that both disease entities have common risk factors, and the amount of certain protein marker of neurodegeneration is increased in PCOS. Reports on the pathomechanism of both diseases point to the possibility of common denominators linking them. Dysregulation of the kynurenine pathway, insulin resistance, and impairment of the hypothalamic-pituitary-gonadal axis, which are correlated with amyloid-beta aggregation are these common areas. This article discusses the relationship between Alzheimer disease and polycystic ovary syndrome, with a particular focus on the role of disorders of tryptophan metabolism in both conditions. Based on a review of the available literature, we concluded that systemic changes occurring in PCOS influence the increased risk of neurodegeneration.
Collapse
Affiliation(s)
- Joachim Sobczuk
- Department of Endocrinology, Diabetes and Isotope Therapy, University Clinical Hospital, 50-367 Wroclaw, Poland
| | | | - Szymon Andrusiów
- Department of Neurology, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Marek Bolanowski
- Department of Endocrinology, Diabetes and Isotope Therapy, University Clinical Hospital, 50-367 Wroclaw, Poland
- Department of Endocrinology, Diabetes and Isotope Therapy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Jacek Daroszewski
- Department of Endocrinology, Diabetes and Isotope Therapy, University Clinical Hospital, 50-367 Wroclaw, Poland
- Department of Endocrinology, Diabetes and Isotope Therapy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| |
Collapse
|
13
|
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
|
14
|
Taenzer M, Löffler-Ragg J, Schroll A, Monfort-Lanzas P, Engl S, Weiss G, Brigo N, Kurz K. Urine Metabolite Analysis to Identify Pathomechanisms of Long COVID: A Pilot Study. Int J Tryptophan Res 2023; 16:11786469231220781. [PMID: 38144169 PMCID: PMC10748708 DOI: 10.1177/11786469231220781] [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: 07/24/2023] [Accepted: 11/28/2023] [Indexed: 12/26/2023] Open
Abstract
Background Around 10% of people who had COVID-9 infection suffer from persistent symptoms such as fatigue, dyspnoea, chest pain, arthralgia/myalgia, sleep disturbances, cognitive dysfunction and impairment of mental health. Different underlying pathomechanisms appear to be involved, in particular inflammation, alterations in amino acid metabolism, autonomic dysfunction and gut dysbiosis. Aim As routine tests are often inconspicuous in patients with Long COVID (LC), similarly to patients suffering from myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), accessible biomarkers indicating dysregulation of specific pathways are urgently needed to identify underlying pathomechanisms and enable personalized medicine treatment. Within this pilot study we aimed to proof traceability of altered metabolism by urine analysis. Patients and Methods Urine metabolome analyses were performed to investigate the metabolic signature of patients with LC (n = 25; 20 women, 5 men) in comparison to healthy controls (Ctrl, n = 8; 7 women, 1 man) and individuals with ME/CFS (n = 8; 2 women, 6 men). Concentrations of neurotransmitter precursors tryptophan, phenylalanine and their downstream metabolites, as well as their association with symptoms (fatigue, anxiety and depression) in the patients were examined. Results and Conclusion Phenylalanine levels were significantly lower in both the LC and ME/CFS patient groups when compared to the Ctrl group. In many LC patients, the concentrations of downstream metabolites of tryptophan and tyrosine, such as serotonin, dopamine and catecholamines, deviated from the reference ranges. Several symptoms (sleep disturbance, pain or autonomic dysfunction) were associated with certain metabolites. Patients experiencing fatigue had lower levels of kynurenine, phenylalanine and a reduced kynurenine to tryptophan ratio (Kyn/Trp). Lower concentrations of gamma-aminobutyric acid (GABA) and higher activity of kynurenine 3-monooxygenase (KMO) were observed in patients with anxiety. Conclusively, our results suggest that amino acid metabolism and neurotransmitter synthesis is disturbed in patients with LC and ME/CFS. The identified metabolites and their associated dysregulations could serve as potential biomarkers for elucidating underlying pathomechanisms thus enabling personalized treatment strategies for these patient populations.
Collapse
Affiliation(s)
- Maja Taenzer
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Judith Löffler-Ragg
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Andrea Schroll
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Pablo Monfort-Lanzas
- Institute of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Sabine Engl
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Natascha Brigo
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Katharina Kurz
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
15
|
Bakker L, Ramakers IHGB, J P M Eussen S, Choe K, van den Hove DLA, Kenis G, Rutten BPF, van Oostenbrugge RJ, Staals J, Ulvik A, Ueland PM, Verhey FRJ, Köhler S. The role of the kynurenine pathway in cognitive functioning after stroke: A prospective clinical study. J Neurol Sci 2023; 454:120819. [PMID: 37852105 DOI: 10.1016/j.jns.2023.120819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND The kynurenine pathway is the main metabolic pathway of tryptophan degradation and has been associated with stroke and impaired cognitive functioning, but studies on its role in post-stroke cognitive impairment (PSCI) are scarce. We aimed to investigate associations between metabolites of the kynurenine pathway at baseline and post-stroke cognitive functioning over time. METHODS Baseline plasma kynurenines were quantified in 198 stroke patients aged 65.4 ± 10.8 years, 138 (69.7%) men, who were followed up over a period of three years after stroke. Baseline and longitudinal associations of kynurenines with PSCI and cognitive domain scores were investigated using linear mixed models, adjusted for several confounders. RESULTS No evidence of associations between kynurenines and odds of PSCI were found. However, considering individual cognitive domains, higher plasma levels of anthranilic acid (AA) were associated with better episodic memory at baseline (β per SD 0.16 [0.05, 0.28]). Additionally, a linear-quadratic association was found for the kynurenic acid/ quinolinic acid ratio (KA/QA), a neuroprotective index, with episodic memory (Wald χ2 = 8.27, p = .016). Higher levels of KA were associated with better processing speed in women only (pinteraction = .008; β per SD 0.15 [95% CI 0.02, 0.27]). These associations did not change over time. CONCLUSIONS Higher levels of KA, AA and KA/QA were associated with better scores on some cognitive domains at baseline. These associations did not change over time. Given the exploratory nature and heterogeneity of findings, these results should be interpreted with caution, and verified in other prospective studies.
Collapse
Affiliation(s)
- Lieke Bakker
- Alzheimer Center Limburg, Maastricht University, 6229 ET Maastricht, the Netherlands; Department of Psychiatry and Neuropsychology, Maastricht University, 6229 ER Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; EURON European Graduate School of Neuroscience, 6229 ER Maastricht, the Netherlands.
| | - Inez H G B Ramakers
- Alzheimer Center Limburg, Maastricht University, 6229 ET Maastricht, the Netherlands; Department of Psychiatry and Neuropsychology, Maastricht University, 6229 ER Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; EURON European Graduate School of Neuroscience, 6229 ER Maastricht, the Netherlands.
| | - Simone J P M Eussen
- Department of Epidemiology, Maastricht University, 6229 HA Maastricht, the Netherlands; School for Cardiovascular Diseases (CARIM), 6229 ER Maastricht, the Netherlands; Care and Public Health Research Institute (CAPHRI), 6229 ER Maastricht, the Netherlands.
| | - Kyonghwan Choe
- Department of Psychiatry and Neuropsychology, Maastricht University, 6229 ER Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; EURON European Graduate School of Neuroscience, 6229 ER Maastricht, the Netherlands.
| | - Daniel L A van den Hove
- Department of Psychiatry and Neuropsychology, Maastricht University, 6229 ER Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; EURON European Graduate School of Neuroscience, 6229 ER Maastricht, the Netherlands; Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, 97080 Wuerzburg, Germany.
| | - Gunter Kenis
- Department of Psychiatry and Neuropsychology, Maastricht University, 6229 ER Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; EURON European Graduate School of Neuroscience, 6229 ER Maastricht, the Netherlands.
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, Maastricht University, 6229 ER Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; EURON European Graduate School of Neuroscience, 6229 ER Maastricht, the Netherlands.
| | - Robert J van Oostenbrugge
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; School for Cardiovascular Diseases (CARIM), 6229 ER Maastricht, the Netherlands; Department of Neurology, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, the Netherlands.
| | - Julie Staals
- School for Cardiovascular Diseases (CARIM), 6229 ER Maastricht, the Netherlands; Department of Neurology, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, the Netherlands.
| | | | | | - Frans R J Verhey
- Alzheimer Center Limburg, Maastricht University, 6229 ET Maastricht, the Netherlands; Department of Psychiatry and Neuropsychology, Maastricht University, 6229 ER Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; EURON European Graduate School of Neuroscience, 6229 ER Maastricht, the Netherlands.
| | - Sebastian Köhler
- Alzheimer Center Limburg, Maastricht University, 6229 ET Maastricht, the Netherlands; Department of Psychiatry and Neuropsychology, Maastricht University, 6229 ER Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, the Netherlands; EURON European Graduate School of Neuroscience, 6229 ER Maastricht, the Netherlands.
| |
Collapse
|
16
|
Sadok I, Jędruchniewicz K. Dietary Kynurenine Pathway Metabolites-Source, Fate, and Chromatographic Determinations. Int J Mol Sci 2023; 24:16304. [PMID: 38003492 PMCID: PMC10671297 DOI: 10.3390/ijms242216304] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Tryptophan metabolism plays an essential role in human health. In mammals, about 95% of dietary tryptophan is metabolized through the kynurenine pathway, which is associated with the development of several pathologies, including neurodegeneration. Some of the kynurenine pathway metabolites are agonists of the aryl hydrocarbon receptor involved in metabolic functions, inflammation, and carcinogenesis. Thus, their origins, fates, and roles are of widespread interest. Except for being produced endogenously, these metabolites can originate from exogenous sources (e.g., food) and undergo absorption in the digestive tract. Recently, a special focus on exogenous sources of tryptophan metabolites was observed. This overview summarizes current knowledge about the occurrence of the kynurenine pathway metabolites (kynurenines) in food and the analytical method utilized for their determination in different food matrices. Special attention was paid to sample preparation and chromatographic analysis, which has proven to be a core technique for the detection and quantification of kynurenines. A discussion of the fate and role of dietary kynurenines has also been addressed. This review will, hopefully, guide further studies on the impact of dietary kynurenines on human health.
