1
|
Li M, Wu Y, Xu Y, Huang X, Gao K, Hu N, Zhu S, Wang C, Liang S. Peripheral tryptophan-kynurenine pathway dysfunction in first-episode schizophrenia. Sci Rep 2025; 15:2432. [PMID: 39827210 PMCID: PMC11742721 DOI: 10.1038/s41598-025-86390-4] [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: 09/02/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
The tryptophan (TRP)-kynurenine (KYN) pathway is involved in the pathogenesis of schizophrenia. This study aimed to investigate the levels of TRP-KYN metabolites in serum and urine of patients with first-episode schizophrenia (FES) and their association with clinical manifestations. This study included 38 drug-naive patients with FES and 43 healthy controls (HCs). Clinical symptoms were evaluated using the Positive and Negative Syndrome Scale (PANSS). Levels of TRP-KYN metabolites in serum and urine were quantified. Patients with FES showed significantly higher serum quinolinic acid/kynurenic acid (QUIN/KYNA) ratio and urine KYN/TRP ratio compared to HCs, while neuroprotective metabolites, including serum KYNA, xanthurenic acid (XA), and urine picolinic acid (PIC) levels, were significantly reduced, along with a decreased urine PIC/QUIN ratio (p < 0.05). The urine KYNA/KYN ratio was negatively correlated with PANSS general psychopathology scores (r = -0.35, p = 0.04) and with PANSS total scores (r = -0.35, p = 0.046). Patients with FES exhibited dysregulation of the peripheral TRP-KYN pathway, characterized by an increased neurotoxic-to-neuroprotective QUIN/KYNA ratio and reduced levels of neuroprotective metabolites. This shift towards increased neurotoxic product generation suggests that the dysregulation of the TRP-KYN pathway could play a role in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Mian Li
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
- The Fourth Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yue Wu
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Yan Xu
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Xin Huang
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Kerun Gao
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Nannan Hu
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Shuangyue Zhu
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Chengpeng Wang
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Sugai Liang
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China.
| |
Collapse
|
2
|
Hatzimanolis A, Foteli S, Xenaki LA, Selakovic M, Dimitrakopoulos S, Vlachos I, Kosteletos I, Soldatos RF, Gazouli M, Chatzipanagiotou S, Stefanis N. Elevated serum kynurenic acid in individuals with first-episode psychosis and insufficient response to antipsychotics. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:61. [PMID: 38987245 PMCID: PMC11237022 DOI: 10.1038/s41537-024-00483-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/21/2024] [Indexed: 07/12/2024]
Abstract
The tryptophan-metabolizing kynurenine pathway (KP) can be activated by enhanced inflammatory responses and has been implicated in the pathophysiology of schizophrenia. However, there is little evidence for KP dysregulation in the early course of psychotic illness. We aimed to investigate the potential immune-mediated hyperactivity of KP in individuals with first-episode psychosis (FEP) and the relationship with symptom severity and treatment response outcomes. Serum immunoassays were performed to measure peripheral levels of inflammatory cytokines (IL-1β, IL-10, TNF-a), KP rate-limiting enzymes (IDO/TDO), and kynurenic acid (KYNA) metabolite in 104 antipsychotic-naïve patients with FEP and 80 healthy controls (HC). The Positive and Negative Syndrome Scale (PANSS) and the Global Assessment of Functioning Scale (GAF) were administered to assess psychopathology and functioning status at admission and following 4-week treatment with antipsychotics. Cytokine and KP components levels were substantially increased in FEP patients compared to HC, before and after antipsychotic treatment. A significant positive correlation between pro-inflammatory IL-1β and KYNA levels was observed among FEP patients, but not in HC. Importantly, within-patient analysis revealed that those with higher baseline KYNA experienced more severe negative symptoms and poorer clinical improvement at follow-up. These findings suggest that KP is upregulated in early psychosis, likely through the induction of IL-1β-dependent pathways, and raised peripheral KYNA might represent a promising indicator of non-response to antipsychotic medication in patients with FEP.
Collapse
Affiliation(s)
- Alex Hatzimanolis
- Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, Athens, Greece.
- Neurobiology Research Institute, Theodore-Theohari Cozzika Foundation, Athens, Greece.
| | - Stefania Foteli
- Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, Athens, Greece
- Department of Medical Biopathology, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, Athens, Greece
| | - Lida-Alkisti Xenaki
- Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, Athens, Greece
| | - Mirjana Selakovic
- Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, Athens, Greece
| | - Stefanos Dimitrakopoulos
- Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, Athens, Greece
| | - Ilias Vlachos
- Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, Athens, Greece
| | - Ioannis Kosteletos
- Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, Athens, Greece
| | - Rigas-Filippos Soldatos
- Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, Athens, Greece
| | - Maria Gazouli
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Stylianos Chatzipanagiotou
- Department of Medical Biopathology, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, Athens, Greece
| | - Nikos Stefanis
- Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, Athens, Greece
- Neurobiology Research Institute, Theodore-Theohari Cozzika Foundation, Athens, Greece
- World Federation of Societies of Biological Psychiatry, First Episode Psychosis Task Force, Barsbüttel, Germany
| |
Collapse
|
3
|
Burghardt KJ, Kajy M, Ward KM, Burghardt PR. Metabolomics, Lipidomics, and Antipsychotics: A Systematic Review. Biomedicines 2023; 11:3295. [PMID: 38137517 PMCID: PMC10741000 DOI: 10.3390/biomedicines11123295] [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: 11/08/2023] [Revised: 12/06/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Antipsychotics are an important pharmacotherapy option for the treatment of many mental illnesses. Unfortunately, selecting antipsychotics is often a trial-and-error process due to a lack of understanding as to which medications an individual patient will find most effective and best tolerated. Metabolomics, or the study of small molecules in a biosample, is an increasingly used omics platform that has the potential to identify biomarkers for medication efficacy and toxicity. This systematic review was conducted to identify metabolites and metabolomic pathways associated with antipsychotic use in humans. Ultimately, 42 studies were identified for inclusion in this review, with all but three studies being performed in blood sources such as plasma or serum. A total of 14 metabolite classes and 12 lipid classes were assessed across studies. Although the studies were highly heterogeneous in approach and mixed in their findings, increases in phosphatidylcholines, decreases in carboxylic acids, and decreases in acylcarnitines were most consistently noted as perturbed in patients exposed to antipsychotics. Furthermore, for the targeted metabolomic and lipidomic studies, seven metabolites and three lipid species had findings that were replicated. The most consistent finding for targeted studies was an identification of a decrease in aspartate with antipsychotic treatment. Studies varied in depth of detail provided for their study participants and in study design. For example, in some cases, there was a lack of detail on specific antipsychotics used or concomitant medications, and the depth of detail on sample handling and analysis varied widely. The conclusions here demonstrate that there is a large foundation of metabolomic work with antipsychotics that requires more complete reporting so that an objective synthesis such as a meta-analysis can take place. This will then allow for validation and clinical application of the most robust findings to move the field forward. Future studies should be carefully controlled to take advantage of the sensitivity of metabolomics while limiting potential confounders that may result from participant heterogeneity and varied analysis approaches.
Collapse
Affiliation(s)
- Kyle J. Burghardt
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University Detroit, Detroit, MI 48201, USA;
| | - Megan Kajy
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University Detroit, Detroit, MI 48201, USA;
| | - Kristen M. Ward
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan Ann Arbor, Detroit, MI 48109, USA;
| | - Paul R. Burghardt
- Department of Nutrition and Food Science, Wayne State University Detroit, Detroit, MI 48201, USA;
| |
Collapse
|
4
|
Jian J, He D, Gao S, Tao X, Dong X. Pharmacokinetics in Pharmacometabolomics: Towards Personalized Medication. Pharmaceuticals (Basel) 2023; 16:1568. [PMID: 38004434 PMCID: PMC10675232 DOI: 10.3390/ph16111568] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/19/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Indiscriminate drug administration may lead to drug therapy results with varying effects on patients, and the proposal of personalized medication can help patients to receive effective drug therapy. Conventional ways of personalized medication, such as pharmacogenomics and therapeutic drug monitoring (TDM), can only be implemented from a single perspective. The development of pharmacometabolomics provides a research method for the realization of precise drug administration, which integrates the environmental and genetic factors, and applies metabolomics technology to study how to predict different drug therapeutic responses of organisms based on baseline metabolic levels. The published research on pharmacometabolomics has achieved satisfactory results in predicting the pharmacokinetics, pharmacodynamics, and the discovery of biomarkers of drugs. Among them, the pharmacokinetics related to pharmacometabolomics are used to explore individual variability in drug metabolism from the level of metabolism of the drugs in vivo and the level of endogenous metabolite changes. By searching for relevant literature with the keyword "pharmacometabolomics" on the two major literature retrieval websites, PubMed and Web of Science, from 2006 to 2023, we reviewed articles in the field of pharmacometabolomics that incorporated pharmacokinetics into their research. This review explains the therapeutic effects of drugs on the body from the perspective of endogenous metabolites and pharmacokinetic principles, and reports the latest advances in pharmacometabolomics related to pharmacokinetics to provide research ideas and methods for advancing the implementation of personalized medication.
Collapse
Affiliation(s)
- Jingai Jian
- School of Medicine, Shanghai University, Shanghai 200444, China; (J.J.); (D.H.)
| | - Donglin He
- School of Medicine, Shanghai University, Shanghai 200444, China; (J.J.); (D.H.)
| | - Songyan Gao
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China;
| | - Xia Tao
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Xin Dong
- School of Medicine, Shanghai University, Shanghai 200444, China; (J.J.); (D.H.)
| |
Collapse
|
5
|
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: 272] [Impact Index Per Article: 136.0] [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
|
6
|
Almulla AF, Vasupanrajit A, Tunvirachaisakul C, Al-Hakeim HK, Solmi M, Verkerk R, Maes M. The tryptophan catabolite or kynurenine pathway in schizophrenia: meta-analysis reveals dissociations between central, serum, and plasma compartments. Mol Psychiatry 2022; 27:3679-3691. [PMID: 35422466 DOI: 10.1038/s41380-022-01552-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/19/2022] [Accepted: 03/25/2022] [Indexed: 02/08/2023]
Abstract
The tryptophan catabolite (TRYCAT) pathway is implicated in the pathophysiology of schizophrenia (SCZ) since the rate-limiting enzyme indoleamine-dioxygenase (IDO) may be induced by inflammatory and oxidative stress mediators. This systematic review searched PubMed, Web of Science, and Google Scholar for papers published from inception until August 2021 and meta-analyzed the association between SCZ and TRYCATs in the central nervous system (CNS) and peripheral blood. We included 61 studies comprising 2813 patients and 2948 healthy controls. In the CNS we found a significant (p < 0.001) increase in the kynurenine/tryptophan (KYN/TRP) (standardized mean difference, SMD = 0.769, 95% confidence interval, CI: 0.456; 1.082) and kynurenic acid (KA)/KYN + TRP (SMD = 0.697, CI: 0.478-0.917) ratios, KA (SMD = 0.646, CI: 0.422; 0.909) and KYN (SMD = 1.238; CI: 0.590; 1.886), while the 3OH-kynurenine (3HK) + KYN-3-monooxygenase (KMO)/KYN ratio was significantly reduced (SMD = -1.089, CI: -1.682; -0.496). There were significant differences between KYN/TRP, (KYN + KA)/TRP, (3HK + KMO)/KYN, KA, and KYN levels among the CNS and peripheral blood, and among serum and plasma KYN. The only useful peripheral marker of CNS TRYCATs findings was the increased KYN/TRP ratio in serum (SMD = 0.211, CI: 0.056; 0.366, p = 0.007), but not in plasma. There was no significant increase in a neurotoxic composite score based on KYN, 3HK, and picolinic, xanthurenic, and quinolinic acid. SCZ is accompanied by increased IDO activity in the CNS and serum, and reduced KMO activity and a shift towards KA production in the CNS. This CNS TRYCATs profile indicates neuroprotective, negative immunoregulatory and anti-inflammatory effects. Peripheral blood levels of TRYCATs are dissociated from CNS findings except for a modest increase in serum IDO activity.