Collapse
Affiliation(s)
- Ilona Sadok
- Laboratory of Separation and Spectroscopic Method Applications, Department of Chemistry, Institute of Biological Sciences, Faculty of Medicine, The John Paul II Catholic University of Lublin, 20-708 Lublin, Poland
| | - Katarzyna Jędruchniewicz
- Laboratory of Separation and Spectroscopic Method Applications, The John Paul II Catholic University of Lublin, 20-708 Lublin, Poland;
| |
Collapse
|
17
|
Önder C, Akdoğan N, Kurgan Ş, Balci N, Serdar CC, Serdar MA, Günhan M. Does smoking influence tryptophan metabolism in periodontal inflammation? A cross-sectional study. J Periodontal Res 2023; 58:1041-1051. [PMID: 37526075 DOI: 10.1111/jre.13166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 06/11/2023] [Accepted: 07/20/2023] [Indexed: 08/02/2023]
Abstract
OBJECTIVES The aim of this study was to identify the effects of smoking and periodontal inflammation on tryptophan-kynurenine metabolism as well as the correlation between these findings and clinical periodontal parameters. BACKGROUND It has been shown that the tryptophan amino acid's primary catabolic pathway, the kynurenine pathway (KP), may serve as a key biomarker for periodontal disease. Although there are studies investigating the effect of smoking on KYN-TRP metabolism, the effect of smoking on periodontal disease through KP has not been revealed so far. METHODS The salivary and serum samples were gathered from 24 nonsmoker (NS-P) stage III, grade B generalized periodontitis and 22 smoker (S-P) stage III, grade C generalized periodontitis patients, in addition to 24 nonsmoker (NS-C) and 24 smoker (S-C) periodontally healthy control individuals. Saliva and serum IL-6, kynurenine (KYN), and tryptophan (TRP) values, and KYN/TRP ratio were analyzed by liquid chromatography-mass spectrometry. Clinical periodontal measurements were recorded. RESULTS Salivary TRP values were significantly higher in both periodontitis groups than control groups (p < .05). Salivary KYN values were highest in NS-P group (p < .05). Salivary KYN values did not differ significantly between periodontitis groups (p = .84). Salivary KYN/TRP ratio was significantly lower in NS-P group compared to other groups (p < .001). Serum TRP value is higher in S-P group than other groups; however, significant difference was found in S-C group (p < .05). Serum KYN values were significantly lower in smokers than nonsmokers. Serum KYN/TRP ratio is higher in NS-P group. NS-P group has the highest salivary IL-6 levels, NS-C group has the lowest values (p < .05). CONCLUSIONS Our results point out that smoking exacerbates inflammation in the periodontium and increases TRP destruction and decreases IDO activity by suppressing KP in serum. As a result, kynurenine and its metabolites may be significant biomarkers in the link between smoking and periodontal disease.
Collapse
Affiliation(s)
- Canan Önder
- Department of Periodontology, Faculty of Dentistry, Ankara University, Ankara, Turkey
| | - Nihan Akdoğan
- Department of Periodontology, Faculty of Dentistry, Ankara University, Ankara, Turkey
| | - Şivge Kurgan
- Department of Periodontology, Faculty of Dentistry, Ankara University, Ankara, Turkey
| | - Nur Balci
- Department of Periodontology, Faculty of Dentistry, Medipol University, İstanbul, Turkey
| | - Ceyhan Ceran Serdar
- Department of Medical Biology and Genetics, Faculty of Medicine, Ankara Medipol University, Ankara, Turkey
| | - Muhittin A Serdar
- Department of Medical Biochemistry, Faculty of Medicine, Acıbadem University, Ankara, Turkey
| | - Meral Günhan
- Department of Periodontology, Faculty of Dentistry, Ankara University, Ankara, Turkey
| |
Collapse
|
18
|
Xue C, Li G, Zheng Q, Gu X, Shi Q, Su Y, Chu Q, Yuan X, Bao Z, Lu J, Li L. Tryptophan metabolism in health and disease. Cell Metab 2023; 35:1304-1326. [PMID: 37352864 DOI: 10.1016/j.cmet.2023.06.004] [Citation(s) in RCA: 282] [Impact Index Per Article: 141.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/10/2023] [Accepted: 06/05/2023] [Indexed: 06/25/2023]
Abstract
Tryptophan (Trp) metabolism primarily involves the kynurenine, 5-hydroxytryptamine, and indole pathways. A variety of bioactive compounds produced via Trp metabolism can regulate various physiological functions, including inflammation, metabolism, immune responses, and neurological function. Emerging evidence supports an intimate relationship between Trp metabolism disorder and diseases. The levels or ratios of Trp metabolites are significantly associated with many clinical features. Additionally, studies have shown that disease progression can be controlled by modulating Trp metabolism. Indoleamine-2,3-dioxygenase, Trp-2,3-dioxygenase, kynurenine-3-monooxygenase, and Trp hydroxylase are the rate-limiting enzymes that are critical for Trp metabolism. These key regulatory enzymes can be targeted for treating several diseases, including tumors. These findings provide novel insights into the treatment of diseases. In this review, we have summarized the recent research progress on the role of Trp metabolites in health and disease along with their clinical applications.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ganglei Li
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qiuxian Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xinyu Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuanshuai Su
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhengyi Bao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
19
|
Shaw C, Hess M, Weimer BC. Microbial-Derived Tryptophan Metabolites and Their Role in Neurological Disease: Anthranilic Acid and Anthranilic Acid Derivatives. Microorganisms 2023; 11:1825. [PMID: 37512997 PMCID: PMC10384668 DOI: 10.3390/microorganisms11071825] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
The gut microbiome provides the host access to otherwise indigestible nutrients, which are often further metabolized by the microbiome into bioactive components. The gut microbiome can also shift the balance of host-produced compounds, which may alter host health. One precursor to bioactive metabolites is the essential aromatic amino acid tryptophan. Tryptophan is mostly shunted into the kynurenine pathway but is also the primary metabolite for serotonin production and the bacterial indole pathway. Balance between tryptophan-derived bioactive metabolites is crucial for neurological homeostasis and metabolic imbalance can trigger or exacerbate neurological diseases. Alzheimer's, depression, and schizophrenia have been linked to diverging levels of tryptophan-derived anthranilic, kynurenic, and quinolinic acid. Anthranilic acid from collective microbiome metabolism plays a complex but important role in systemic host health. Although anthranilic acid and its metabolic products are of great importance for host-microbe interaction in neurological health, literature examining the mechanistic relationships between microbial production, host regulation, and neurological diseases is scarce and at times conflicting. This narrative review provides an overview of the current understanding of anthranilic acid's role in neurological health and disease, with particular focus on the contribution of the gut microbiome, the gut-brain axis, and the involvement of the three major tryptophan pathways.
Collapse
Affiliation(s)
- Claire Shaw
- Department of Population Health and Reproduction, 100K Pathogen Genome Project, University of California Davis, Davis, CA 95616, USA
- Department of Animal Science, College of Agricultural and Environmental Sciences, University of California Davis, Davis, CA 95616, USA
| | - Matthias Hess
- Department of Animal Science, College of Agricultural and Environmental Sciences, University of California Davis, Davis, CA 95616, USA
| | - Bart C Weimer
- Department of Population Health and Reproduction, 100K Pathogen Genome Project, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
20
|
Dehhaghi M, Heng B, Guillemin GJ. The kynurenine pathway in traumatic brain injuries and concussion. Front Neurol 2023; 14:1210453. [PMID: 37360356 PMCID: PMC10289013 DOI: 10.3389/fneur.2023.1210453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Up to 10 million people per annum experience traumatic brain injury (TBI), 80-90% of which are categorized as mild. A hit to the brain can cause TBI, which can lead to secondary brain injuries within minutes to weeks after the initial injury through unknown mechanisms. However, it is assumed that neurochemical changes due to inflammation, excitotoxicity, reactive oxygen species, etc., that are triggered by TBI are associated with the emergence of secondary brain injuries. The kynurenine pathway (KP) is an important pathway that gets significantly overactivated during inflammation. Some KP metabolites such as QUIN have neurotoxic effects suggesting a possible mechanism through which TBI can cause secondary brain injury. That said, this review scrutinizes the potential association between KP and TBI. A more detailed understanding of the changes in KP metabolites during TBI is essential to prevent the onset or at least attenuate the severity of secondary brain injuries. Moreover, this information is crucial for the development of biomarker/s to probe the severity of TBI and predict the risk of secondary brain injuries. Overall, this review tries to fill the knowledge gap about the role of the KP in TBI and highlights the areas that need to be studied.
Collapse
|
21
|
Kesarwani P, Kant S, Zhao Y, Prabhu A, Buelow KL, Miller CR, Chinnaiyan P. Quinolinate promotes macrophage-induced immune tolerance in glioblastoma through the NMDAR/PPARγ signaling axis. Nat Commun 2023; 14:1459. [PMID: 36927729 PMCID: PMC10020159 DOI: 10.1038/s41467-023-37170-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 03/01/2023] [Indexed: 03/18/2023] Open
Abstract
There has been considerable scientific effort dedicated to understanding the biologic consequence and therapeutic implications of aberrant tryptophan metabolism in brain tumors and neurodegenerative diseases. A majority of this work has focused on the upstream metabolism of tryptophan; however, this has resulted in limited clinical application. Using global metabolomic profiling of patient-derived brain tumors, we identify the downstream metabolism of tryptophan and accumulation of quinolinate (QA) as a metabolic node in glioblastoma and demonstrate its critical role in promoting immune tolerance. QA acts as a metabolic checkpoint in glioblastoma by inducing NMDA receptor activation and Foxo1/PPARγ signaling in macrophages, resulting in a tumor supportive phenotype. Using a genetically-engineered mouse model designed to inhibit production of QA, we identify kynureninase as a promising therapeutic target to revert the potent immune suppressive microenvironment in glioblastoma. These findings offer an opportunity to revisit the biologic consequence of this pathway as it relates to oncogenesis and neurodegenerative disease and a framework for developing immune modulatory agents to further clinical gains in these otherwise incurable diseases.
Collapse
Affiliation(s)
- Pravin Kesarwani
- Department of Radiation Oncology, Corewell Health East, Royal Oak, MI, USA
| | - Shiva Kant
- Department of Radiation Oncology, Corewell Health East, Royal Oak, MI, USA
| | - Yi Zhao
- Department of Radiation Oncology, Corewell Health East, Royal Oak, MI, USA
| | - Antony Prabhu
- Department of Radiation Oncology, Corewell Health East, Royal Oak, MI, USA
| | - Katie L Buelow
- Department of Radiation Oncology, Corewell Health East, Royal Oak, MI, USA
| | - C Ryan Miller
- Department of Pathology, Division of Neuropathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Prakash Chinnaiyan
- Department of Radiation Oncology, Corewell Health East, Royal Oak, MI, USA.