Collapse
Affiliation(s)
- Abbas F Almulla
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Asara Vasupanrajit
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | | | - Marco Solmi
- Department of Psychiatry, University of Ottawa, Ottawa, ON, Canada.,Department of Mental Health, The Ottawa Hospital, Ottawa, ON, Canada.,Ottawa Hospital Research Institute (OHRI), Clinical Epidemiology Program, University of Ottawa, Ottawa, ON, Canada
| | - Robert Verkerk
- Laboratory of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand. .,Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria. .,Department of Psychiatry, IMPACT Strategic Research Centre, Deakin University, Geelong, VIC, Australia.
| |
Collapse
|
7
|
Dash S, Syed YA, Khan MR. Understanding the Role of the Gut Microbiome in Brain Development and Its Association With Neurodevelopmental Psychiatric Disorders. Front Cell Dev Biol 2022; 10:880544. [PMID: 35493075 PMCID: PMC9048050 DOI: 10.3389/fcell.2022.880544] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome has a tremendous influence on human physiology, including the nervous system. During fetal development, the initial colonization of the microbiome coincides with the development of the nervous system in a timely, coordinated manner. Emerging studies suggest an active involvement of the microbiome and its metabolic by-products in regulating early brain development. However, any disruption during this early developmental process can negatively impact brain functionality, leading to a range of neurodevelopment and neuropsychiatric disorders (NPD). In this review, we summarize recent evidence as to how the gut microbiome can influence the process of early human brain development and its association with major neurodevelopmental psychiatric disorders such as autism spectrum disorders, attention-deficit hyperactivity disorder, and schizophrenia. Further, we discuss how gut microbiome alterations can also play a role in inducing drug resistance in the affected individuals. We propose a model that establishes a direct link of microbiome dysbiosis with the exacerbated inflammatory state, leading to functional brain deficits associated with NPD. Based on the existing research, we discuss a framework whereby early diet intervention can boost mental wellness in the affected subjects and call for further research for a better understanding of mechanisms that govern the gut-brain axis may lead to novel approaches to the study of the pathophysiology and treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Somarani Dash
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Yasir Ahmed Syed
- School of Biosciences and Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Cardiff, United Kingdom
| | - Mojibur R. Khan
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, India
- *Correspondence: Mojibur R. Khan,
| |
Collapse
|
8
|
Yang Q, Zhang Y, Yang K, Niu Y, Fan F, Chen S, Luo X, Tan S, Wang Z, Tong J, Yang F, Li CSR, Tan Y. Associations of the serum kynurenine pathway metabolites with P50 auditory gating in non-smoking patients with first-episode schizophrenia. Front Psychiatry 2022; 13:1036421. [PMID: 36339840 PMCID: PMC9632432 DOI: 10.3389/fpsyt.2022.1036421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/05/2022] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE Our study aimed to investigate the associations between the serum level of kynurenine pathway (KP) metabolites and P50 auditory gating in non-smoking patients with first-episode schizophrenia (FES). MATERIALS AND METHODS In this study, 82 non-smoking patients with FES and 73 healthy controls (HC). P50 auditory gating was measured using a fully functional digital 64-channel EEG system, and the components included S1 amplitude, S2 amplitude, gating ratio (S2/S1), and amplitude difference (S1-S2). Serum levels of kynurenine and kynurenine acid were assessed using a combination of liquid chromatography with tandem mass spectrometry. Psychopathology was assessed by the Positive and Negative Syndrome Scale (PANSS). RESULTS The serum kynurenine (251.46 ± 65.93 ng/ml vs. 320.65 ± 65.89 ng/ml, t = -6.38, p < 0.001), and kynurenine acid levels (5.19 ± 2.22 ng/ml vs. 13.26 ± 4.23 ng/ml, t = -14.73, p < 0.001), S1 amplitude [2.88 (1.79, 3.78) μV vs. 3.08 (2.46, 4.56) μV, Z = -2.17, p = 0.030] and S1-S2 [1.60 (0.63, 2.49) μV vs. 1.92 (1.12, 2.93) μV, Z = -2.23, p = 0.026] in patients with FES were significantly lower than those in HC. The serum kynurenine and kynurenine acid levels were negatively associated with S1-S2 (r = -0.32, p = 0.004 and r = -0.42, p < 0.001; respectively) and positively correlated with S2/S1 ratio (r = 0.34, p = 0.002 and r = 0.35, p = 0.002; respectively) in patients. CONCLUSION Our findings suggested that neuroactive metabolites of the KP might play an important role in sensory gating deficit in first episode patients with schizophrenia. Furthermore, metabolites of the KP may be a new target for the treatment of cognitive impairments in schizophrenia.
Collapse
Affiliation(s)
- Qingyan Yang
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Yong Zhang
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Kebing Yang
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Yajuan Niu
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Fengmei Fan
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Song Chen
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Xingguang Luo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Shuping Tan
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Zhiren Wang
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Jinghui Tong
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Fude Yang
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Chiang-Shan R Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Yunlong Tan
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| |
Collapse
|
9
|
Evaluation of different internal standardization approaches for the quantification of melatonin in cell culture samples by multiple heart-cutting two dimensional liquid chromatography tandem mass spectrometry. J Chromatogr A 2021; 1663:462752. [PMID: 34954534 DOI: 10.1016/j.chroma.2021.462752] [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: 10/22/2021] [Revised: 12/04/2021] [Accepted: 12/14/2021] [Indexed: 11/23/2022]
Abstract
We evaluate here different analytical strategies for the chromatographic separation and determination of N-acetyl-5-methoxytryptamine (MEL) and its oxidative metabolites N1-acetyl-N2-formyl-5-methoxykynuramine (AFMK), N1-acetyl-5-methoxykynuramine (AMK) and cyclic 3-hydroxymelatonin (c3OHM) in cell culture samples. Two dimensional liquid chromatography (2D-LC) in the multiple heart-cutting mode was compared with regular 1D chromatographic separations of MEL and its oxidative metabolites. Our results showed that the use of trifluoroacetic acid (TFA) as mobile phase modifier was required to obtain a satisfactory resolution and peak shapes particularly for c3OHM. As TFA is not compatible with ESI ionization the application of the MHC mode was mandatory for a proper chromatographic separation. We evaluate also different internal standardization approaches based on the combined use of a surrogate standard (5-methoxytryptophol) and an internal standard (6-methoxytryptamine) for MEL quantification in cell culture samples obtaining unsatisfactory results both by 1D- and 2D-LC-ESI-MS/MS (from 9 ± 2 to 186 ± 38%). We demonstrate that only the application of isotope dilution Mass Spectrometry through the use of an in house synthesized 13C isotopically labelled analogue provided quantitative MEL recoveries both by using 1D- and 2D-LC-ESI-MS/MS (99±1 and 98±1. Respectively) in androgen-insensitive human prostate carcinoma PC3 cells.
Collapse
|
10
|
Büki A, Kekesi G, Horvath G, Vécsei L. A Potential Interface between the Kynurenine Pathway and Autonomic Imbalance in Schizophrenia. Int J Mol Sci 2021; 22:10016. [PMID: 34576179 PMCID: PMC8467675 DOI: 10.3390/ijms221810016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023] Open
Abstract
Schizophrenia is a neuropsychiatric disorder characterized by various symptoms including autonomic imbalance. These disturbances involve almost all autonomic functions and might contribute to poor medication compliance, worsened quality of life and increased mortality. Therefore, it has a great importance to find a potential therapeutic solution to improve the autonomic disturbances. The altered level of kynurenines (e.g., kynurenic acid), as tryptophan metabolites, is almost the most consistently found biochemical abnormality in schizophrenia. Kynurenic acid influences different types of receptors, most of them involved in the pathophysiology of schizophrenia. Only few data suggest that kynurenines might have effects on multiple autonomic functions. Publications so far have discussed the implication of kynurenines and the alteration of the autonomic nervous system in schizophrenia independently from each other. Thus, the coupling between them has not yet been addressed in schizophrenia, although their direct common points, potential interfaces indicate the consideration of their interaction. The present review gathers autonomic disturbances, the impaired kynurenine pathway in schizophrenia, and the effects of kynurenine pathway on autonomic functions. In the last part of the review, the potential interaction between the two systems in schizophrenia, and the possible therapeutic options are discussed.
Collapse
Affiliation(s)
- Alexandra Büki
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., H-6720 Szeged, Hungary; (A.B.); (G.K.); (G.H.)
| | - Gabriella Kekesi
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., H-6720 Szeged, Hungary; (A.B.); (G.K.); (G.H.)
| | - Gyongyi Horvath
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., H-6720 Szeged, Hungary; (A.B.); (G.K.); (G.H.)
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6., H-6725 Szeged, Hungary
- MTA-SZTE Neuroscience Research Group, H-6725 Szeged, Hungary
- Interdisciplinary Excellence Center, Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6., H-6725 Szeged, Hungary
| |
Collapse
|
11
|
Li R, Yang L, Guan S, Lin M, Lai H, Liu K, Liu Z, Zhang X. UPLC-MS-Based Serum Metabolic Profiling Reveals Potential Biomarkers for Predicting Propofol Responsiveness in Females. J Proteome Res 2021; 20:4578-4588. [PMID: 34384217 DOI: 10.1021/acs.jproteome.1c00554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Although previous studies have shown that certain factors interfere with the sensitivity of propofol, the mechanisms for interindividual variability in response to propofol remain unclear. This study aimed to screen the metabolites to predict patients' sensitivity to propofol and to identify metabolic pathways to explore possible mechanisms associated with propofol resistance. Sera from 40 female patients undergoing elective hysteroscopic surgery in a prospective cohort propofol study were obtained before the administration of propofol. The patients' responsiveness to propofol was differentiated based on propofol effect-site concentration. Serum samples from two sets, a discovery set (n = 24) and an independent validation set (n = 16), were analyzed using ultraperformance liquid chromatography coupled with mass spectrometry based untargeted metabolomics. In the discovery set, 494 differential metabolites were screened out, and then 391 potential candidate biomarkers with the area under receiver operating characteristic curve >0.80 were selected. Pathway analysis showed that the pathway of glycerophospholipid metabolism was the most influential pathway. In the independent validation set, six potential biomarkers enabled the discrimination of poor responders from good and intermediate responders, which might be applied to predict propofol sensitivity. The mass spectrometry data are available via MetaboLights (http://www.ebi.ac.uk/metabolights/login) with the identifier MTBLS2311.
Collapse
Affiliation(s)
- Ruiyun Li
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Lu Yang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Su Guan
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Ming Lin
- Department of Anesthesiology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China
| | - Hanjin Lai
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Kun Liu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Zimeng Liu
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Xuyu Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
12
|
Morrens M, Coppens V, Walther S. Do Immune Dysregulations and Oxidative Damage Drive Mood and Psychotic Disorders? Neuropsychobiology 2021; 79:251-254. [PMID: 30991415 DOI: 10.1159/000496622] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/07/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium, .,University Department of Psychiatry, Campus Duffel, Duffel, Belgium,
| | - Violette Coppens
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium.,University Department of Psychiatry, Campus Duffel, Duffel, Belgium
| | - Sebastian Walther
- Translational Research Center, University Hospital of Psychiatry, University of Bern, Bern, Switzerland
| |
Collapse
|
13
|
Noyan H, Erdağ E, Tüzün E, Yaylım İ, Küçükhüseyin Ö, Hakan MT, Gülöksüz S, Rutten BPF, Saka MC, Atbaşoğlu C, Alptekin K, van Os J, Üçok A. Association of the kynurenine pathway metabolites with clinical, cognitive features and IL-1β levels in patients with schizophrenia spectrum disorder and their siblings. Schizophr Res 2021; 229:27-37. [PMID: 33609988 DOI: 10.1016/j.schres.2021.01.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/11/2021] [Accepted: 01/19/2021] [Indexed: 11/16/2022]
Abstract
OBJECTIVE There is evidence suggesting that tryptophan (TRP)-kynurenine (KYN) pathway dysregulation is involved in the pathophysiology of schizophrenia and is regulated by inflammatory cytokines. The study investigate for the first time whether this dysregulation occurs in advanced stages of the disease as a byproduct or emerges as one of the early and inherited manifestations of schizophrenia. METHOD Sera of 148 patients with schizophrenia spectrum disorders (SCZ), 139 unaffected siblings (SIB) and 210 controls were investigated. Serum interleukin (IL)-1β levels were measured by ELISA, and TRP, KYN and kynurenic acid (KYNA) levels were measured by a high-performance liquid chromatography system. Also, we collected clinical data by applying Comprehensive Assessment of Symptoms and History in SCZ, and SIS-R in SIB and control groups. RESULTS Compared to controls, SCZ and SIB groups had lower TRP and higher KYNA levels. TRP levels showed significant differences only between SCZ and controls (p < 0.01). KYNA levels of both SCZ (p ≤ 0.001) and SIB (p < 0.05) were higher than controls. No statistical significance was found for KYN levels across groups. SCZ and SIB groups had higher serum IL-1β levels than controls (p ≤ 0.001). CONCLUSIONS Patients with SCZ and their siblings exhibited similar clinical features and TRP metabolite levels suggesting that TRP-KYN dysregulation may be an inherited component of the disease putatively conferring increased risk to schizophrenia. Elevation of IL-1β is one of the factors promoting overconsumption of the TRP-KYN pathway leading to increased production of neuroregulatory KYNA and presumably to neurodegeneration.