- Oakland University William Beaumont School of Medicine, Royal Oak, MI, USA.
| |
Collapse
|
22
|
Ge X, Zheng M, Hu M, Fang X, Geng D, Liu S, Wang L, Zhang J, Guan L, Zheng P, Xie Y, Pan W, Zhou M, Zhou L, Tang R, Zheng K, Yu Y, Huang XF. Butyrate ameliorates quinolinic acid-induced cognitive decline in obesity models. J Clin Invest 2023; 133:154612. [PMID: 36787221 PMCID: PMC9927952 DOI: 10.1172/jci154612] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/20/2022] [Indexed: 02/15/2023] Open
Abstract
Obesity is a risk factor for neurodegenerative disease associated with cognitive dysfunction, including Alzheimer's disease. Low-grade inflammation is common in obesity, but the mechanism between inflammation and cognitive impairment in obesity is unclear. Accumulative evidence shows that quinolinic acid (QA), a neuroinflammatory neurotoxin, is involved in the pathogenesis of neurodegenerative processes. We investigated the role of QA in obesity-induced cognitive impairment and the beneficial effect of butyrate in counteracting impairments of cognition, neural morphology, and signaling. We show that in human obesity, there was a negative relationship between serum QA levels and cognitive function and decreased cortical gray matter. Diet-induced obese mice had increased QA levels in the cortex associated with cognitive impairment. At single-cell resolution, we confirmed that QA impaired neurons, altered the dendritic spine's intracellular signal, and reduced brain-derived neurotrophic factor (BDNF) levels. Using Caenorhabditis elegans models, QA induced dopaminergic and glutamatergic neuron lesions. Importantly, the gut microbiota metabolite butyrate was able to counteract those alterations, including cognitive impairment, neuronal spine loss, and BDNF reduction in both in vivo and in vitro studies. Finally, we show that butyrate prevented QA-induced BDNF reductions by epigenetic enhancement of H3K18ac at BDNF promoters. These findings suggest that increased QA is associated with cognitive decline in obesity and that butyrate alleviates neurodegeneration.
Collapse
Affiliation(s)
- Xing Ge
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Mingxuan Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Minmin Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Xiaoli Fang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Sha Liu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Li Wang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Jun Zhang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Li Guan
- The Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Peng Zheng
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, New South Wales, Australia
| | - Yuanyi Xie
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, New South Wales, Australia
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Menglu Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Limian Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Xu-Feng Huang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China.,Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, New South Wales, Australia
| |
Collapse
|
23
|
Sheng C, Chu X, He Y, Ding Q, Jia S, Shi Q, Sun R, Song L, Du W, Liang Y, Chen N, Yang Y, Wang X. Alterations in Peripheral Metabolites as Key Actors in Alzheimer's Disease. Curr Alzheimer Res 2023; 20:379-393. [PMID: 37622711 DOI: 10.2174/1567205020666230825091147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/24/2023] [Accepted: 07/20/2023] [Indexed: 08/26/2023]
Abstract
Growing evidence supports that Alzheimer's disease (AD) could be regarded as a metabolic disease, accompanying central and peripheral metabolic disturbance. Nowadays, exploring novel and potentially alternative hallmarks for AD is needed. Peripheral metabolites based on blood and gut may provide new biochemical insights about disease mechanisms. These metabolites can influence brain energy homeostasis, maintain gut mucosal integrity, and regulate the host immune system, which may further play a key role in modulating the cognitive function and behavior of AD. Recently, metabolomics has been used to identify key AD-related metabolic changes and define metabolic changes during AD disease trajectory. This review aims to summarize the key blood- and microbial-derived metabolites that are altered in AD and identify the potential metabolic biomarkers of AD, which will provide future targets for precision therapeutic modulation.
Collapse
Affiliation(s)
- Can Sheng
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Xu Chu
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Yan He
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Qingqing Ding
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Shulei Jia
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qiguang Shi
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Ran Sun
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Li Song
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Wenying Du
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Yuan Liang
- Department of Clinical Medicine, Jining Medical University, Jining, 272067, China
| | - Nian Chen
- Department of Clinical Medicine, Jining Medical University, Jining, 272067, China
| | - Yan Yang
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Xiaoni Wang
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
| |
Collapse
|
24
|
Jo JK, Lee G, Nguyen CD, Park SE, Kim EJ, Kim HW, Seo SH, Cho KM, Kwon SJ, Kim JH, Son HS. Effects of Donepezil Treatment on Brain Metabolites, Gut Microbiota, and Gut Metabolites in an Amyloid Beta-Induced Cognitive Impairment Mouse Pilot Model. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196591. [PMID: 36235127 PMCID: PMC9572896 DOI: 10.3390/molecules27196591] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 12/12/2022]
Abstract
Accumulated clinical and biomedical evidence indicates that the gut microbiota and their metabolites affect brain function and behavior in various central nervous system disorders. This study was performed to investigate the changes in brain metabolites and composition of the fecal microbial community following injection of amyloid β (Aβ) and donepezil treatment of Aβ-injected mice using metataxonomics and metabolomics. Aβ treatment caused cognitive dysfunction, while donepezil resulted in the successful recovery of memory impairment. The Aβ + donepezil group showed a significantly higher relative abundance of Verrucomicrobia than the Aβ group. The relative abundance of 12 taxa, including Blautia and Akkermansia, differed significantly between the groups. The Aβ + donepezil group had higher levels of oxalate, glycerol, xylose, and palmitoleate in feces and oxalate, pyroglutamic acid, hypoxanthine, and inosine in brain tissues than the Aβ group. The levels of pyroglutamic acid, glutamic acid, and phenylalanine showed similar changes in vivo and in vitro using HT-22 cells. The major metabolic pathways in the brain tissues and gut microbiota affected by Aβ or donepezil treatment of Aβ-injected mice were related to amino acid pathways and sugar metabolism, respectively. These findings suggest that alterations in the gut microbiota might influence the induction and amelioration of Aβ-induced cognitive dysfunction via the gut–brain axis. This study could provide basic data on the effects of Aβ and donepezil on gut microbiota and metabolites in an Aβ-induced cognitive impairment mouse model.
Collapse
Affiliation(s)
- Jae-Kwon Jo
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Gihyun Lee
- Department of Korean Medicine, Dongshin University, Naju 58245, Korea
| | - Cong Duc Nguyen
- Department of Korean Medicine, Dongshin University, Naju 58245, Korea
| | - Seong-Eun Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Eun-Ju Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Hyun-Woo Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | | | | | | | - Jae-Hong Kim
- Department of Acupuncture and Moxibustion Medicine, College of Korean Medicine, Dongshin University, Naju 58245, Korea
- Correspondence: (J.-H.K.); (H.-S.S.); Tel.: +82-62-350-7209 (J.-H.K.); +82-2-3290-3053 (H.-S.S.)
| | - Hong-Seok Son
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
- Correspondence: (J.-H.K.); (H.-S.S.); Tel.: +82-62-350-7209 (J.-H.K.); +82-2-3290-3053 (H.-S.S.)
| |
Collapse
|
25
|
Pradhan SS, Thota SM, Rajaratnam S, Bhagavatham SKS, Pulukool SK, Rathnakumar S, Phalguna KS, Dandamudi RB, Pargaonkar A, Joseph P, Joshy EV, Sivaramakrishnan V. Integrated multi-omics analysis of Huntington disease identifies pathways that modulate protein aggregation. Dis Model Mech 2022; 15:dmm049492. [PMID: 36052548 PMCID: PMC10655815 DOI: 10.1242/dmm.049492] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 08/15/2022] [Indexed: 11/20/2022] Open
Abstract
Huntington disease (HD) is a neurodegenerative disease associated with polyglutamine expansion in the protein huntingtin (HTT). Although the length of the polyglutamine repeat correlates with age at disease onset and severity, psychological, cognitive and behavioral complications point to the existence of disease modifiers. Mitochondrial dysfunction and metabolic deregulation are both associated with the HD but, despite multi-omics characterization of patients and model systems, their mechanisms have remained elusive. Systems analysis of multi-omics data and its validation by using a yeast model could help to elucidate pathways that modulate protein aggregation. Metabolomics analysis of HD patients and of a yeast model of HD was, therefore, carried out. Our analysis showed a considerable overlap of deregulated metabolic pathways. Further, the multi-omics analysis showed deregulated pathways common in human, mice and yeast model systems, and those that are unique to them. The deregulated pathways include metabolic pathways of various amino acids, glutathione metabolism, longevity, autophagy and mitophagy. The addition of certain metabolites as well as gene knockouts targeting the deregulated metabolic and autophagy pathways in the yeast model system showed that these pathways do modulate protein aggregation. Taken together, our results showed that the modulation of deregulated pathways influences protein aggregation in HD, and has implications for progression and prognosis. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Sai S. Pradhan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| | - Sai M. Thota
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| | - Saiswaroop Rajaratnam
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| | - Sai K. S. Bhagavatham
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| | - Sujith K. Pulukool
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| | - Sriram Rathnakumar
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| | - Kanikaram S. Phalguna
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| | - Rajesh B. Dandamudi
- Department of Chemistry, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh 515 134, India
| | - Ashish Pargaonkar
- Application Division, Agilent Technologies Ltd., Bengaluru 560048, India
| | - Prasanth Joseph
- Application Division, Agilent Technologies Ltd., Bengaluru 560048, India
| | - E. V. Joshy
- Department of Neurology, Sri Sathya Sai Institute of Higher Medical Sciences, Whitefield, Bengaluru, Karnataka 560066, India
| | - Venketesh Sivaramakrishnan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| |
Collapse
|
26
|
Mahalakshmi AM, Paneyala S, Ray B, Essa MM, Dehhaghi M, Heng B, Guillemin GJ, Babu Chidambaram S. Alterations in Tryptophan Metabolism Affect Vascular Functions: Connected to Ageing Population Vulnerability to COVID-19 Infection? Int J Tryptophan Res 2022; 15:11786469221083946. [PMID: 35645571 PMCID: PMC9133873 DOI: 10.1177/11786469221083946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 12/11/2021] [Indexed: 11/17/2022] Open
Affiliation(s)
- Arehally M Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, Karnataka, India
| | | | - Bipul Ray
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, Karnataka, India
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman
- Ageing and Dementia Research Group, Sultan Qaboos University, Muscat, Oman
- Visiting Professor, Biomedical Sciences Department, University of Pacific, Sacramento, CA, USA
| | - Mona Dehhaghi
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
- PANDIS.org
| | - Benjamin Heng
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
- PANDIS.org
| | - Gilles J Guillemin
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
- PANDIS.org
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, Karnataka, India
| |
Collapse
|
27
|
Nociceptor-derived Reg3γ prevents endotoxic death by targeting kynurenine pathway in microglia. Cell Rep 2022; 38:110462. [PMID: 35263589 DOI: 10.1016/j.celrep.2022.110462] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 01/11/2022] [Accepted: 02/09/2022] [Indexed: 11/21/2022] Open
Abstract
Nociceptors can fine-tune local or systemic immunity, but the mechanisms of nociceptive modulation in endotoxic death remain largely unknown. Here, we identified C-type lectin Reg3γ as a nociceptor-enriched hormone that protects the host from endotoxic death. During endotoxemia, nociceptor-derived Reg3γ penetrates the brain and suppresses the expression of microglial indoleamine dioxygenase 1, a critical enzyme of the kynurenine pathway, via the Extl3-Bcl10 axis. Endotoxin-administered nociceptor-null mice and nociceptor-specific Reg3γ-deficient mice exhibit a high mortality rate accompanied by decreased brain HK1 phosphorylation and ATP production despite normal peripheral inflammation. Such metabolic arrest is only observed in the brain, and aberrant production of brain quinolinic acid, a neurotoxic metabolite of the kynurenine pathway, causes HK1 suppression. Strikingly, the central administration of Reg3γ protects mice from endotoxic death by enhancing brain ATP production. By identifying nociceptor-derived Reg3γ as a microglia-targeted hormone, this study provides insights into the understanding of tolerance to endotoxic death.
Collapse
|
28
|
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: 6.0] [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
|
29
|
Trézéguet V, Fatrouni H, Merched AJ. Immuno-Metabolic Modulation of Liver Oncogenesis by the Tryptophan Metabolism. Cells 2021; 10:cells10123469. [PMID: 34943977 PMCID: PMC8700200 DOI: 10.3390/cells10123469] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/25/2021] [Accepted: 12/01/2021] [Indexed: 01/06/2023] Open
Abstract
Metabolic rewiring in tumor cells is a major hallmark of oncogenesis. Some of the oncometabolites drive suppressive and tolerogenic signals from the immune system, which becomes complicit to the advent and the survival of neoplasia. Tryptophan (TRP) catabolism through the kynurenine (KYN) pathway was reported to play immunosuppressive actions across many types of cancer. Extensive debate of whether the culprit of immunosuppression was the depletion of TRP or rather KYN accumulation in the tumor microenvironment has been ongoing for years. Results from clinical trials assessing the benefit of inhibiting key limiting enzymes of this pathway such as indoleamine 2,3-dioxygenase (IDO1) or tryptophan 2,3-dioxygenase (TDO2) failed to meet the expectations. Bearing in mind the complexity of the tumoral terrain and the existence of different cancers with IDO1/TDO2 expressing and non-expressing tumoral cells, here we present a comprehensive analysis of the TRP global metabolic hub and the driving potential of the process of oncogenesis with the main focus on liver cancers.
Collapse
|
30
|
Shayesteh S, Guillemin GJ, Rashidian A, Faghir-Ghanesefat H, Mani AR, Tavangar SM, Dehpour AR. 1-Methyl tryptophan, an indoleamine 2,3-dioxygenase inhibitor, attenuates cardiac and hepatic dysfunction in rats with biliary cirrhosis. Eur J Pharmacol 2021; 908:174309. [PMID: 34252442 DOI: 10.1016/j.ejphar.2021.174309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/30/2022]
Abstract
Kynurenine Pathway (KP) is the dominant metabolic route of tryptophan which is catalyzed by indoleamine-2,3-dioxygenase (IDO). This pathway is upregulated in liver disease where the level of KP metabolites correlates with the severity of disease. Cirrhosis is associated with cardiac dysfunction, which manifests itself during severe physiological challenges such as liver transplantation. Cardiac dysfunction in cirrhosis is linked to systemic inflammation and impaired cardiac beta-adrenergic signaling pathways. The KP pathway is involved in modulation of cardiac signaling and is upregulated by systemic inflammation. Therefore, this study aimed to evaluate the effect of IDO inhibition on development of cardiac dysfunction in an experimental model of cirrhosis. Cirrhosis was induced by bile duct ligation (BDL). Experimental groups were given either 1-methyl tryptophan (1-MT, 1, 3, 9 mg/kg), or saline. 28 days after BDL, cardiac chronotropic response to epinephrine was assessed ex vivo. HPLC was employed to measure hepatic and cardiac levels of tryptophan, kynurenine and kynurenic acid. Cirrhosis in rats was associated with impaired cardiac chronotropic responsiveness to adrenergic stimulation. 1-MT dose-dependently improved cirrhosis-induced chronotropic dysfunction as well as elevated serum levels of CRP and IL-6 in BDL rats. Hepatic and cardiac kynurenine/tryptophan ratio were elevated in cirrhotic rats and were reduced following 1-MT administration. Chronic administration of 1-MT could also reduce hepatic inflammation, fibrosis and ductular proliferation. 1-MT attenuates cardiac dysfunction in rats with biliary cirrhosis. This protective effect is not limited to the cardiac function as liver histopathologic changes were also improved following chronic 1-MT administration.
Collapse
Affiliation(s)
- Sevda Shayesteh
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Gilles J Guillemin
- Neuroinflammation Group, Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Amir Rashidian
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hedyeh Faghir-Ghanesefat
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali R Mani
- Division of Medicine, University College London, London, UK
| | - Seyed Mohammad Tavangar
- Department of Pathology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad R Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Leclercq S, Schwarz M, Delzenne NM, Stärkel P, de Timary P. Alterations of kynurenine pathway in alcohol use disorder and abstinence: a link with gut microbiota, peripheral inflammation and psychological symptoms. Transl Psychiatry 2021; 11:503. [PMID: 34599147 PMCID: PMC8486842 DOI: 10.1038/s41398-021-01610-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/11/2021] [Accepted: 08/20/2021] [Indexed: 02/06/2023] Open
Abstract
The gut-brain communication is mostly driven by the immune, metabolic and neural pathways which remained poorly explored in patients with alcohol use disorder (AUD). The metabolites arising from the tryptophan-kynurenine pathway have gained considerable attention since they are at the interface between intestinal bacteria, host immune response and brain functions. This study described the circulating levels of kynurenine metabolites in AUD patients, at the onset (T1) and end (T2) of a 3-week detoxification program, and tested correlations between those metabolites and inflammatory markers, the gut microbiota and the psychological symptoms. Increased concentration of the neurotoxic metabolite quinolinic acid (QUIN) and decreased levels of the neuroprotector metabolite kynurenic acid (KYNA) which both modulate glutamatergic neurotransmission were observed in AUD patients, particularly at T2. The inflammatory marker hsCRP was associated with several metabolic ratios of the kynurenine pathway. Tryptophan, KYNA and QUIN were correlated with depression, alcohol craving and reaction time, respectively. Analysis of gut microbiota revealed that bacteria known as short-chain fatty acid producers, as well as bacterial metabolites including butyrate and medium-chain fatty acids were associated with some metabolites of the tryptophan-kynurenine pathway. Targeting the glutamatergic neurotransmission through the modulation of the kynurenine pathway, by manipulating the gut microbiota, might represent an interesting alternative for modulating alcohol-related behavior.
Collapse
Affiliation(s)
- Sophie Leclercq
- grid.7942.80000 0001 2294 713XInstitute of Neuroscience, Université catholique de Louvain (UCLouvain), Brussels, Belgium ,grid.7942.80000 0001 2294 713XMetabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Markus Schwarz
- grid.411095.80000 0004 0477 2585Institute of Laboratory Medicine, LMU Klinikum Munich, Munich, Germany
| | - Nathalie M. Delzenne
- grid.7942.80000 0001 2294 713XMetabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Peter Stärkel
- grid.7942.80000 0001 2294 713XLaboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université catholique de Louvain (UCLouvain), Brussels, Belgium ,grid.48769.340000 0004 0461 6320Department of Hepatogastroenterology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Philippe de Timary
- Institute of Neuroscience, Université catholique de Louvain (UCLouvain), Brussels, Belgium. .,Department of Adult Psychiatry, Cliniques universitaires Saint-Luc, Brussels, Belgium.
| |
Collapse
|
32
|
Teruya T, Chen YJ, Kondoh H, Fukuji Y, Yanagida M. Whole-blood metabolomics of dementia patients reveal classes of disease-linked metabolites. Proc Natl Acad Sci U S A 2021; 118:e2022857118. [PMID: 34493657 PMCID: PMC8449400 DOI: 10.1073/pnas.2022857118] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 08/08/2021] [Indexed: 01/03/2023] Open
Abstract
Dementia is caused by factors that damage neurons. We quantified small molecular markers in whole blood of dementia patients, using nontargeted liquid chromatography-mass spectroscopy (LC-MS). Thirty-three metabolites, classified into five groups (A to E), differed significantly in dementia patients, compared with healthy elderly subjects. Seven A metabolites present in plasma, including quinolinic acid, kynurenine, and indoxyl-sulfate, increased. Possibly they act as neurotoxins in the central nervous system (CNS). The remaining 26 compounds (B to E) decreased, possibly causing a loss of support or protection of the brain in dementia. Six B metabolites, normally enriched in red blood cells (RBCs), all contain trimethylated ammonium moieties. These metabolites include ergothioneine and structurally related compounds that have scarcely been investigated as dementia markers, validating the examination of RBC metabolites. Ergothioneine, a potent antioxidant, is significantly decreased in various cognition-related disorders, such as mild cognitive impairment and frailty. C compounds also include some oxidoreductants and are normally abundant in RBCs (NADP+, glutathione, adenosine triphosphate, pantothenate, S-adenosyl-methionine, and gluconate). Their decreased levels in dementia patients may also contribute to depressed brain function. Twelve D metabolites contains plasma compounds, such as amino acids, glycerophosphocholine, dodecanoyl-carnitine, and 2-hydroxybutyrate, which normally protect the brain, but their diminution in dementia may reduce that protection. Seven D compounds have been identified previously as dementia markers. B to E compounds may be critical to maintain the CNS by acting directly or indirectly. How RBC metabolites act in the CNS and why they diminish significantly in dementia remain to be determined.