Collapse
Affiliation(s)
- Handan Noyan
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.
| | - Ece Erdağ
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Erdem Tüzün
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - İlhan Yaylım
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Özlem Küçükhüseyin
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Mehmet Tolgahan Hakan
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Sinan Gülöksüz
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands.
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands.
| | - Meram Can Saka
- Department of Psychiatry, School of Medicine, Ankara University, Ankara, Turkey
| | - Cem Atbaşoğlu
- Department of Psychiatry, School of Medicine, Ankara University, Ankara, Turkey
| | - Köksal Alptekin
- Department of Psychiatry, Dokuz Eylul University School of Medicine, Izmir, Turkey.
| | - Jim van Os
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands; Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Psychosis Studies, King's College London, Institute of Psychiatry, London, UK
| | - Alp Üçok
- Department of Psychiatry, Faculty of Medicine, Istanbul University, Istanbul, Turkey.
| |
Collapse
|
14
|
Cao B, Chen Y, Ren Z, Pan Z, McIntyre RS, Wang D. Dysregulation of kynurenine pathway and potential dynamic changes of kynurenine in schizophrenia: A systematic review and meta-analysis. Neurosci Biobehav Rev 2021; 123:203-214. [PMID: 33513412 DOI: 10.1016/j.neubiorev.2021.01.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/26/2022]
Abstract
The kynurenine (KYN) pathway is postulated to play various roles in immune system dysregulation of schizophrenia (SCZ). We conducted a meta-analysis to explore the association between six key metabolites of KYN pathway (i.e., tryptophan (TRP), KYN, quinolinic acid (QUIN), and kynurenic acid (KYNA)) and SCZ. Priori Bonferroni adjustments were conducted for multiple comparisons. In total, 42 studies that examined the relationship between the metabolites in KYN pathway mentioned above and SCZ in 4217 participants and nine studies that examined alterations of these metabolites after antipsychotic treatments were included. The results demonstrate that (1) subjects with prescribed medication had significantly higher KYN levels when compared to controls; (2) higher KYN levels in cerebrospinal fluid (CSF), lower plasma KYN levels and higher CSF KYNA levels were associated with SCZ; (3) the KYN levels were higher in subjects with SCZ after antipsychotic treatments when compared with baseline. The evidence provides valuable insight of the potential underlying involvement of the KYN pathway in the pathogenesis of SCZ.
Collapse
Affiliation(s)
- Bing Cao
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University (SWU), Chongqing, 400715, PR China; National Demonstration Center for Experimental Psychology Education (Southwest University), Chongqing, PR China.
| | - Yan Chen
- Dalla Lana School of Public Health, University of Toronto, 155 College St., Toronto, ON, Canada
| | - Zhongyu Ren
- College of Physical Education, Southwest University, Chongqing, PR China
| | - Zihang Pan
- Duke-NUS Medical School, Singapore, Singapore; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Dongfang Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
15
|
Reavis ZW, Mirjankar N, Sarangi S, Boyle SH, Kuhn CM, Matson WR, Babyak MA, Matson SA, Siegler IC, Kaddurah-Daouk R, Suarez EC, Williams RB, Grichnik K, Stafford-Smith M, Georgiades A. Sex and race differences of cerebrospinal fluid metabolites in healthy individuals. Metabolomics 2021; 17:13. [PMID: 33462762 PMCID: PMC8041469 DOI: 10.1007/s11306-020-01757-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Analyses of cerebrospinal fluid (CSF) metabolites in large, healthy samples have been limited and potential demographic moderators of brain metabolism are largely unknown. OBJECTIVE Our objective in this study was to examine sex and race differences in 33 CSF metabolites within a sample of 129 healthy individuals (37 African American women, 29 white women, 38 African American men, and 25 white men). METHODS CSF metabolites were measured with a targeted electrochemistry-based metabolomics platform. Sex and race differences were quantified with both univariate and multivariate analyses. Type I error was controlled for by using a Bonferroni adjustment (0.05/33 = .0015). RESULTS Multivariate Canonical Variate Analysis (CVA) of the 33 metabolites showed correct classification of sex at an average rate of 80.6% and correct classification of race at an average rate of 88.4%. Univariate analyses revealed that men had significantly higher concentrations of cysteine (p < 0.0001), uric acid (p < 0.0001), and N-acetylserotonin (p = 0.049), while women had significantly higher concentrations of 5-hydroxyindoleacetic acid (5-HIAA) (p = 0.001). African American participants had significantly higher concentrations of 3-hydroxykynurenine (p = 0.018), while white participants had significantly higher concentrations of kynurenine (p < 0.0001), indoleacetic acid (p < 0.0001), xanthine (p = 0.001), alpha-tocopherol (p = 0.007), cysteine (p = 0.029), melatonin (p = 0.036), and 7-methylxanthine (p = 0.037). After the Bonferroni adjustment, the effects for cysteine, uric acid, and 5-HIAA were still significant from the analysis of sex differences and kynurenine and indoleacetic acid were still significant from the analysis of race differences. CONCLUSION Several of the metabolites assayed in this study have been associated with mental health disorders and neurological diseases. Our data provide some novel information regarding normal variations by sex and race in CSF metabolite levels within the tryptophan, tyrosine and purine pathways, which may help to enhance our understanding of mechanisms underlying sex and race differences and potentially prove useful in the future treatment of disease.
Collapse
Affiliation(s)
- Zackery W Reavis
- Department of Pharmacology & Cancer Biology, School of Medicine, Duke University, Durham, NC, USA
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | | | | | - Stephen H Boyle
- Department of Psychiatry & Behavioral Sciences, School of Medicine, Duke University, Durham, NC, USA
| | - Cynthia M Kuhn
- Department of Pharmacology & Cancer Biology, School of Medicine, Duke University, Durham, NC, USA
| | | | - Michael A Babyak
- Department of Psychiatry & Behavioral Sciences, School of Medicine, Duke University, Durham, NC, USA
| | | | - Ilene C Siegler
- Department of Psychiatry & Behavioral Sciences, School of Medicine, Duke University, Durham, NC, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry & Behavioral Sciences, School of Medicine, Duke University, Durham, NC, USA
| | - Edward C Suarez
- Department of Psychiatry & Behavioral Sciences, School of Medicine, Duke University, Durham, NC, USA
| | - Redford B Williams
- Department of Psychiatry & Behavioral Sciences, School of Medicine, Duke University, Durham, NC, USA
| | | | - Mark Stafford-Smith
- Department of Anesthesiology, School of Medicine, Duke University, Durham, NC, USA
| | - Anastasia Georgiades
- Department of Psychiatry & Behavioral Sciences, School of Medicine, Duke University, Box 3454 DUMC, Durham, NC, 27710, USA.
| |
Collapse
|
16
|
Muneer A. Kynurenine Pathway of Tryptophan Metabolism in Neuropsychiatric Disorders: Pathophysiologic and Therapeutic Considerations. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2020; 18:507-526. [PMID: 33124585 PMCID: PMC7609208 DOI: 10.9758/cpn.2020.18.4.507] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/04/2020] [Accepted: 06/19/2020] [Indexed: 12/12/2022]
Abstract
Under physiological conditions 95% of the ingested essential amino acid tryptophan is metabolized by the kynurenine pathway (KP) to yield the ubiquitous co-enzyme nicotinamide adenine dinucleotide, fulfilling cellular energy require-ments. Importantly, the intermediaries of KP exert crucial effects throughout the body, including the central nervous system. Besides, KP metabolites are implicated in diverse disease processes such as inflammation/immune disorders, endocrine/metabolic conditions, cancers and neuropsychiatric diseases. A burgeoning body of research indicates that the KP plays a pathogenic role in major psychiatric diseases like mood disorders and schizophrenia. Triggered by inflammatory processes, the balance between neurotoxic and neuroprotective branches of the KP is disturbed. In preclinical models these discrepancies result in behaviors reminiscent of depression and psychosis. In clinical samples, recent studies are discovering key kynurenine pathway abnormalities which incriminate it in the pathogenesis of the main psychiatric disorders. Harnessing this knowledge has the potential to find disease biomarkers helpful in identifying and prognosticating neuropsychiatric disorders. Concurrently, earnest research efforts directed towards manipulating the KP hold the promise of discovering novel pharmacological agents that have therapeutic value. In this manuscript, an in-depth appraisal of the extant literature is done to understand the working of KP as this applies to neuropsychiatric disorders. It is concluded that this pathway plays an overarching role in the development of major psychiatric disorders, the KP metabolites have the potential to serve as disease markers and new medications based on KP modulation can bring lasting cures for patients suffering from these intractable conditions.
Collapse
Affiliation(s)
- Ather Muneer
- Islamic International Medical College, Riphah International University, Rawalpindi, Pakistan
| |
Collapse
|
17
|
Bilgiç A, Abuşoğlu S, Sadıç Çelikkol Ç, Oflaz MB, Akça ÖF, Sivrikaya A, Baysal T, Ünlü A. Altered kynurenine pathway metabolite levels in toddlers and preschool children with autism spectrum disorder. Int J Neurosci 2020; 132:826-834. [DOI: 10.1080/00207454.2020.1841187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Ayhan Bilgiç
- Department of Child and Adolescent Psychiatry, Meram School of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Sedat Abuşoğlu
- Department of Biochemistry, School of Medicine, Selcuk University, Konya, Turkey
| | - Çağla Sadıç Çelikkol
- Department of Child and Adolescent Psychiatry, Meram School of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Mehmet Burhan Oflaz
- Department of Pediatric Cardiology, Meram School of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Ömer Faruk Akça
- Department of Child and Adolescent Psychiatry, Meram School of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Abdullah Sivrikaya
- Department of Biochemistry, School of Medicine, Selcuk University, Konya, Turkey
| | - Tamer Baysal
- Department of Pediatric Cardiology, Meram School of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Ali Ünlü
- Department of Biochemistry, School of Medicine, Selcuk University, Konya, Turkey
| |
Collapse
|
18
|
Blood-based kynurenine pathway alterations in schizophrenia spectrum disorders: A meta-analysis. Schizophr Res 2020; 223:43-52. [PMID: 32981827 DOI: 10.1016/j.schres.2020.09.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 08/06/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The kynurenine pathway (KP) has been proposed as indirect link between systemic immune responses and clinical symptom development in schizophrenia spectrum disorders (SSD). Empirical evidence for such immune-related KP shifts in SSD has however resulted in divergent findings. METHODS We conducted a systematic literature search in PubMed. Thirty papers (total number of patients n = 1506; controls: n = 1432) reported on peripheral concentrations of KP metabolites in SSD patients versus controls. Six KP metabolites were included in a meta-analysis, with secondary analysis of covariate and subgroup effects of patients' symptomatic state, age and duration of illness. RESULTS Tryptophan (SMD: -0.30; p = .003) and Xanthurenic Acid (SMD: -0.80; p < .001) were significantly decreased in SSD compared to controls, while Quinolinic Acid (SMD: -0.40; p = .08) and Kynurenic Acid (SMD: -0.39; p = .04) were only significantly decreased in patients with acute or highly symptomatic illness. Finally, in relatively older patient cohorts Kynurenine (SMD: -0.31; p = .02) and Kynurenic Acid (SMD: -0.40; p = .002) were found to be decreased. CONCLUSION A partial downregulation of the KP is observed in SSD patients, in particular during acute symptomatic states and in older age, effects that were independent from each other. In contrast, younger and stable or remitted patients display limited to no KP metabolite abnormalities. The current meta-analysis illustrates the dynamic nature of KP abnormalities. It should be noted that all included studies investigated peripheral KP metabolites, which do not necessarily reflect central KP metabolite abnormalities in schizophrenic patients.