Collapse
Affiliation(s)
- Takayuki Teruya
- G0 Cell Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan
| | - Yung-Ju Chen
- G0 Cell Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan
| | - Hiroshi Kondoh
- Geriatric Unit, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Yasuhide Fukuji
- National Hospital Organization Ryukyu Hospital, Okinawa 904-1201, Japan
| | - Mitsuhiro Yanagida
- G0 Cell Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan;
| |
Collapse
|
33
|
Limana da Silveira T, Lopes Machado M, Bicca Obetine Baptista F, Farina Gonçalves D, Duarte Hartmann D, Marafiga Cordeiro L, Franzen da Silva A, Lenz Dalla Corte C, Aschner M, Antunes Soares FA. Caenorhabditis elegans as a model for studies on quinolinic acid-induced NMDAR-dependent glutamatergic disorders. Brain Res Bull 2021; 175:90-98. [PMID: 34271120 DOI: 10.1016/j.brainresbull.2021.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 12/21/2022]
Abstract
Quinolinic acid (QUIN) is an agonist of the neurotransmitter glutamate (Glu) capable of binding to N-methyl-D-aspartate receptors (NMDAR) increasing glutamatergic signaling. QUIN is known for being an endogenous neurotoxin, able to induce neurodegeneration. In Caenorhabditis elegans, the mechanism by which QUIN induces behavioral and metabolic toxicity has not been fully elucidated. The effects of QUIN on behavioral and metabolic parameters in nmr-1 and nmr-2 NMDA receptors in transgenic and wild-type (WT) worms were performed to decipher the pathway by which QUIN exerts its toxicity. QUIN increased locomotion parameters such as wavelength and movement amplitude medium, as well as speed and displacement, without modifying the number of body bends in an NMDAR-dependent-manner. QUIN increased the response time to the chemical stimulant 1-octanol, which is modulated by glutamatergic neurotransmission in the ASH neuron. Brood size increased after exposure to QUIN, dependent upon nmr-2/NMDA-receptor, with no change in lifespan. Oxygen consumption, mitochondrial membrane potential, and the flow of coupled and unbound electrons to ATP production were reduced by QUIN in wild-type animals, but did not alter citrate synthase activity, altering the functionality but the mitochondrial viability. Notably, QUIN modified fine locomotor and chemosensory behavioral parameters, as well as metabolic parameters, analogous to previously reported effects in mammals. Our results indicate that QUIN can be used as a neurotoxin to elicit glutamatergic dysfunction in C. elegans in a way analogous to other animal models.
Collapse
Affiliation(s)
- Tássia Limana da Silveira
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Marina Lopes Machado
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Fabiane Bicca Obetine Baptista
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Débora Farina Gonçalves
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Diane Duarte Hartmann
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil; Universidade Regional do Noroeste do Estado do Rio Grande do Sul
| | - Larissa Marafiga Cordeiro
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Aline Franzen da Silva
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Cristiane Lenz Dalla Corte
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University, Forchheimer 209, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Felix Alexandre Antunes Soares
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil.
| |
Collapse
|
34
|
Wangpaichitr M, Theodoropoulos G, Nguyen DJM, Wu C, Spector SA, Feun LG, Savaraj N. Cisplatin Resistance and Redox-Metabolic Vulnerability: A Second Alteration. Int J Mol Sci 2021; 22:7379. [PMID: 34298999 PMCID: PMC8304747 DOI: 10.3390/ijms22147379] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/01/2021] [Accepted: 07/05/2021] [Indexed: 01/17/2023] Open
Abstract
The development of drug resistance in tumors is a major obstacle to effective cancer chemotherapy and represents one of the most significant complications to improving long-term patient outcomes. Despite early positive responsiveness to platinum-based chemotherapy, the majority of lung cancer patients develop resistance. The development of a new combination therapy targeting cisplatin-resistant (CR) tumors may mark a major improvement as salvage therapy in these patients. The recent resurgence in research into cellular metabolism has again confirmed that cancer cells utilize aerobic glycolysis ("the Warburg effect") to produce energy. Hence, this observation still remains a characteristic hallmark of altered metabolism in certain cancer cells. However, recent evidence promotes another concept wherein some tumors that acquire resistance to cisplatin undergo further metabolic alterations that increase tumor reliance on oxidative metabolism (OXMET) instead of glycolysis. Our review focuses on molecular changes that occur in tumors due to the relationship between metabolic demands and the importance of NAD+ in redox (ROS) metabolism and the crosstalk between PARP-1 (Poly (ADP ribose) polymerase-1) and SIRTs (sirtuins) in CR tumors. Finally, we discuss a role for the tumor metabolites of the kynurenine pathway (tryptophan catabolism) as effectors of immune cells in the tumor microenvironment during acquisition of resistance in CR cells. Understanding these concepts will form the basis for future targeting of CR cells by exploiting redox-metabolic changes and their consequences on immune cells in the tumor microenvironment as a new approach to improve overall therapeutic outcomes and survival in patients who fail cisplatin.
Collapse
Affiliation(s)
- Medhi Wangpaichitr
- Department of Veterans Affairs, Miami VA Healthcare System, Research Service (151), Miami, FL 33125, USA; (G.T.); (D.J.M.N.); (C.W.); (S.A.S.)
- Department of Surgery, Cardiothoracic Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - George Theodoropoulos
- Department of Veterans Affairs, Miami VA Healthcare System, Research Service (151), Miami, FL 33125, USA; (G.T.); (D.J.M.N.); (C.W.); (S.A.S.)
| | - Dan J. M. Nguyen
- Department of Veterans Affairs, Miami VA Healthcare System, Research Service (151), Miami, FL 33125, USA; (G.T.); (D.J.M.N.); (C.W.); (S.A.S.)
| | - Chunjing Wu
- Department of Veterans Affairs, Miami VA Healthcare System, Research Service (151), Miami, FL 33125, USA; (G.T.); (D.J.M.N.); (C.W.); (S.A.S.)
| | - Sydney A. Spector
- Department of Veterans Affairs, Miami VA Healthcare System, Research Service (151), Miami, FL 33125, USA; (G.T.); (D.J.M.N.); (C.W.); (S.A.S.)
| | - Lynn G. Feun
- Department of Medicine, Hematology/Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (L.G.F.); (N.S.)
| | - Niramol Savaraj
- Department of Medicine, Hematology/Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (L.G.F.); (N.S.)
- Department of Veterans Affairs, Miami VA Healthcare System, Hematology/Oncology, 1201 NW 16 Street, Room D1010, Miami, FL 33125, USA
| |
Collapse
|
35
|
Kassab SE, Mowafy S. Structural Basis of Selective Human Indoleamine-2,3-dioxygenase 1 (hIDO1) Inhibition. ChemMedChem 2021; 16:3149-3164. [PMID: 34174026 DOI: 10.1002/cmdc.202100253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/23/2021] [Indexed: 11/08/2022]
Abstract
hIDO1 is a heme-dioxygenase overexpressed in the tumor microenvironment and is implicated in the survival of cancer cells. Metabolism of tryptophan to N-formyl-kynurenine by hIDO1 leads to immune suppression to result in cancer cell immune escape. In this article, we discuss the discovery of selective hIDO1 inhibitors for therapeutic intervention that have been promoted to clinical trials and for which crystallographic structural information is available for the respective inhibitor-enzyme complex. The structural insights are based on the complex crystal structures and the relative biological data profiles. The structural basis of selective hIDO1 inhibition, as discussed herein, opens new avenues to the discovery of novel inhibitors with improved activity profiles, selectivity, and distinct structure frameworks.
Collapse
Affiliation(s)
- Shaymaa Emam Kassab
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, El-Buhaira, 22516, Egypt
| | - Samar Mowafy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Misr International University, Cairo, 11431, Egypt.,Department of Chemistry, University of Washington, Seattle, Washington, 98195, United States of America
| |
Collapse
|
36
|
Neuroprotection in Glaucoma: NAD +/NADH Redox State as a Potential Biomarker and Therapeutic Target. Cells 2021; 10:cells10061402. [PMID: 34198948 PMCID: PMC8226607 DOI: 10.3390/cells10061402] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023] Open
Abstract
Glaucoma is the leading cause of irreversible blindness worldwide. Its prevalence and incidence increase exponentially with age and the level of intraocular pressure (IOP). IOP reduction is currently the only therapeutic modality shown to slow glaucoma progression. However, patients still lose vision despite best treatment, suggesting that other factors confer susceptibility. Several studies indicate that mitochondrial function may underlie both susceptibility and resistance to developing glaucoma. Mitochondria meet high energy demand, in the form of ATP, that is required for the maintenance of optimum retinal ganglion cell (RGC) function. Reduced nicotinamide adenine dinucleotide (NAD+) levels have been closely correlated to mitochondrial dysfunction and have been implicated in several neurodegenerative diseases including glaucoma. NAD+ is at the centre of various metabolic reactions culminating in ATP production—essential for RGC function. In this review we present various pathways that influence the NAD+(H) redox state, affecting mitochondrial function and making RGCs susceptible to degeneration. Such disruptions of the NAD+(H) redox state are generalised and not solely induced in RGCs because of high IOP. This places the NAD+(H) redox state as a potential systemic biomarker for glaucoma susceptibility and progression; a hypothesis which may be tested in clinical trials and then translated to clinical practice.
Collapse
|
37
|
Cathomas F, Guetter K, Seifritz E, Klaus F, Kaiser S. Quinolinic acid is associated with cognitive deficits in schizophrenia but not major depressive disorder. Sci Rep 2021; 11:9992. [PMID: 33976271 PMCID: PMC8113521 DOI: 10.1038/s41598-021-89335-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 04/20/2021] [Indexed: 01/16/2023] Open
Abstract
Tryptophan and its catabolites (TRYCATs) have been suggested to link peripheral immune system activation and central neurotransmitter abnormalities with relevance to the etio-pathophysiology of schizophrenia (SZ) and major depressive disorder (MDD). The relationship to different psychopathological dimensions within these disorders however remains to be elucidated. We thus investigated potential group differences of tryptophan, kynurenine, kynurenic acid, 3-hydroxy kynurenine and quinolinic acid in the plasma of 19 healthy controls (HC), 45 patients with SZ and 43 patients with MDD and correlated plasma proteins with the "motivation and pleasure" dimension and cognition. After correcting for the covariates age, sex, body mass index, smoking and medication, patients with MDD showed lower kynurenine and 3-hydroxy kynurenine levels compared to HC. Quinolinic acid correlated negatively with composite cognitive score in patients with SZ, indicating that more severe cognitive impairments were associated with increased plasma levels of quinolinic acid. No correlations were found in patients with MDD. These results indicate that MDD and SZ are associated with dysregulation of the kynurenine pathway. Quinolinic acid might be specifically implicated in the pathophysiology of cognitive deficits in patients with SZ. Further studies are needed to determine whether TRYCATs are causally involved in the etiology of these neuropsychiatric disorders.
Collapse
Affiliation(s)
- Flurin Cathomas
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Lenggstrasse 31, 8032, Zürich, Switzerland. .,Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, Mount Sinai, NY, USA.
| | - Karoline Guetter
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Lenggstrasse 31, 8032, Zürich, Switzerland
| | - Erich Seifritz
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Lenggstrasse 31, 8032, Zürich, Switzerland.,Neuroscience Center Zurich, ETH Zurich and University of Zurich, 8057, Zurich, Switzerland
| | - Federica Klaus
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Lenggstrasse 31, 8032, Zürich, Switzerland.,Department of Psychiatry, University of California San Diego, San Diego, USA
| | - Stefan Kaiser
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Chemin du Petit-Bel-Air, 1225, Chêne-Bourg, Switzerland
| |
Collapse
|
38
|
Quan W, Jiao Y, Li Y, Xue C, Liu G, Wang Z, Qin F, He Z, Zeng M, Chen J. Metabolic changes from exposure to harmful Maillard reaction products and high-fat diet on Sprague-Dawley rats. Food Res Int 2021; 141:110129. [DOI: 10.1016/j.foodres.2021.110129] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 12/17/2020] [Accepted: 01/06/2021] [Indexed: 12/11/2022]
|
39
|
Quan W, Jiao Y, Xue C, Li Y, Liu G, He Z, Qin F, Zeng M, Chen J. The Effect of Exogenous Free Nε-(Carboxymethyl)Lysine on Diabetic-Model Goto-Kakizaki Rats: Metabolomics Analysis in Serum and Urine. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:783-793. [PMID: 33401897 DOI: 10.1021/acs.jafc.0c06445] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The current study investigated the effects of exogenous free Nε-(carboxymethyl) lysine (CML) from daily diet on diabetic-model Goto-Kakizaki rats. Rats were fed with free CML (2 mg/kg body weight) for 8 weeks, then metabolomics evaluation was performed on serum and urine, and biochemical and histopathologic examinations were conducted to verify metabolic results. Diabetic rats fed with free CML showed significantly increased (P < 0.05) fasting blood glucose (11.1 ± 1.07 mmol/L) and homeostasis model assessment values (homeostatic model assessment of insulin resistance: 16.0 ± 4.24; homeostatic model assessment of beta cell function: 6.66 ± 2.01; and modified beta cell function index: 11.5 ± 2.66) and a significantly altered (P < 0.05) oxidative stress level when compared to the control group. Serum and urine metabolomics showed a significantly altered (P < 0.05) level of aminomalonic acid, 2-oxoadipic acid, l-malic acid, β-alanine, 2-oxoglutaric acid, d-threitol, N-acetyl-leucine, methylmalonic acid, l-cysteine, thymine, glycine, l-alanine, 4-hydroxyproline, hexadecane, succinic acid, l-ornithine, gluconolactone, maleic acid, l-lactate, tryptophan, 5-methoxyindoleacetate, γ-aminobutyric acid, homoserine, maltose, and quinolinic acid. Our results indicated that these metabolites altered by exposure to exogenous free CML were mapped to the citric acid cycle and amino acid and carbohydrate metabolism, which might be related to increased progression of diabetes and some other diabetic complications, including diabetic brain and neurological diseases, retinopathy, nephropathy, and impaired wound healing.
Collapse
Affiliation(s)
- Wei Quan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ye Jiao
- School of Chemistry and Food Engineering, Changsha University of Science & Technology, Changsha 410114, China
| | - Chaoyi Xue
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yong Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Guoping Liu
- Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Zhiyong He
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Fang Qin
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Maomao Zeng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jie Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
40
|
Whiley L, Chappell KE, D'Hondt E, Lewis MR, Jiménez B, Snowden SG, Soininen H, Kłoszewska I, Mecocci P, Tsolaki M, Vellas B, Swann JR, Hye A, Lovestone S, Legido-Quigley C, Holmes E. Metabolic phenotyping reveals a reduction in the bioavailability of serotonin and kynurenine pathway metabolites in both the urine and serum of individuals living with Alzheimer's disease. Alzheimers Res Ther 2021; 13:20. [PMID: 33422142 PMCID: PMC7797094 DOI: 10.1186/s13195-020-00741-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 12/07/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Both serotonergic signalling disruption and systemic inflammation have been associated with the pathogenesis of Alzheimer's disease (AD). The common denominator linking the two is the catabolism of the essential amino acid, tryptophan. Metabolism via tryptophan hydroxylase results in serotonin synthesis, whilst metabolism via indoleamine 2,3-dioxygenase (IDO) results in kynurenine and its downstream derivatives. IDO is reported to be activated in times of host systemic inflammation and therefore is thought to influence both pathways. To investigate metabolic alterations in AD, a large-scale metabolic phenotyping study was conducted on both urine and serum samples collected from a multi-centre clinical cohort, consisting of individuals clinically diagnosed with AD, mild cognitive impairment (MCI) and age-matched controls. METHODS Metabolic phenotyping was applied to both urine (n = 560) and serum (n = 354) from the European-wide AddNeuroMed/Dementia Case Register (DCR) biobank repositories. Metabolite data were subsequently interrogated for inter-group differences; influence of gender and age; comparisons between two subgroups of MCI - versus those who remained cognitively stable at follow-up visits (sMCI); and those who underwent further cognitive decline (cMCI); and the impact of selective serotonin reuptake inhibitor (SSRI) medication on metabolite concentrations. RESULTS Results revealed significantly lower metabolite concentrations of tryptophan pathway metabolites in the AD group: serotonin (urine, serum), 5-hydroxyindoleacetic acid (urine), kynurenine (serum), kynurenic acid (urine), tryptophan (urine, serum), xanthurenic acid (urine, serum), and kynurenine/tryptophan ratio (urine). For each listed metabolite, a decreasing trend in concentrations was observed in-line with clinical diagnosis: control > MCI > AD. There were no significant differences in the two MCI subgroups whilst SSRI medication status influenced observations in serum, but not urine. CONCLUSIONS Urine and serum serotonin concentrations were found to be significantly lower in AD compared with controls, suggesting the bioavailability of the neurotransmitter may be altered in the disease. A significant increase in the kynurenine/tryptophan ratio suggests that this may be a result of a shift to the kynurenine metabolic route due to increased IDO activity, potentially as a result of systemic inflammation. Modulation of the pathways could help improve serotonin bioavailability and signalling in AD patients.
Collapse
Affiliation(s)
- Luke Whiley
- UK Dementia Research Institute, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
- Health Futures Institute, Murdoch University, Perth, WA, 6105, Australia
- The Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia
| | - Katie E Chappell
- Section of Bioanalytical Chemistry W12 0NN, UK, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
- National Phenome Centre, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Ellie D'Hondt
- imec, Exascience Life Lab, Kapeldreef 75, B-3001, Leuven, Belgium
| | - Matthew R Lewis
- Section of Bioanalytical Chemistry W12 0NN, UK, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
- National Phenome Centre, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Beatriz Jiménez
- National Phenome Centre, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Stuart G Snowden
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Present address: Core Metabolomics and Lipidomics Laboratory, Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Hilkka Soininen
- Department of Neurology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | | | - Patrizia Mecocci
- Institute of Gerontology and Geriatrics, University of Perugia, Perugia, Italy
| | - Magda Tsolaki
- 3rd Department of Neurology, Aristotle University, Thessaloniki, Greece
| | - Bruno Vellas
- INSERM U 558, University of Toulouse, Toulouse, France
| | - Jonathan R Swann
- Section of Bioanalytical Chemistry W12 0NN, UK, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Abdul Hye
- INSERM U 558, University of Toulouse, Toulouse, France
| | - Simon Lovestone
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK
- Current affiliation at Janssen-Cilag Ltd, High Wycombe, UK
| | - Cristina Legido-Quigley
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Elaine Holmes
- UK Dementia Research Institute, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK.
- Health Futures Institute, Murdoch University, Perth, WA, 6105, Australia.
- The Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia.
- Section for Nutrition Research, Imperial College, Hammersmith Campus Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
41
|
Cattelan Souza L, de Brito MLO, Jesse CR, Boeira SP, de Gomes MG, Goes ATR, Fabbro LD, Machado FR, Prigol M, Nogueira CW. Involvement of kynurenine pathway in depressive-like behaviour induced by nandrolone decanoate in mice. Steroids 2020; 164:108727. [PMID: 32891681 DOI: 10.1016/j.steroids.2020.108727] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 08/24/2020] [Accepted: 08/30/2020] [Indexed: 11/17/2022]
Abstract
Nandrolone decanoate (ND) belongs to the class II of anabolic-androgenic steroids (AAS), which is composed of 19-nor-testosterone-derivatives. AAS represent a group of synthetic testosterone that is used in clinical treatment. However, these drugs are widely abused among individuals as a means of promoting muscle growth or enhancing athletic performance. AAS in general and ND in particular have been associated with several behavioral disturbances, such as anxiety, aggressiveness and depression. A factor that contributes to the development of depression is the brain activation of indoleamine 2,3-dioxygenase (IDO), the rate-limiting enzyme of kynurenine pathway (KP). In the present study, we examined the involvement of KP in depressive phenotype induced by a ND treatment (10 mg/kg/day/s.c., for 28 days) that mimics human abuse system (e.g. supraphysiological doses) in C57B/6J mice. Our results showed that ND caused depressive like-behavior in the tail suspension test and anhedonic-like state measured in the sucrose preference test. ND administration decreased the levels of brain-derived neurotrophic factor and neurotrophin-3 and reduced Na+,K+-ATPase activity in the hippocampus, striatum and prefrontal cortex. We also found that ND elicited KP activation, as reflected by the increase of IDO activity and kynurenine levels in these brain regions. Moreover, ND decreased serotonin levels and increased 5-hydroxyindoleacetic acid levels in the brain. Treatment with IDO inhibitor 1-methyl-dl-trypthophan (1 mg/kg/i.p.) reversed the behavioral and neurochemical alterations induced by ND. These results indicate for the first time that KP plays a key role in depressive-like behavior and neurotoxicity induced by supraphysiologicaldoses of ND in mice.