Collapse
|
19
|
Parksepp M, Leppik L, Koch K, Uppin K, Kangro R, Haring L, Vasar E, Zilmer M. Metabolomics approach revealed robust changes in amino acid and biogenic amine signatures in patients with schizophrenia in the early course of the disease. Sci Rep 2020; 10:13983. [PMID: 32814830 PMCID: PMC7438522 DOI: 10.1038/s41598-020-71014-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023] Open
Abstract
The primary objective of this study was to evaluate how schizophrenia (SCH) spectrum disorders and applied antipsychotic (AP) treatment affect serum level of amino acids (AAs) and biogenic amines (BAs) in the early course of the disorder. We measured 21 different AAs and 10 BAs in a sample of antipsychotic (AP)-naïve first-episode psychosis (FEP) patients (n = 52) at baseline, after 0.6-year as well as after 5.1-year treatment compared to control subjects (CSs, n = 37). Serum levels of metabolites were determined with AbsoluteIDQ p180 kit using flow injection analysis tandem mass spectrometry and liquid chromatography technique. Elevated level of taurine and reduced level of proline and alpha-aminoadipic acid (alpha-AAA) were established as metabolites with significant change in AP-naïve FEP patients compared to CSs. The following 0.6-year treatment restored these alterations. However, further continuous 5.1-year AP treatment changed the metabolic profile substantially. Significantly elevated levels of asparagine, glutamine, methionine, ornithine and taurine, alongside with decreased levels of aspartate, glutamate and alpha-AAA were observed in the patient group compared to CSs. These biomolecule profile alterations provide further insights into the pathophysiology of SCH spectrum disorders and broaden our understanding of the impact of AP treatment in the early stages of the disease.
Collapse
Affiliation(s)
- Madis Parksepp
- Department of Psychiatry, Institute of Clinical Medicine, University of Tartu, 31 Raja Street, 50417, Tartu, Estonia
- Psychiatry Clinic of Viljandi Hospital, 6 Pargi tee Street, 71024, Viljandi County, Estonia
| | - Liisa Leppik
- Psychiatry Clinic of Viljandi Hospital, 6 Pargi tee Street, 71024, Viljandi County, Estonia
| | - Kadri Koch
- Psychiatry Clinic of Tartu University Hospital, 31 Raja Street, 50417, Tartu, Estonia
| | - Kärt Uppin
- Psychiatry Clinic of Tartu University Hospital, 31 Raja Street, 50417, Tartu, Estonia
| | - Raul Kangro
- Institute of Mathematics and Statistics, University of Tartu, 18 Narva mnt, 51009, Tartu, Estonia
| | - Liina Haring
- Department of Psychiatry, Institute of Clinical Medicine, University of Tartu, 31 Raja Street, 50417, Tartu, Estonia.
- Psychiatry Clinic of Tartu University Hospital, 31 Raja Street, 50417, Tartu, Estonia.
- Institute of Biomedicine and Translational Medicine, Centre of Excellence for Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia.
| | - Eero Vasar
- Institute of Biomedicine and Translational Medicine, Centre of Excellence for Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Mihkel Zilmer
- Institute of Biomedicine and Translational Medicine, Centre of Excellence for Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| |
Collapse
|
20
|
Zhang Z, Zhang M, Luo Y, Ni X, Lu H, Wen Y, Fan N. Preliminary comparative analysis of kynurenine pathway metabolites in chronic ketamine users, schizophrenic patients, and healthy controls. Hum Psychopharmacol 2020; 35:e2738. [PMID: 32352599 DOI: 10.1002/hup.2738] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 01/02/2023]
Abstract
OBJECTIVE The serum kynurenine pathway metabolites kynurenic acid (KYNA), kynurenine (KYN), and tryptophan (TRP) were examined in chronic ketamine users and in schizophrenic patients. The correlations of the metabolites with sociodemographic data, clinical characteristics, and drug use status were analyzed. METHODS Seventy-nine healthy controls, 78 ketamine users, and 80 schizophrenic patients were recruited. Serum TRP, KYN, and KYNA levels were measured by high-performance liquid chromatography following tandem mass spectrometry (MS/MS). Psychotic symptoms were evaluated using the positive and negative syndrome scale (PANSS), the Beck Depression Inventory (BDI), and the Beck Anxiety Inventory (BAI). RESULTS Serum levels of TRP, KYNA, and KYN (in ketamine users only) were lower in ketamine users and schizophrenic patients than in controls (p < .05). TRP and KYN were lower in ketamine users than in schizophrenic patients (p < .01). KYNA levels were positively correlated with the current frequency of ketamine use in ketamine users (p = .031), and serum KYNA levels were negatively correlated with the duration of schizophrenia (p = .015). CONCLUSION TRP, KYNA, and KYN were lower in chronic ketamine users than in controls, and the alterations were in the same direction as those observed in schizophrenic patients.
Collapse
Affiliation(s)
- Zhaohua Zhang
- Department of Substance Dependence, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Minling Zhang
- Department of Substance Dependence, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Yayan Luo
- Department of Substance Dependence, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Xiaojia Ni
- Department of Substance Dependence, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Haoyang Lu
- Department of Substance Dependence, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Yuguan Wen
- Department of Substance Dependence, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Ni Fan
- Department of Substance Dependence, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| |
Collapse
|
21
|
Steen NE, Dieset I, Hope S, Vedal TSJ, Smeland OB, Matson W, Kaddurah-Daouk R, Agartz I, Melle I, Djurovic S, Jönsson EG, Bogdanov M, Andreassen OA. Metabolic dysfunctions in the kynurenine pathway, noradrenergic and purine metabolism in schizophrenia and bipolar disorders. Psychol Med 2020; 50:595-606. [PMID: 30867076 DOI: 10.1017/s0033291719000400] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND We aimed at exploring potential pathophysiological processes across psychotic disorders, applying metabolomics in a large and well-characterized sample of patients and healthy controls. METHODS Patients with schizophrenia and bipolar disorders (N = 212) and healthy controls (N = 68) had blood sampling with subsequent metabolomics analyses using electrochemical coulometric array detection. Concentrations of 52 metabolites including tyrosine, tryptophan and purine pathways were compared between patients and controls while controlling for demographic and clinical characteristics. Significant findings were further tested in medication-free subsamples. RESULTS Significantly decreased plasma concentrations in patients compared to healthy controls were found for 3-hydroxykynurenine (3OHKY, p = 0.0008), xanthurenic acid (XANU, p = 1.5×10-5), vanillylmandelic acid (VMA, p = 4.5×10-5) and metanephrine (MN, p = 0.0001). Plasma concentration of xanthine (XAN) was increased in the patient group (p = 3.5×10-5). Differences of 3OHKY, XANU, VMA and XAN were replicated across schizophrenia spectrum disorders and bipolar disorders subsamples of medication-free individuals. CONCLUSIONS Although prone to residual confounding, the present results suggest the kynurenine pathway of tryptophan metabolism, noradrenergic and purinergic system dysfunction as trait factors in schizophrenia spectrum and bipolar disorders. Of special interest is XANU, a metabolite previously not found to be associated with bipolar disorders.
Collapse
Affiliation(s)
- Nils Eiel Steen
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingrid Dieset
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Sigrun Hope
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurohabilitation, Oslo University Hospital, Oslo, Norway
| | - Trude S J Vedal
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Olav B Smeland
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neuroscience, University of California San Diego, La Jolla, CA92093, USA
| | | | - Rima Kaddurah-Daouk
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Ingrid Agartz
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Ingrid Melle
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Erik G Jönsson
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | | | - Ole A Andreassen
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
22
|
Dysregulation of kynurenine metabolism is related to proinflammatory cytokines, attention, and prefrontal cortex volume in schizophrenia. Mol Psychiatry 2020; 25:2860-2872. [PMID: 30940904 PMCID: PMC7577855 DOI: 10.1038/s41380-019-0401-9] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 11/09/2022]
Abstract
The kynurenine pathway (KP) of tryptophan (TRP) catabolism links immune system activation with neurotransmitter signaling. The KP metabolite kynurenic acid (KYNA) is increased in the brains of people with schizophrenia. We tested the extent to which: (1) brain KP enzyme mRNAs, (2) brain KP metabolites, and (3) plasma KP metabolites differed on the basis of elevated cytokines in schizophrenia vs. control groups and the extent to which plasma KP metabolites were associated with cognition and brain volume in patients displaying elevated peripheral cytokines. KP enzyme mRNAs and metabolites were assayed in two independent postmortem brain samples from a total of 71 patients with schizophrenia and 72 controls. Plasma KP metabolites, cognition, and brain volumes were measured in an independent cohort of 96 patients with schizophrenia and 81 healthy controls. Groups were stratified based on elevated vs. normal proinflammatory cytokine mRNA levels. In the prefrontal cortex (PFC), kynurenine (KYN)/TRP ratio, KYNA levels, and mRNA for enzymes, tryptophan dioxygenase (TDO) and kynurenine aminotransferases (KATI/II), were significantly increased in the high cytokine schizophrenia subgroup. KAT mRNAs significantly correlated with mRNA for glial fibrillary acidic protein in patients. In plasma, the high cytokine schizophrenia subgroup displayed an elevated KYN/TRP ratio, which correlated inversely with attention and dorsolateral prefrontal cortex (DLPFC) volume. This study provides further evidence for the role of inflammation in a subgroup of patients with schizophrenia and suggests a molecular mechanism through which inflammation could lead to schizophrenia. Proinflammatory cytokines may elicit conversion of TRP to KYN in the periphery and increase the N-methyl-D-aspartate receptor antagonist KYNA via increased KAT mRNA and possibly more enzyme synthesis activity in brain astrocytes, leading to DLPFC volume loss, and attention impairment in schizophrenia.
Collapse
|
23
|
Trovão N, Prata J, VonDoellinger O, Santos S, Barbosa M, Coelho R. Peripheral Biomarkers for First-Episode Psychosis-Opportunities from the Neuroinflammatory Hypothesis of Schizophrenia. Psychiatry Investig 2019; 16:177-184. [PMID: 30836740 PMCID: PMC6444098 DOI: 10.30773/pi.2018.12.19.1] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/19/2018] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVE Schizophrenia is a disabling disorder of unknown aetiology, lacking definite diagnostic method and cure. A reliable biological marker of schizophrenia is highly demanded, for which traceable immune mediators in blood could be promising candidates. We aimed to gather the best findings of neuroinflammatory markers for first-episode psychosis (FEP). METHODS We performed an extensive narrative review of online literature on inflammation-related markers found in human FEP patients only. RESULTS Changes to cytokine levels have been increasingly reported in schizophrenia. The peripheral levels of IL-1 (or its receptor antagonist), soluble IL-2 receptor, IL-4, IL-6, IL-8, and TNF-α have been frequently reported as increased in FEP, in a suggestive continuum from high-risk stages for psychosis. Microglia and astrocytes establish the link between this immune signalling and the synthesis of noxious tryptophan catabolism products, that cause structural damage and directly hamper normal neurotransmission. Amongst these, only 3-hydroxykynurenine has been consistently described in the blood of FEP patients. CONCLUSION Peripheral molecules stemming from brain inflammation might provide insightful biomarkers of schizophrenia, as early as FEP or even prodromal phases, although more time- and clinically-adjusted studies are essential for their validation.