Collapse
Affiliation(s)
- Leandro Cattelan Souza
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, CEP 97650-000, Itaqui, RS, Brazil; Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil.
| | - Maicon Lenon Otenio de Brito
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, CEP 97650-000, Itaqui, RS, Brazil
| | - Cristiano Ricardo Jesse
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, CEP 97650-000, Itaqui, RS, Brazil
| | - Silvana Peterini Boeira
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, CEP 97650-000, Itaqui, RS, Brazil
| | - Marcelo Gomes de Gomes
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, CEP 97650-000, Itaqui, RS, Brazil
| | - André Tiago Rossito Goes
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, CEP 97650-000, Itaqui, RS, Brazil
| | - Lucian Del Fabbro
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, CEP 97650-000, Itaqui, RS, Brazil
| | - Franciele Romero Machado
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, CEP 97650-000, Itaqui, RS, Brazil
| | - Marina Prigol
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, CEP 97650-000, Itaqui, RS, Brazil
| | - Cristina Wayne Nogueira
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| |
Collapse
|
42
|
Skonieczna-Żydecka K, Jakubczyk K, Maciejewska-Markiewicz D, Janda K, Kaźmierczak-Siedlecka K, Kaczmarczyk M, Łoniewski I, Marlicz W. Gut Biofactory-Neurocompetent Metabolites within the Gastrointestinal Tract. A Scoping Review. Nutrients 2020; 12:E3369. [PMID: 33139656 PMCID: PMC7693392 DOI: 10.3390/nu12113369] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota have gained much scientific attention recently. Apart from unravelling the taxonomic data, we should understand how the altered microbiota structure corresponds to functions of this complex ecosystem. The metabolites of intestinal microorganisms, especially bacteria, exert pleiotropic effects on the human organism and contribute to the host systemic balance. These molecules play key roles in regulating immune and metabolic processes. A subset of them affect the gut brain axis signaling and balance the mental wellbeing. Neurotransmitters, short chain fatty acids, tryptophan catabolites, bile acids and phosphatidylcholine, choline, serotonin, and L-carnitine metabolites possess high neuroactive potential. A scoping literature search in PubMed/Embase was conducted up until 20 June 2020, using three major search terms "microbiota metabolites" AND "gut brain axis" AND "mental health". This review aimed to enhance our knowledge regarding the gut microbiota functional capacity, and support current and future attempts to create new compounds for future clinical interventions.
Collapse
Affiliation(s)
- Karolina Skonieczna-Żydecka
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (K.S.-Ż.); (K.J.); (D.M.-M.); (K.J.)
| | - Karolina Jakubczyk
- Department of Surgical Oncology, Medical University of Gdansk, Smoluchowskiego 17, 80-214 Gdańsk, Poland;
| | - Dominika Maciejewska-Markiewicz
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (K.S.-Ż.); (K.J.); (D.M.-M.); (K.J.)
| | - Katarzyna Janda
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (K.S.-Ż.); (K.J.); (D.M.-M.); (K.J.)
| | | | - Mariusz Kaczmarczyk
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland;
| | - Igor Łoniewski
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (K.S.-Ż.); (K.J.); (D.M.-M.); (K.J.)
| | - Wojciech Marlicz
- Department of Gastroenterology, Pomeranian Medical University, 71-252 Szczecin, Poland
- The Centre for Digestive Diseases Endoklinika, 70-535 Szczecin, Poland
| |
Collapse
|
43
|
Wang H, Shen Y, Chuang H, Chiu C, Ye Y, Zhao L. Neuroinflammation in Alzheimer's Disease: Microglia, Molecular Participants and Therapeutic Choices. Curr Alzheimer Res 2020; 16:659-674. [PMID: 31580243 DOI: 10.2174/1567205016666190503151648] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 03/21/2019] [Accepted: 04/30/2019] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease is the world's most common dementing illness. It is pathologically characterized by β-amyloid accumulation, extracellular senile plaques and intracellular neurofibrillary tangles formation, and neuronal necrosis and apoptosis. Neuroinflammation has been widely recognized as a crucial process that participates in AD pathogenesis. In this review, we briefly summarized the involvement of microglia in the neuroinflammatory process of Alzheimer's disease. Its roles in the AD onset and progression are also discussed. Numerous molecules, including interleukins, tumor necrosis factor alpha, chemokines, inflammasomes, participate in the complex process of AD-related neuroinflammation and they are selectively discussed in this review. In the end of this paper from an inflammation- related perspective, we discussed some potential therapeutic choices.
Collapse
Affiliation(s)
- Haijun Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yin Shen
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Haoyu Chuang
- Department of Neurosurgery, Tainan Municipal An-Nan Hospital, Tainan, Taiwan.,Department of Neurosurgery, China Medical University Bei-Gang Hospital, Yun-Lin, Taiwan.,School of Medicine, China Medical University, Taichung, Taiwan
| | - Chengdi Chiu
- School of Medicine, China Medical University, Taichung, Taiwan.,Department of Neurosurgery, China Medical University Hospital, Taichung, Taiwan
| | - Youfan Ye
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lei Zhao
- Department of Infectious Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
44
|
Braidy N, Alicajic H, Pow D, Smith J, Jugder BE, Brew BJ, Nicolazzo JA, Guillemin GJ. Potential Mechanism of Cellular Uptake of the Excitotoxin Quinolinic Acid in Primary Human Neurons. Mol Neurobiol 2020; 58:34-54. [PMID: 32894500 DOI: 10.1007/s12035-020-02046-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/28/2020] [Indexed: 01/18/2023]
Abstract
In Alzheimer's disease (AD), excessive amounts of quinolinic acid (QUIN) accumulate within the brain parenchyma and dystrophic neurons. QUIN also regulates glutamate uptake into neurons, which may be due to modulation of Na+-dependent excitatory amino acid transporters (EAATs). To determine the biological relationships between QUIN and glutamate dysfunction, we first quantified the functionality and kinetics of [3H]QUIN uptake in primary human neurons using liquid scintillation. We then measured changes in the protein expression of the glutamate transporter EAAT3 and EAAT1b in primary neurons treated with QUIN and the EAAT inhibitor L-trans-pyrrolidine-2,4-dicarboxylic acid (2,4-PDC) using western blotting and immunohistochemistry. Immunohistochemistry was further used to elucidate intracellular transport of exogenous QUIN and the lysosomal-associated membrane protein 2 (LAMP2). Structural insights into the binding between QUIN and EAAT3 were further investigated using molecular docking techniques. We report significant temperature-dependent high-affinity transport leading to neuronal uptake of [3H]QUIN with a Km of 42.2 μM, and a Vmax of 9.492 pmol/2 min/mg protein, comparable with the uptake of glutamate. We also found that QUIN increases expression of the EAAT3 monomer while decreasing the functional trimer. QUIN uptake into primary neurons was shown to involve EAAT3 as uptake was significantly attenuated following EAAT inhibition. We also demonstrated that QUIN increases the expression of aberrant EAAT1b protein in neurons further implicating QUIN-induced glutamate dysfunction. Furthermore, we demonstrated that QUIN is metabolised exclusively in lysosomes. The involvement of EAAT3 as a modulator for QUIN uptake was further confirmed using molecular docking. This study is the first to characterise a mechanism for QUIN uptake into primary human neurons involving EAAT3, opening potential targets to attenuate QUIN-induced excitotoxicity in neuroinflammatory diseases.
Collapse
Affiliation(s)
- Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia.
- School of Medicine, Huzhou University, Wuxing District, Huzhou, Zhejiang, China.
| | - Hayden Alicajic
- Neuropharmacology group, MND and Neurodegenerative diseases Research Centre, Macquarie University, Sydney, NSW, 2019, Australia
| | - David Pow
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Jason Smith
- Department of Chemistry and Biomolecular sciences, Macquarie University, Sydney, NSW, Australia
| | - Bat-Erdene Jugder
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Bruce J Brew
- St Vincent's Centre for Applied Medical Research, Sydney, Australia
- Department of Neurology and HIV Medicine, St Vincent's Hospital, Sydney, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Gilles J Guillemin
- Neuropharmacology group, MND and Neurodegenerative diseases Research Centre, Macquarie University, Sydney, NSW, 2019, Australia.
| |
Collapse
|
45
|
Geng J, Weitz AC, Dornevil K, Hendrich MP, Liu A. Kinetic and Spectroscopic Characterization of the Catalytic Ternary Complex of Tryptophan 2,3-Dioxygenase. Biochemistry 2020; 59:2813-2822. [PMID: 32659080 DOI: 10.1021/acs.biochem.0c00179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The first step of the kynurenine pathway for l-tryptophan (l-Trp) degradation is catalyzed by heme-dependent dioxygenases, tryptophan 2,3-dioxygenase (TDO) and indoleamine 2,3-dioxygenase. In this work, we employed stopped-flow optical absorption spectroscopy to study the kinetic behavior of the Michaelis complex of Cupriavidus metallidurans TDO (cmTDO) to improve our understanding of oxygen activation and initial oxidation of l-Trp. On the basis of the stopped-flow results, rapid freeze-quench (RFQ) experiments were performed to capture and characterize this intermediate by Mössbauer spectroscopy. By incorporating the chlorite dismutase-chlorite system to produce high concentrations of solubilized O2, we were able to capture the Michaelis complex of cmTDO in a nearly quantitative yield. The RFQ-Mössbauer results confirmed the identity of the Michaelis complex as an O2-bound ferrous species. They revealed remarkable similarities between the electronic properties of the Michaelis complex and those of the O2 adduct of myoglobin. We also found that the decay of this reactive intermediate is the rate-limiting step of the catalytic reaction. An inverse α-secondary substrate kinetic isotope effect was observed with a kH/kD of 0.87 ± 0.03 when (indole-d5)-l-Trp was employed as the substrate. This work provides an important piece of spectroscopic evidence of the chemical identity of the Michaelis complex of bacterial TDO.
Collapse
Affiliation(s)
- Jiafeng Geng
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Andrew C Weitz
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Kednerlin Dornevil
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States.,Department of Chemistry, University of Texas at San Antonio, San Antonio, Texas 78249, United States
| | - Michael P Hendrich
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Aimin Liu
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States.,Department of Chemistry, University of Texas at San Antonio, San Antonio, Texas 78249, United States
| |
Collapse
|
46
|
Marson GV, de Castro RJS, Belleville MP, Hubinger MD. Spent brewer's yeast as a source of high added value molecules: a systematic review on its characteristics, processing and potential applications. World J Microbiol Biotechnol 2020; 36:95. [PMID: 32583032 DOI: 10.1007/s11274-020-02866-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/08/2020] [Indexed: 01/10/2023]
Abstract
Development of new strategies to add-value to agro-industrial by-products are of environmental and economical importance. Innovative and low-cost sources of protein and bioactive peptides have been explored worldwide. Spent brewer's yeast (SBY) is the second most relevant by-product from the brewing industry, and despite its nutritional (about 50% protein, dry weight) and technological potential, it is still underused or needs to be disposed of. SBY cells need to be disrupted to release intracellular and cell wall proteins. This procedure has been performed using autolysis, glass bead milling, enzymatic hydrolysis and ultrasound processing. Enzymatic treatment is usually performed without prior purification and is a challenging process, which involves multiple factors, but has been successfully used as a strategy to add value to agro-industrial by-products. Scope and approach: in this review, we particularly focused on enzymatic hydrolysis as a strategy to promote SBY valorisation, illustrating the state-of-the-art processes used to produce protein extracts from this material as well as exploring fundamental concepts related to the particularities of yeast cell disruption and protein hydrolysis. Furthermore, innovative applications of value-added yeast by-products in food, biotechnological and pharmaceutical industries are presented and discussed. Key findings and conclusions: the discovery of valuable compounds found in spent yeasts as well as the development of new processing methodologies have been widening the possibilities of reuse and transformation of SBY as an ingredient and innovative matrix. Once released, yeast proteins and peptides may be applied as an innovative non-animal protein source or a functional and bioactive ingredient.
Collapse
Affiliation(s)
- Gabriela Vollet Marson
- Institut Européen des Membranes, Université de Montpellier, CNRS, ENSCM, UM, CC 047, 2 Place Eugène Bataillon, 34095, Montpellier, France. .,Laboratory of Process Engineering, Department of Food Engineering, School of Food Engineering, UNICAMP, Rua Monteiro Lobato, 80, Campinas, SP, 13083-862, Brazil.
| | - Ruann Janser Soares de Castro
- Department of Food Science, School of Food Engineering, UNICAMP, Rua Monteiro Lobato, 80, Campinas, SP, 13083-862, Brazil
| | - Marie-Pierre Belleville
- Institut Européen des Membranes, Université de Montpellier, CNRS, ENSCM, UM, CC 047, 2 Place Eugène Bataillon, 34095, Montpellier, France
| | - Miriam Dupas Hubinger
- Laboratory of Process Engineering, Department of Food Engineering, School of Food Engineering, UNICAMP, Rua Monteiro Lobato, 80, Campinas, SP, 13083-862, Brazil
| |
Collapse
|
47
|
Bosi A, Banfi D, Bistoletti M, Giaroni C, Baj A. Tryptophan Metabolites Along the Microbiota-Gut-Brain Axis: An Interkingdom Communication System Influencing the Gut in Health and Disease. Int J Tryptophan Res 2020; 13:1178646920928984. [PMID: 32577079 PMCID: PMC7290275 DOI: 10.1177/1178646920928984] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/02/2020] [Indexed: 12/12/2022] Open
Abstract
The ‘microbiota-gut-brain axis’ plays a fundamental role in maintaining host homeostasis, and different immune, hormonal, and neuronal signals participate to this interkingdom communication system between eukaryota and prokaryota. The essential aminoacid tryptophan, as a precursor of several molecules acting at the interface between the host and the microbiota, is fundamental in the modulation of this bidirectional communication axis. In the gut, tryptophan undergoes 3 major metabolic pathways, the 5-HT, kynurenine, and AhR ligand pathways, which may be directly or indirectly controlled by the saprophytic flora. The importance of tryptophan metabolites in the modulation of the gastrointestinal tract is suggested by several preclinical and clinical studies; however, a thorough revision of the available literature has not been accomplished yet. Thus, this review attempts to cover the major aspects on the role of tryptophan metabolites in host-microbiota cross-talk underlaying regulation of gut functions in health conditions and during disease states, with particular attention to 2 major gastrointestinal diseases, such as irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD), both characterized by psychiatric disorders. Research in this area opens the possibility to target tryptophan metabolism to ameliorate the knowledge on the pathogenesis of both diseases, as well as to discover new therapeutic strategies based either on conventional pharmacological approaches or on the use of pre- and probiotics to manipulate the microbial flora.
Collapse
Affiliation(s)
- Annalisa Bosi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Davide Banfi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
48
|
Nassan FL, Gunn JA, Hill MM, Williams PL, Hauser R. Association of urinary concentrations of phthalate metabolites with quinolinic acid among women: A potential link to neurological disorders. ENVIRONMENT INTERNATIONAL 2020; 138:105643. [PMID: 32179323 PMCID: PMC7136979 DOI: 10.1016/j.envint.2020.105643] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/03/2020] [Accepted: 03/05/2020] [Indexed: 05/28/2023]
Abstract
BACKGROUND Quinolinic acid (QA), a neuroactive metabolite produced during tryptophan degradation, is implicated in the pathogenesis of several neurological disorders. Phthalates are structurally similar to QA, and exposure to phthalates has demonstrated increased QA production and excretion in rodent studies. We recently showed that very high exposure to dibutyl phthalate was associated with higher concentrations of urinary QA in men. However, no human studies examined the associations between background (low) phthalate exposures and QA. OBJECTIVES To examine the associations of urinary concentrations of phthalate metabolites with QA. METHODS Female participants (N = 126) who participated in a prospective cohort study at the Massachusetts General Hospital Fertility Center provided 758 urine samples (273 during pregnancy and 485 during non-pregnancy). Concentrations of 11 phthalate metabolites and QA in urine were measured. We used multivariable linear mixed effect models to estimate the percent change in urinary QA concentrations associated with a doubling (100%) of urinary phthalate metabolite concentration, and evaluated whether there was effect modification by pregnancy status. RESULTS Women's mean (standard deviation) age was 34.2 (4.0) years with a body mass index of 23.5 (3.7) kg/m2. The women were primarily Caucasian (92%), had at least a college degree (98%), and none were current smokers. Pairwise Spearman correlations between concentrations for phthalate metabolites and QA measured in the same urine samples ranged from 0.36 for MEHP to 0.68 for dibutyl phthalate (DBP) metabolites. In multivariable-adjusted models, the percent change in urinary QA concentrations was significantly higher for each doubling of several urinary phthalate metabolite concentrations. For example, each doubling of DBP metabolites was associated with a 13.7% (95%CI: 10.6, 16.9)% higher QA. Associations between the low molecular weight phthalate metabolites and QA were stronger among samples collected during pregnancy as compared to non-pregnancy samples from the same women. CONCLUSIONS Urinary concentrations of several phthalate metabolites were positively associated with QA among women. These findings, along with the known neurotoxicity of QA, warrant the need to examine whether QA concentrations may serve as a pathway for the adverse neurodevelopment outcomes found in children's health studies.
Collapse
Affiliation(s)
- Feiby L Nassan
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| | | | | | - Paige L Williams
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Russ Hauser
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Vincent Obstetrics and Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
49
|
Abstract
Chronic low-grade inflammation has been observed in major depression and other major psychiatric disorders and has been implicated in metabolic changes that are commonly associated with these disorders. This raises the possibility that the effects of dysfunctional metabolism may facilitate changes in neuronal structure and function which contribute to neuroprogression. Such changes may have implications for the progress from major depression to dementia in the elderly patient. The purpose of this review is to examine the contribution of inflammation and hypercortisolaemia, which are frequently associated with major depression, to neurodegeneration and how they detrimentally impact on brain energy metabolism. A key factor in these adverse events is insulin insensitivity caused by pro-inflammatory cytokines in association with desensitised glucocorticoid receptors. Identifying the possible metabolic changes initiated by inflammation opens new targets to ameliorate the adverse metabolic changes. This has resulted in the identification of dietary and drug targets which are of interest in the development of a new generation of psychotropic drugs.
Collapse
|
50
|
Esmaeili S, Ghobadi N, Akbari V, Moradi S, Shahlaie M, Ghobadi S, Jalalvand AR, Amani M, Khodarahmi R. Pyridine-2,3-dicarboxylate, quinolinic acid, induces 1N4R Tau amyloid aggregation in vitro: Another evidence for the detrimental effect of the inescapable endogenous neurotoxin. Chem Biol Interact 2020; 315:108884. [PMID: 31678113 DOI: 10.1016/j.cbi.2019.108884] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/18/2019] [Accepted: 10/26/2019] [Indexed: 01/19/2023]
Abstract
Quinolinic acid (QA) known as a neuro-active metabolite associated with the kynurenine pathway. At high concentrations, QA is often involved in the initiation and development of several human neurologic diseases, like Alzheimer's disease. Because of the QA action as the NMDA receptor, it is considered as a potent excitotoxin in vivo. Since it is probable that different mechanisms are employed by QA, activation of NMDA receptors cannot fully explain the revealed toxicity and it is even believed that there are multiple unknown mechanisms/targets leading to QA cytotoxicity. Herein we report accelerated amyloid oligomerization of 1N4R Tau under the effect of QA, in vitro, then the molecular structure, morphology and toxicity of the protein aggregate were documented by using various theoretical/experimental approaches. The possible mechanism of action of QA-induced Tau oligomerization has also been explored.
Collapse
Affiliation(s)
- Sajjad Esmaeili
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nazanin Ghobadi
- Department of Chemistry, School of Science, Alzahra University, Vanak, Tehran, Iran; Department of Chemistry and Biochemistry, Ohio University, Athens, OH, 45701, United States
| | - Vali Akbari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sajad Moradi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohsen Shahlaie
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sirous Ghobadi
- Department of Biology, Faculty of Sciences, Razi University, Kermanshah, Iran
| | - Ali Reza Jalalvand
- Research Center of Oils and Fats (RCOF), Food and Drug Administration (FDA), Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mojtaba Amani
- Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Reza Khodarahmi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Pharmacognosy and Biotechnology, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|