Collapse
Affiliation(s)
- Nuno Trovão
- Department of Psychiatry, Vila Nova de Gaia/ Espinho Hospital Center, Vila Nova de Gaia, Portugal.,Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Department of Psychiatry, Faculty of Medicine of University of Porto, Porto, Portugal
| | - Joana Prata
- Department of Psychiatry, Vila Nova de Gaia/ Espinho Hospital Center, Vila Nova de Gaia, Portugal.,Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Department of Psychiatry, Faculty of Medicine of University of Porto, Porto, Portugal
| | - Orlando VonDoellinger
- Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Department of Psychiatry, Tâmega e Sousa Hospital Center, Penafiel, Portugal
| | - Susana Santos
- Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal
| | - Mário Barbosa
- Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Rui Coelho
- Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Department of Psychiatry, Faculty of Medicine of University of Porto, Porto, Portugal
| |
Collapse
|
24
|
Everett JR. Pharmacometabonomics: The Prediction of Drug Effects Using Metabolic Profiling. Handb Exp Pharmacol 2019; 260:263-299. [PMID: 31823071 DOI: 10.1007/164_2019_316] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metabonomics, also known as metabolomics, is concerned with the study of metabolite profiles in humans, animals, plants and other systems in order to assess their health or other status and their responses to experimental interventions. Metabonomics is thus widely used in disease diagnosis and in understanding responses to therapies such as drug administration. Pharmacometabonomics, also known as pharmacometabolomics, is a related methodology but with a prognostic as opposed to diagnostic thrust. Pharmacometabonomics aims to predict drug effects including efficacy, safety, metabolism and pharmacokinetics, prior to drug administration, via an analysis of pre-dose metabolite profiles. This article will review the development of pharmacometabonomics as a new field of science that has much promise in helping to deliver more effective personalised medicine, a major goal of twenty-first century healthcare.
Collapse
Affiliation(s)
- Jeremy R Everett
- Medway Metabonomics Research Group, University of Greenwich, Kent, UK.
| |
Collapse
|
25
|
Abstract
This paper discusses the current evidence from animal and human studies for a central role of inflammation in schizophrenia. In animal models, pre- or perinatal elicitation of the immune response may increase immune reactivity throughout life, and similar findings have been described in humans. Levels of pro-inflammatory markers, such as cytokines, have been found to be increased in the blood and cerebrospinal fluid of patients with schizophrenia. Numerous epidemiological and clinical studies have provided evidence that various infectious agents are risk factors for schizophrenia and other psychoses. For example, a large-scale epidemiological study performed in Denmark clearly showed that severe infections and autoimmune disorders are such risk factors. The vulnerability-stress-inflammation model may help to explain the role of inflammation in schizophrenia because stress can increase pro-inflammatory cytokines and may even contribute to a chronic pro-inflammatory state. Schizophrenia is characterized by risk genes that promote inflammation and by environmental stress factors and alterations of the immune system. Typical alterations of dopaminergic, serotonergic, noradrenergic, and glutamatergic neurotransmission described in schizophrenia have also been found in low-level neuroinflammation and consequently may be key factors in the generation of schizophrenia symptoms. Further support for the relevance of a low-level neuroinflammatory process in schizophrenia is provided by the loss of central nervous system volume and microglial activation demonstrated in neuroimaging studies. Last but not least, the benefit of anti-inflammatory medications found in some studies and the intrinsic anti-inflammatory and immunomodulatory effects of antipsychotics provide further support for the role of inflammation in this debilitating disease.
Collapse
Affiliation(s)
- Norbert Müller
- Department of Psychiatry and Psychotherapy Ludwig Maximilian University and Marion von Tessin Memory Center, Munich, Germany
| |
Collapse
|
26
|
Cai H, Zhou X, Dougherty GG, Reddy RD, Haas GL, Montrose DM, Keshavan M, Yao JK. Pregnenolone-progesterone-allopregnanolone pathway as a potential therapeutic target in first-episode antipsychotic-naïve patients with schizophrenia. Psychoneuroendocrinology 2018; 90:43-51. [PMID: 29433072 PMCID: PMC5864547 DOI: 10.1016/j.psyneuen.2018.02.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/29/2017] [Accepted: 02/04/2018] [Indexed: 11/23/2022]
Abstract
Neurosteroids are both endogenous and exogenous steroids that rapidly alter neuronal excitability through interactions with ligand-gated ion channels and other cell surface receptors. They are originated from cholesterol and have important implications for schizophrenia (SZ) pathophysiology and treatment strategies. Specifically, pregnenolone (PREG), progesterone (PROG) and allopregnanolone (ALLO) exhibit similar psychotropic properties. Using enzyme immunoassay, we compared the neurosteroids in PREG downstream pathways in plasma between healthy controls (HC, n = 43) and first-episode antipsychotic-naïve patients with SZ (FEAN-SZ, n = 53) before antipsychotic drug (APD) treatment. Comparisons were also made particularly along PREG-PROG-ALLO pathway in the same FEAN-SZ patients across multiple time points following initiation of treatment for 12 months (m). Firstly, at baseline, levels of PREG were significantly higher and those of ALLO were lower in FEAN-SZ than in HC, whereas PROG, cortisol, dehydroepiandrosterone (DHEA) and dehydroepiandrosterone sulfate (DHEAS) were not different. Consequently, the molar ratios of ALLO/PREG and ALLO/PROG in FEAN-SZ were significantly reduced. Secondly, in response to APD at 1 month, ALLO levels in FEAN-SZ were markedly elevated, whereas PREG and PROG levels decreased. Thirdly, among FEAN-SZ, lower levels of PROG (reflecting higher conversion to ALLO) at baseline may predict better therapeutic outcome after 1 month of APD treatment. These findings point to the perturbations of the PREG-PROG-ALLO pathway early in psychosis, and further study of this pathway may inform alternative and innovative therapeutic targets for SZ.
Collapse
Affiliation(s)
- HuaLin Cai
- Medical Research Service, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; Departments of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15216, USA; The Second Xiangya Hospital and Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
| | - Xiang Zhou
- Medical Research Service, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; Departments of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - George G Dougherty
- Medical Research Service, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Ravinder D Reddy
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Gretchen L Haas
- Medical Research Service, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Debra M Montrose
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Matcheri Keshavan
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Psychiatry, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey K Yao
- Medical Research Service, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; Departments of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15216, USA; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
27
|
Suvisaari J, Mantere O, Keinänen J, Mäntylä T, Rikandi E, Lindgren M, Kieseppä T, Raij TT. Is It Possible to Predict the Future in First-Episode Psychosis? Front Psychiatry 2018; 9:580. [PMID: 30483163 PMCID: PMC6243124 DOI: 10.3389/fpsyt.2018.00580] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/23/2018] [Indexed: 12/26/2022] Open
Abstract
The outcome of first-episode psychosis (FEP) is highly variable, ranging from early sustained recovery to antipsychotic treatment resistance from the onset of illness. For clinicians, a possibility to predict patient outcomes would be highly valuable for the selection of antipsychotic treatment and in tailoring psychosocial treatments and psychoeducation. This selective review summarizes current knowledge of prognostic markers in FEP. We sought potential outcome predictors from clinical and sociodemographic factors, cognition, brain imaging, genetics, and blood-based biomarkers, and we considered different outcomes, like remission, recovery, physical comorbidities, and suicide risk. Based on the review, it is currently possible to predict the future for FEP patients to some extent. Some clinical features-like the longer duration of untreated psychosis (DUP), poor premorbid adjustment, the insidious mode of onset, the greater severity of negative symptoms, comorbid substance use disorders (SUDs), a history of suicide attempts and suicidal ideation and having non-affective psychosis-are associated with a worse outcome. Of the social and demographic factors, male gender, social disadvantage, neighborhood deprivation, dysfunctional family environment, and ethnicity may be relevant. Treatment non-adherence is a substantial risk factor for relapse, but a small minority of patients with acute onset of FEP and early remission may benefit from antipsychotic discontinuation. Cognitive functioning is associated with functional outcomes. Brain imaging currently has limited utility as an outcome predictor, but this may change with methodological advancements. Polygenic risk scores (PRSs) might be useful as one component of a predictive tool, and pharmacogenetic testing is already available and valuable for patients who have problems in treatment response or with side effects. Most blood-based biomarkers need further validation. None of the currently available predictive markers has adequate sensitivity or specificity used alone. However, personalized treatment of FEP will need predictive tools. We discuss some methodologies, such as machine learning (ML), and tools that could lead to the improved prediction and clinical utility of different prognostic markers in FEP. Combination of different markers in ML models with a user friendly interface, or novel findings from e.g., molecular genetics or neuroimaging, may result in computer-assisted clinical applications in the near future.
Collapse
Affiliation(s)
- Jaana Suvisaari
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Outi Mantere
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland.,Department of Psychiatry, McGill University, Montreal, QC, Canada.,Bipolar Disorders Clinic, Douglas Mental Health University Institute, Montreal, QC, Canada.,Department of Psychiatry, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jaakko Keinänen
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland.,Department of Psychiatry, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Teemu Mäntylä
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland.,Department of Neuroscience and Biomedical Engineering, and Advanced Magnetic Imaging Center, Aalto NeuroImaging, Aalto University School of Science, Espoo, Finland.,Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Eva Rikandi
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland.,Department of Neuroscience and Biomedical Engineering, and Advanced Magnetic Imaging Center, Aalto NeuroImaging, Aalto University School of Science, Espoo, Finland.,Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Maija Lindgren
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Tuula Kieseppä
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland.,Department of Psychiatry, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tuukka T Raij
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland.,Department of Neuroscience and Biomedical Engineering, and Advanced Magnetic Imaging Center, Aalto NeuroImaging, Aalto University School of Science, Espoo, Finland
| |
Collapse
|
28
|
Everett JR. NMR-based pharmacometabonomics: A new paradigm for personalised or precision medicine. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2017; 102-103:1-14. [PMID: 29157489 DOI: 10.1016/j.pnmrs.2017.04.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 04/23/2017] [Accepted: 04/24/2017] [Indexed: 06/07/2023]
Abstract
Metabolic profiling by NMR spectroscopy or hyphenated mass spectrometry, known as metabonomics or metabolomics, is an important tool for systems-based approaches in biology and medicine. The experiments are typically done in a diagnostic fashion where changes in metabolite profiles are interpreted as a consequence of an intervention or event; be that a change in diet, the administration of a drug, physical exertion or the onset of a disease. By contrast, pharmacometabonomics takes a prognostic approach to metabolic profiling, in order to predict the effects of drug dosing before it occurs. Differences in pre-dose metabolite profiles between groups of subjects are used to predict post-dose differences in response to drug administration. Thus the paradigm is inverted and pharmacometabonomics is the metabolic equivalent of pharmacogenomics. Although the field is still in its infancy, it is expected that pharmacometabonomics, alongside pharmacogenomics, will assist with the delivery of personalised or precision medicine to patients, which is a critical goal of 21st century healthcare.
Collapse
Affiliation(s)
- Jeremy R Everett
- Medway Metabonomics Group, University of Greenwich, Chatham Maritime, Kent ME4 4TB, UK.
| |
Collapse
|
29
|
Bridging Autism Spectrum Disorders and Schizophrenia through inflammation and biomarkers - pre-clinical and clinical investigations. J Neuroinflammation 2017; 14:179. [PMID: 28870209 PMCID: PMC5584030 DOI: 10.1186/s12974-017-0938-y] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/08/2017] [Indexed: 12/15/2022] Open
Abstract
In recent years, evidence supporting a link between inflammation and neuropsychiatric disorders has been mounting. Autism spectrum disorders (ASD) and schizophrenia share some clinical similarities which we hypothesize might reflect the same biological basis, namely, in terms of inflammation. However, the diagnosis of ASD and schizophrenia relies solely on clinical symptoms, and to date, there is no clinically useful biomarker to diagnose or monitor the course of such illnesses. The focus of this review is the central role that inflammation plays in ASD and schizophrenia. It spans from pre-clinical animal models to clinical research and excludes in vitro studies. Four major areas are covered: (1) microglia, the inflammatory brain resident myeloid cells, (2) biomarkers, including circulating cytokines, oxidative stress markers, and microRNA players, known to influence cellular processes at brain and immune levels, (3) effect of anti-psychotics on biomarkers and other predictors of response, and (4) impact of gender on response to immune activation, biomarkers, and response to anti-psychotic treatments.
Collapse
|
30
|
Central Nervous System Infection with Borna Disease Virus Causes Kynurenine Pathway Dysregulation and Neurotoxic Quinolinic Acid Production. J Virol 2017; 91:JVI.00673-17. [PMID: 28446679 PMCID: PMC5487560 DOI: 10.1128/jvi.00673-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 12/27/2022] Open
Abstract
Central nervous system infection of neonatal and adult rats with Borna disease virus (BDV) results in neuronal destruction and behavioral abnormalities with differential immune-mediated involvement. Neuroactive metabolites generated from the kynurenine pathway of tryptophan degradation have been implicated in several human neurodegenerative disorders. Here, we report that brain expression of key enzymes in the kynurenine pathway are significantly, but differentially, altered in neonatal and adult rats with BDV infection. Gene expression analysis of rat brains following neonatal infection showed increased expression of kynurenine amino transferase II (KATII) and kynurenine-3-monooxygenase (KMO) enzymes. Additionally, indoleamine 2,3-dioxygenase (IDO) expression was only modestly increased in a brain region- and time-dependent manner in neonatally infected rats; however, its expression was highly increased in adult infected rats. The most dramatic impact on gene expression was seen for KMO, whose activity promotes the production of neurotoxic quinolinic acid. KMO expression was persistently elevated in brain regions of both newborn and adult BDV-infected rats, with increases reaching up to 86-fold. KMO protein levels were increased in neonatally infected rats and colocalized with neurons, the primary target cells of BDV infection. Furthermore, quinolinic acid was elevated in neonatally infected rat brains. We further demonstrate increased expression of KATII and KMO, but not IDO, in vitro in BDV-infected C6 astroglioma cells. Our results suggest that BDV directly impacts the kynurenine pathway, an effect that may be exacerbated by inflammatory responses in immunocompetent hosts. Thus, experimental models of BDV infection may provide new tools for discriminating virus-mediated from immune-mediated impacts on the kynurenine pathway and their relative contribution to neurodegeneration.IMPORTANCE BDV causes persistent, noncytopathic infection in vitro yet still elicits widespread neurodegeneration of infected neurons in both immunoincompetent and immunocompetent hosts. Here, we show that BDV infection induces expression of key enzymes of the kynurenine pathway in brains of newborn and adult infected rats and cultured astroglioma cells, shunting tryptophan degradation toward the production of neurotoxic quinolinic acid. Thus, our findings newly implicate this metabolic pathway in BDV-induced neurodegeneration. Given the importance of the kynurenine pathway in a wide range of human infections and neurodegenerative and neuropsychiatric disorders, animal models of BDV infection may serve as important tools for contrasting direct viral and indirect antiviral immune-mediated impacts on kynurenine pathway dysregulation and the ensuing neurodevelopmental and neuropathological consequences.
Collapse
|
31
|
Metabolomics biomarkers to predict acamprosate treatment response in alcohol-dependent subjects. Sci Rep 2017; 7:2496. [PMID: 28566752 PMCID: PMC5451388 DOI: 10.1038/s41598-017-02442-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 04/12/2017] [Indexed: 01/25/2023] Open
Abstract
Precision medicine for alcohol use disorder (AUD) allows optimal treatment of the right patient with the right drug at the right time. Here, we generated multivariable models incorporating clinical information and serum metabolite levels to predict acamprosate treatment response. The sample of 120 patients was randomly split into a training set (n = 80) and test set (n = 40) five independent times. Treatment response was defined as complete abstinence (no alcohol consumption during 3 months of acamprosate treatment) while nonresponse was defined as any alcohol consumption during this period. In each of the five training sets, we built a predictive model using a least absolute shrinkage and section operator (LASSO) penalized selection method and then evaluated the predictive performance of each model in the corresponding test set. The models predicted acamprosate treatment response with a mean sensitivity and specificity in the test sets of 0.83 and 0.31, respectively, suggesting our model performed well at predicting responders, but not non-responders (i.e. many non-responders were predicted to respond). Studies with larger sample sizes and additional biomarkers will expand the clinical utility of predictive algorithms for pharmaceutical response in AUD.
Collapse
|
32
|
Correlations of Kynurenic Acid, 3-Hydroxykynurenine, sIL-2R, IFN-α, and IL-4 with Clinical Symptoms During Acute Relapse of Schizophrenia. Neurotox Res 2017; 32:17-26. [DOI: 10.1007/s12640-017-9714-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/09/2017] [Accepted: 02/24/2017] [Indexed: 12/13/2022]
|
33
|
The kynurenine pathway in schizophrenia and bipolar disorder. Neuropharmacology 2017; 112:297-306. [DOI: 10.1016/j.neuropharm.2016.05.020] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 05/26/2016] [Accepted: 05/27/2016] [Indexed: 11/20/2022]
|
34
|
Kantae V, Krekels EHJ, Esdonk MJV, Lindenburg P, Harms AC, Knibbe CAJ, Van der Graaf PH, Hankemeier T. Integration of pharmacometabolomics with pharmacokinetics and pharmacodynamics: towards personalized drug therapy. Metabolomics 2016; 13:9. [PMID: 28058041 PMCID: PMC5165030 DOI: 10.1007/s11306-016-1143-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/26/2016] [Indexed: 02/05/2023]
Abstract
Personalized medicine, in modern drug therapy, aims at a tailored drug treatment accounting for inter-individual variations in drug pharmacology to treat individuals effectively and safely. The inter-individual variability in drug response upon drug administration is caused by the interplay between drug pharmacology and the patients' (patho)physiological status. Individual variations in (patho)physiological status may result from genetic polymorphisms, environmental factors (including current/past treatments), demographic characteristics, and disease related factors. Identification and quantification of predictors of inter-individual variability in drug pharmacology is necessary to achieve personalized medicine. Here, we highlight the potential of pharmacometabolomics in prospectively informing on the inter-individual differences in drug pharmacology, including both pharmacokinetic (PK) and pharmacodynamic (PD) processes, and thereby guiding drug selection and drug dosing. This review focusses on the pharmacometabolomics studies that have additional value on top of the conventional covariates in predicting drug PK. Additionally, employing pharmacometabolomics to predict drug PD is highlighted, and we suggest not only considering the endogenous metabolites as static variables but to include also drug dose and temporal changes in drug concentration in these studies. Although there are many endogenous metabolite biomarkers identified to predict PK and more often to predict PD, validation of these biomarkers in terms of specificity, sensitivity, reproducibility and clinical relevance is highly important. Furthermore, the application of these identified biomarkers in routine clinical practice deserves notable attention to truly personalize drug treatment in the near future.
Collapse
Affiliation(s)
- Vasudev Kantae
- Division of Analytical Biosciences, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Elke H. J. Krekels
- Division of Pharmacology, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Michiel J. Van Esdonk
- Division of Pharmacology, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Peter Lindenburg
- Division of Analytical Biosciences, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Amy C. Harms
- Division of Analytical Biosciences, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Catherijne A. J. Knibbe
- Division of Pharmacology, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Piet H. Van der Graaf
- Division of Pharmacology, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Certara QSP, Canterbury Innovation Centre, Canterbury, UK
| | - Thomas Hankemeier
- Division of Analytical Biosciences, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| |
Collapse
|
35
|
Everett JR. From Metabonomics to Pharmacometabonomics: The Role of Metabolic Profiling in Personalized Medicine. Front Pharmacol 2016; 7:297. [PMID: 27660611 PMCID: PMC5014868 DOI: 10.3389/fphar.2016.00297] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/23/2016] [Indexed: 01/08/2023] Open
Abstract
Variable patient responses to drugs are a key issue for medicine and for drug discovery and development. Personalized medicine, that is the selection of medicines for subgroups of patients so as to maximize drug efficacy and minimize toxicity, is a key goal of twenty-first century healthcare. Currently, most personalized medicine paradigms rely on clinical judgment based on the patient's history, and on the analysis of the patients' genome to predict drug effects i.e., pharmacogenomics. However, variability in patient responses to drugs is dependent upon many environmental factors to which human genomics is essentially blind. A new paradigm for predicting drug responses based on individual pre-dose metabolite profiles has emerged in the past decade: pharmacometabonomics, which is defined as “the prediction of the outcome (for example, efficacy or toxicity) of a drug or xenobiotic intervention in an individual based on a mathematical model of pre-intervention metabolite signatures.” The new pharmacometabonomics paradigm is complementary to pharmacogenomics but has the advantage of being sensitive to environmental as well as genomic factors. This review will chart the discovery and development of pharmacometabonomics, and provide examples of its current utility and possible future developments.
Collapse
Affiliation(s)
- Jeremy R Everett
- Medway Metabonomics Research Group, University of Greenwich Kent, UK
| |
Collapse
|
36
|
Goff DC, Romero K, Paul J, Mercedes Perez-Rodriguez M, Crandall D, Potkin SG. Biomarkers for drug development in early psychosis: Current issues and promising directions. Eur Neuropsychopharmacol 2016; 26:923-37. [PMID: 27005595 DOI: 10.1016/j.euroneuro.2016.01.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/20/2016] [Accepted: 01/23/2016] [Indexed: 12/14/2022]
Abstract
A major goal of current research in schizophrenia is to understand the biology underlying onset and early progression and to develop interventions that modify these processes. Biomarkers can play a critical role in identifying disease state, factors contributing to underlying progression, as well as predicting and monitoring response to treatment. Once biomarker-based therapeutics are established, biomarkers can guide treatment selection. It is increasingly clear that a wide range of potential biomarkers should be examined in schizophrenia, given the large number of genetic and environmental factors that have been identified as risk factors. New models for analysis of biomarkers are needed that represent the central nervous system as a highly complex, dynamic, and interactive system. Many tools are available with which to study relevant brain chemistry, but most are indirect measures and represent only a small fraction of the potential etiologic factors contributing to the molecular, structural and functional components of schizophrenia. This review represents the work of the International Society for CNS Clinical Trials and Methodology (ISCTM) Biomarkers Working Group. It discusses advantages and disadvantages of different categories of biomarkers and provides a summary of evidence that biomarkers representing inflammation, oxidative stress, endocannabinoids, glucocorticoid, and biogenic amines systems are dysregulated and potentially interactive in early phase schizophrenia. As has been recently demonstrated in several neurodevelopmental and neurodegenerative disorders, a multi-modal, longitudinal strategy involving a diverse array of biomarkers and new approaches to statistical modeling are needed to improve early interventions based on the fuller understanding.
Collapse
Affiliation(s)
| | | | - Jeffrey Paul
- Astellas Pharma Global Development, Northbrook, IL, USA
| | | | | | | |
Collapse
|
37
|
Fazio F, Lionetto L, Curto M, Iacovelli L, Cavallari M, Zappulla C, Ulivieri M, Napoletano F, Capi M, Corigliano V, Scaccianoce S, Caruso A, Miele J, De Fusco A, Di Menna L, Comparelli A, De Carolis A, Gradini R, Nisticò R, De Blasi A, Girardi P, Bruno V, Battaglia G, Nicoletti F, Simmaco M. Xanthurenic Acid Activates mGlu2/3 Metabotropic Glutamate Receptors and is a Potential Trait Marker for Schizophrenia. Sci Rep 2015; 5:17799. [PMID: 26643205 PMCID: PMC4672300 DOI: 10.1038/srep17799] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 11/06/2015] [Indexed: 11/09/2022] Open
Abstract
The kynurenine pathway of tryptophan metabolism has been implicated in the pathophysiology of psychiatric disorders, including schizophrenia. We report here that the kynurenine metabolite, xanturenic acid (XA), interacts with, and activates mGlu2 and mGlu3 metabotropic glutamate receptors in heterologous expression systems. However, the molecular nature of this interaction is unknown, and our data cannot exclude that XA acts primarily on other targets, such as the vesicular glutamate transporter, in the CNS. Systemic administration of XA in mice produced antipsychotic-like effects in the MK-801-induced model of hyperactivity. This effect required the presence of mGlu2 receptors and was abrogated by the preferential mGlu2/3 receptor antagonist, LY341495. Because the mGlu2 receptor is a potential drug target in the treatment of schizophrenia, we decided to measure serum levels of XA and other kynurenine metabolites in patients affected by schizophrenia. Serum XA levels were largely reduced in a large cohort of patients affected by schizophrenia, and, in patients with first-episode schizophrenia, levels remained low after 12 months of antipsychotic medication. As opposed to other kynurenine metabolites, XA levels were also significantly reduced in first-degree relatives of patients affected by schizophrenia. We suggest that lowered serum XA levels might represent a novel trait marker for schizophrenia.
Collapse
Affiliation(s)
| | - Luana Lionetto
- Advanced Molecular Diagnostics Unit, Sant’Andrea Hospital, Rome, Italy
| | - Martina Curto
- School of Medicine and Psychology NESMOS Department, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| | - Luisa Iacovelli
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Michele Cavallari
- School of Medicine and Psychology NESMOS Department, Sant’Andrea Hospital, Sapienza University, Rome, Italy
- Center for Neurological Imaging, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, U.S.A
| | | | - Martina Ulivieri
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Flavia Napoletano
- School of Medicine and Psychology NESMOS Department, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| | - Matilde Capi
- School of Medicine and Psychology NESMOS Department, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| | - Valentina Corigliano
- School of Medicine and Psychology NESMOS Department, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| | - Sergio Scaccianoce
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Alessandra Caruso
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Jessica Miele
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | | | | | - Anna Comparelli
- School of Medicine and Psychology NESMOS Department, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| | - Antonella De Carolis
- School of Medicine and Psychology NESMOS Department, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| | - Roberto Gradini
- I.R.C.C.S. Neuromed, Pozzilli, Italy
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Robert Nisticò
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
- I.R.C.C.S. Fondazione Santa Lucia, Rome, Italy
| | - Antonio De Blasi
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Paolo Girardi
- School of Medicine and Psychology NESMOS Department, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| | - Valeria Bruno
- I.R.C.C.S. Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | | | - Ferdinando Nicoletti
- I.R.C.C.S. Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Maurizio Simmaco
- Advanced Molecular Diagnostics Unit, Sant’Andrea Hospital, Rome, Italy
- School of Medicine and Psychology NESMOS Department, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| |
Collapse
|
38
|
Müller N, Weidinger E, Leitner B, Schwarz MJ. The role of inflammation in schizophrenia. Front Neurosci 2015; 9:372. [PMID: 26539073 PMCID: PMC4612505 DOI: 10.3389/fnins.2015.00372] [Citation(s) in RCA: 302] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 09/28/2015] [Indexed: 12/16/2022] Open
Abstract
High levels of pro-inflammatory substances such as cytokines have been described in the blood and cerebrospinal fluid of schizophrenia patients. Animal models of schizophrenia show that under certain conditions an immune disturbance during early life, such as an infection-triggered immune activation, might trigger lifelong increased immune reactivity. A large epidemiological study clearly demonstrated that severe infections and autoimmune disorders are risk factors for schizophrenia. Genetic studies have shown a strong signal for schizophrenia on chromosome 6p22.1, in a region related to the human leucocyte antigen (HLA) system and other immune functions. Another line of evidence demonstrates that chronic (dis)stress is associated with immune activation. The vulnerability-stress-inflammation model of schizophrenia includes the contribution of stress on the basis of increased genetic vulnerability for the pathogenesis of schizophrenia, because stress may increase pro-inflammatory cytokines and even contribute to a lasting pro-inflammatory state. Immune alterations influence the dopaminergic, serotonergic, noradrenergic, and glutamatergic neurotransmission. The activated immune system in turn activates the enzyme indoleamine 2,3-dioxygenase (IDO) of the tryptophan/kynurenine metabolism which influences the serotonergic and glutamatergic neurotransmission via neuroactive metabolites such as kynurenic acid. The described loss of central nervous system volume and the activation of microglia, both of which have been clearly demonstrated in neuroimaging studies of schizophrenia patients, match the assumption of a (low level) inflammatory neurotoxic process. Further support for the inflammatory hypothesis comes from the therapeutic benefit of anti-inflammatory medication. Metaanalyses have shown an advantageous effect of cyclo-oxygenase-2 inhibitors in early stages of schizophrenia. Moreover, intrinsic anti-inflammatory, and immunomodulatory effects of antipsychotic drugs are known since a long time. Anti-inflammatory effects of antipsychotics, therapeutic effects of anti-inflammtory compounds, genetic, biochemical, and immunological findings point to a major role of inflammation in schizophrenia.
Collapse
Affiliation(s)
- Norbert Müller
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Munich, Germany
| | - Elif Weidinger
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Munich, Germany
| | - Bianka Leitner
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Munich, Germany
| | - Markus J Schwarz
- Department of Laboratory Medicine, Ludwig Maximilian University Munich, Germany
| |
Collapse
|
39
|
Elevated baseline serum glutamate as a pharmacometabolomic biomarker for acamprosate treatment outcome in alcohol-dependent subjects. Transl Psychiatry 2015; 5:e621. [PMID: 26285131 PMCID: PMC4564571 DOI: 10.1038/tp.2015.120] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 06/04/2015] [Accepted: 07/12/2015] [Indexed: 12/20/2022] Open
Abstract
Acamprosate has been widely used since the Food and Drug Administration approved the medication for treatment of alcohol use disorders (AUDs) in 2004. Although the detailed molecular mechanism of acamprosate remains unclear, it has been largely known that acamprosate inhibits glutamate action in the brain. However, AUD is a complex and heterogeneous disorder. Thus, biomarkers are required to prescribe this medication to patients who will have the highest likelihood of responding positively. To identify pharmacometabolomic biomarkers of acamprosate response, we utilized serum samples from 120 alcohol-dependent subjects, including 71 responders (maintained continuous abstinence) and 49 non-responders (any alcohol use) during 12 weeks of acamprosate treatment. Notably, baseline serum glutamate levels were significantly higher in responders compared with non-responders. Importantly, serum glutamate levels of responders are normalized after acamprosate treatment, whereas there was no significant glutamate change in non-responders. Subsequent functional studies in animal models revealed that, in the absence of alcohol, acamprosate activates glutamine synthetase, which synthesizes glutamine from glutamate and ammonia. These results suggest that acamprosate reduces serum glutamate levels for those who have elevated baseline serum glutamate levels among responders. Taken together, our findings demonstrate that elevated baseline serum glutamate levels are a potential biomarker associated with positive acamprosate response, which is an important step towards development of a personalized approach to treatment for AUD.
Collapse
|
40
|
Fond G, d'Albis MA, Jamain S, Tamouza R, Arango C, Fleischhacker WW, Glenthøj B, Leweke M, Lewis S, McGuire P, Meyer-Lindenberg A, Sommer IE, Winter-van Rossum I, Kapur S, Kahn RS, Rujescu D, Leboyer M. The promise of biological markers for treatment response in first-episode psychosis: a systematic review. Schizophr Bull 2015; 41:559-73. [PMID: 25759473 PMCID: PMC4393702 DOI: 10.1093/schbul/sbv002] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Successful treatment of first-episode psychosis is one of the major factors that impacts long-term prognosis. Currently, there are no satisfactory biological markers (biomarkers) to predict which patients with a first-episode psychosis will respond to which treatment. In addition, a non-negligible rate of patients does not respond to any treatment or may develop side effects that affect adherence to the treatments as well as negatively impact physical health. Thus, there clearly is a pressing need for defining biomarkers that may be helpful to predict response to treatment and sensitivity to side effects in first-episode psychosis. The present systematic review provides (1) trials that assessed biological markers associated with antipsychotic response or side effects in first-episode psychosis and (2) potential biomarkers associated with biological disturbances that may guide the choice of conventional treatments or the prescription of innovative treatments. Trials including first-episode psychoses are few in number. Most of the available data focused on pharmacogenetics markers with so far only preliminary results. To date, these studies yielded-beside markers for metabolism of antipsychotics-no or only a few biomarkers for response or side effects, none of which have been implemented in daily clinical practice. Other biomarkers exploring immunoinflammatory, oxidative, and hormonal disturbances emerged as biomarkers of first-episode psychoses in the last decades, and some of them have been associated with treatment response. In addition to pharmacogenetics, further efforts should focus on the association of emergent biomarkers with conventional treatments or with innovative therapies efficacy, where some preliminary data suggest promising results.
Collapse
Affiliation(s)
| | | | | | - Ryad Tamouza
- Jean Dausset Laboratory & INSERM, UMRS 940, Hôpital Saint Louis, Paris, France
| | - Celso Arango
- Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón, IiSGM, School of Medicine, Universidad Complutense, CIBERSAM, Madrid, Spain
| | | | - Birte Glenthøj
- Center for Neuropsychiatric Schizophrenia Research & Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Psychiatric Hospital Center Glostrup, University of Copenhagen, Faculty of Health and Medical Sciences, Denmark
| | - Markus Leweke
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty Mannheim, Mannheim, Germany
| | - Shôn Lewis
- Institute of Brain, Behaviour and Mental Health, University of Manchester, Manchester, UK
| | - Phillip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, King's College London, London, UK
| | - Andreas Meyer-Lindenberg
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty Mannheim, Mannheim, Germany
| | - Iris E Sommer
- Department of Psychiatry, Brain Center Rudolf Magnus, UMC Utrecht, Utrecht, The Netherlands
| | - Inge Winter-van Rossum
- Department of Psychiatry, Brain Center Rudolf Magnus, UMC Utrecht, Utrecht, The Netherlands
| | - Shitij Kapur
- Institute of Psychiatry, King's College London, London, UK
| | - René S Kahn
- Department of Psychiatry, Brain Center Rudolf Magnus, UMC Utrecht, Utrecht, The Netherlands
| | - Dan Rujescu
- Department of Psychiatry, University of Halle, Halle, Germany
| | | |
Collapse
|
41
|
Everett JR. Pharmacometabonomics in humans: a new tool for personalized medicine. Pharmacogenomics 2015; 16:737-54. [PMID: 25929853 DOI: 10.2217/pgs.15.20] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Pharmacogenomics is now over 50 years old and has had some impact in clinical practice, through its use to select patient subgroups who will enjoy efficacy without side effects when treated with certain drugs. However, pharmacogenomics, has had less impact than initially predicted. One reason for this is that many diseases, and the way in which the patients respond to drug treatments, have both genetic and environmental elements. Pure genomics is almost blind to the environmental elements. A new methodology has emerged, termed pharmacometabonomics that is concerned with the prediction of drug effects through the analysis of predose, biofluid metabolite profiles, which reflect both genetic and environmental influences on human physiology. In this review we will cover what pharmacometabonomics is, how it works, what applications exist and what the future might hold in this exciting new area.
Collapse
|
42
|
Parrott JM, O'Connor JC. Kynurenine 3-Monooxygenase: An Influential Mediator of Neuropathology. Front Psychiatry 2015; 6:116. [PMID: 26347662 PMCID: PMC4542134 DOI: 10.3389/fpsyt.2015.00116] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/03/2015] [Indexed: 12/13/2022] Open
Abstract
Mounting evidence demonstrates that kynurenine metabolism may play an important pathogenic role in the development of multiple neurological and neuropsychiatric disorders. The kynurenine pathway consists of two functionally distinct branches that generate both neuroactive and oxidatively reactive metabolites. In the brain, the rate-limiting enzyme for one of these branches, kynurenine 3-monooxygenase (KMO), is predominantly expressed in microglia and has emerged as a pivotal point of metabolic regulation. KMO substrate and expression levels are upregulated by pro-inflammatory cytokines and altered by functional genetic mutations. Increased KMO metabolism results in the formation of metabolites that activate glutamate receptors and elevate oxidative stress, while recent evidence has revealed neurodevelopmental consequences of reduced KMO activity. Together, the evidence suggests that KMO is positioned at a critical metabolic junction to influence the development or trajectory of a myriad of neurological diseases. Understanding the mechanism(s) by which alterations in KMO activity are able to impair neuronal function, and viability will enhance our knowledge of related disease pathology and provide insight into novel therapeutic opportunities. This review will discuss the influence of KMO on brain kynurenine metabolism and the current understanding of molecular mechanisms by which altered KMO activity may contribute to neurodevelopment, neurodegenerative, and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Jennifer M Parrott
- Department of Pharmacology, School of Medicine, University of Texas Health Science Center at San Antonio , San Antonio, TX , USA ; Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio , San Antonio, TX , USA
| | - Jason C O'Connor
- Department of Pharmacology, School of Medicine, University of Texas Health Science Center at San Antonio , San Antonio, TX , USA ; Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio , San Antonio, TX , USA ; Mood Disorders Translational Research Core, University of Texas Health Science Center at San Antonio , San Antonio, TX , USA ; Audie L. Murphy Memorial VA Hospital, South Texas Veterans Health System , San Antonio, TX , USA
| |
Collapse
|
43
|
van Beveren NJM, Schwarz E, Noll R, Guest PC, Meijer C, de Haan L, Bahn S. Evidence for disturbed insulin and growth hormone signaling as potential risk factors in the development of schizophrenia. Transl Psychiatry 2014; 4:e430. [PMID: 25158005 PMCID: PMC4150237 DOI: 10.1038/tp.2014.52] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 05/07/2014] [Accepted: 05/21/2014] [Indexed: 01/03/2023] Open
Abstract
Molecular abnormalities in metabolic, hormonal and immune pathways are present in peripheral body fluids of a significant subgroup of schizophrenia patients. The authors have tested whether such disturbances also occur in psychiatrically ill and unaffected siblings of schizophrenia patients with the aim of identifying potential contributing factors to disease vulnerability. The subjects were recruited as part of the Genetic Risk and OUtcome of Psychosis (GROUP) study. The authors used multiplexed immunoassays to measure the levels of 184 molecules in serum from 112 schizophrenia patients, 133 siblings and 87 unrelated controls. Consistent with the findings of previous studies, serum from schizophrenia patients contained higher levels of insulin, C-peptide and proinsulin, decreased levels of growth hormone and altered concentrations of molecules involved in inflammation. In addition, significant differences were found in the levels of some of these proteins in siblings diagnosed with mood disorders (n=16) and in unaffected siblings (n=117). Most significantly, the insulin/growth hormone ratio was higher across all groups compared with the controls. Taken together, these findings suggest the presence of a molecular endophenotype involving disruption of insulin and growth factor signaling pathways as an increased risk factor for schizophrenia.
Collapse
Affiliation(s)
- N J M van Beveren
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands,Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands,Department 'Nieuwe Kennis', Delta Center for Mental Health Care, Rotterdam, The Netherlands,Department of Neuroscience, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands. E-mail: or
| | - E Schwarz
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - R Noll
- Department of Psychology, DeSales University, Center Valley, PA, USA
| | - P C Guest
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - C Meijer
- Department of Psychiatry, Academic Medical Center, Amsterdam, The Netherlands
| | - L de Haan
- Department of Psychiatry, Academic Medical Center, Amsterdam, The Netherlands
| | - S Bahn
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands,Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK,Department of Neuroscience, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands. E-mail: or
| |
Collapse
|
44
|
Chiappelli J, Pocivavsek A, Nugent KL, Notarangelo FM, Kochunov P, Rowland LM, Schwarcz R, Hong LE. Stress-induced increase in kynurenic acid as a potential biomarker for patients with schizophrenia and distress intolerance. JAMA Psychiatry 2014; 71:761-8. [PMID: 24806441 PMCID: PMC4219570 DOI: 10.1001/jamapsychiatry.2014.243] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
IMPORTANCE Several lines of evidence have linked the endogenous neuromodulator kynurenic acid (KYNA) to schizophrenia. The pathophysiology of schizophrenia is commonly associated with stress, and stress plays a key regulatory role in the first, rate-limiting step of the kynurenine pathway, which produces KYNA. OBJECTIVE To determine whether the level of KYNA changes following psychological stress and whether this change is associated with stress-related behavior. DESIGN, SETTING, AND PARTICIPANTS The KYNA level was measured in saliva samples taken at baseline and at 2 times following a laboratory-based psychological stress challenge in 128 participants (64 patients with schizophrenia from outpatient clinics and 64 healthy controls from the community). EXPOSURE Laboratory-based psychological stress challenge. MAIN OUTCOMES AND MEASURES Quitting the stressful task early was used as a behavioral marker of distress intolerance. RESULTS Patients with schizophrenia showed a significantly higher rate of distress intolerance compared with healthy controls (P = .003). Salivary KYNA levels increased significantly between baseline and 20 minutes following the stress task in both patients and controls (mean [SEM], 6.72nM [0.65nM] vs 8.43nM [1.05nM], respectively; P = .007). Patients who were unable to tolerate the stressful tasks and quit early showed significantly higher levels of KYNA than patients who tolerated the psychological stressor (P = .02) or healthy controls (P = .02). In patients with distress intolerance, KYNA elevation significantly correlated with the severity of clinical symptoms (ρ = 0.64; P = .008). CONCLUSIONS AND RELEVANCE Distress intolerance is more common in patients with schizophrenia. Patients with this behavioral phenotype have elevated salivary KYNA levels. This stress response behavior-linked biomarker may aid heterogeneity reduction in schizophrenia and other stress-related psychiatric conditions.
Collapse
Affiliation(s)
- Joshua Chiappelli
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore
| | - Ana Pocivavsek
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore
| | - Katie L. Nugent
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore
| | - Francesca M. Notarangelo
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore
| | - Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore
| | - Laura M. Rowland
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore
| | - L. Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore
| |
Collapse
|
45
|
Abstract
Increased proinflammatory markers like cytokines have been described in the blood and cerebrospinal fluid of patients suffering from schizophrenia. Animal models have shown that a hit in early life to the immune system might trigger a lifelong increased immune reactivity. Many epidemiological and clinical studies show the role of various infectious agents as risk factors for schizophrenia with overlap to other psychoses. The first large-scale epidemiological study in psychiatry from Denmark clearly demonstrates severe infections and autoimmune disorders during lifetime to be risk factors for schizophrenia. Genetic studies have shown the strongest signal for schizophrenia on chromosome 6p22.1, in a region related to the major histocompatibility complex and other immune functions. The vulnerability-stress-inflammation model is important as stress may increase proinflammatory cytokines and even contribute to a lasting proinflammatory state. The immune system itself is considered an important further piece in the puzzle, as in autoimmune disorders in general, which are always linked to three factors: genes, the environment and the immune system. Alterations of dopaminergic, serotonergic, noradrenergic and glutamatergic neurotransmission have been shown with low-level neuroinflammation and may directly be involved in the generation of schizophrenic symptoms. Loss of central nervous system volume and microglial activation has been demonstrated in schizophrenia in neuroimaging studies, which supports the assumption of a low-level neuroinflammatory process. Further support comes from the therapeutic benefit of anti-inflammatory medications in specific studies and the anti-inflammatory and immunomodulatory intrinsic effects of antipsychotics.
Collapse
Affiliation(s)
- Norbert Müller
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilian University of Munich, Munich, Germany
| |
Collapse
|
46
|
Altamura AC, Buoli M, Pozzoli S. Role of immunological factors in the pathophysiology and diagnosis of bipolar disorder: comparison with schizophrenia. Psychiatry Clin Neurosci 2014; 68:21-36. [PMID: 24102953 DOI: 10.1111/pcn.12089] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 04/05/2013] [Accepted: 05/26/2013] [Indexed: 01/02/2023]
Abstract
Several lines of evidence point to the key role of neurobiological mechanisms and shared genetic background in schizophrenia and bipolar disorder. For both disorders, neurodevelopmental and neurodegenerative processes have been postulated to be relevant for the pathogenesis as well as dysregulation of immuno-inflammatory pathways. Inflammation is a complex biological response to harmful stimuli and it is mediated by cytokines cascades, cellular immune responses, oxidative factors and hormone regulation. Cytokines, in particular, are supposed to play a critical role in infectious and inflammatory processes, mediating the cross-talk between the brain and the immune system; they also possibly contribute to the development of the central nervous system. From this perspective, even though mixed results have been reported, it seems that both schizophrenia and bipolar disorder are associated with an imbalance in inflammatory cytokines; in fact, some of these could represent biological markers of illness and could be possible targets for pharmacological treatments. In light of these considerations, the purpose of the present paper was to provide a comprehensive and critical review of the existing literature about immunological abnormalities in bipolar disorder with particular attention to the similarities and differences with schizophrenia.
Collapse
|
47
|
Kaddurah-Daouk R, Weinshilboum RM. Pharmacometabolomics: Implications for Clinical Pharmacology and Systems Pharmacology. Clin Pharmacol Ther 2013; 95:154-67. [DOI: 10.1038/clpt.2013.217] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 10/28/2013] [Indexed: 12/24/2022]
|
48
|
Activation of kynurenine pathway in ex vivo fibroblasts from patients with bipolar disorder or schizophrenia: cytokine challenge increases production of 3-hydroxykynurenine. J Psychiatr Res 2013; 47:1815-23. [PMID: 24012176 DOI: 10.1016/j.jpsychires.2013.08.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 08/10/2013] [Accepted: 08/14/2013] [Indexed: 02/02/2023]
Abstract
Accumulating data suggest a causative link between immune stimulation, disturbed metabolism of tryptophan, and pathogenesis of bipolar disorder and schizophrenia. The goal of this study was to examine the production of kynurenic acid (KYNA), 3-hydroxykynurenine (3-HK) and the expression of kynurenine pathway enzymes involved in their synthesis and metabolism in cultured skin fibroblasts obtained from patients with bipolar disorder, schizophrenia or from healthy control individuals. The assessment was performed under basal conditions or following treatment with interferon (IFN)-γ, tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, or their combinations, in cells exposed to exogenous kynurenine. In both groups of patients, the baseline production of KYNA and 3-HK was increased, as compared to control subjects. Case-treatment analyses revealed significant interactions between bipolar case status and IL-1β, IL-6, IFN-γ + TNF-α, or IFN-γ + IL-1β, as well as between schizophrenia case status and IL-1β, IFN-γ + TNF-α, or IFN-γ + IL-1β, in terms of higher 3-HK. Noteworthy, no case-treatment interactions in terms of KYNA production were found. Observed changes did not appear to correlate with the expression of genes encoding kynurenine aminotransferases (KATs), kynureninase (KYNU) or kynurenine-3-monooxygenase (KMO). The single nucleotide polymorphisms (SNPs), rs1053230 and rs2275163, in KMO influenced KYNA levels yet did not explain the case-treatment discrepancies. In conclusion, our present findings indicate the utility of skin-derived fibroblasts for kynurenines research and support the concept of kynurenine pathway alterations in bipolar disorder and schizophrenia. The increase in ratio between neurotoxic 3-HK and neuroinhibitory/neuroprotective KYNA following exposure to cytokines may account for altered neurogenesis and structural abnormalities characteristic for both diseases.
Collapse
|
49
|
Abstract
BACKGROUND Pharmacometabonomics is a new branch of science, first described in 2006 and defined as 'the prediction of the effects of a drug on the basis of a mathematical model of pre-dose metabolite profiles'. Pharmacometabonomics has been used to predict drug metabolism, pharmacokinetics (PK), drug safety and drug efficacy in both animals and humans and is complementary to both pharmacogenomics (PGx) and pharmacoproteomics. METHODS A literature review using the search terms pharmacometabonomics, pharmacometabolomics, pharmaco-metabonomics, pharmaco-metabolomics and the singular form of all those terms was conducted in October 2012 using PubMed and Web of Science. The review was updated until mid April 2013. RESULTS Since the original description of pharmacometabonomics in 2006, 21 original publications and eight reviews have emerged, covering a broad range of applications from the prediction of PK to the prediction of drug metabolism, efficacy and safety in humans and animals. CONCLUSIONS Pharmacometabonomics promises to be an important new approach to the delivery of personalized medicine to improve both drug efficacy and safety for patients in the future. Pharmacometabonomics is particularly powerful as it is sensitive to both genetic and environmental factors such as diet, drug intake and most importantly, a person's microbiome. PGx is now over 50 years old and although it has not achieved as much as some hoped, it is starting to have important applications in personalized medicine. We predict that pharmacometabonomics will be equally important in the next few decades and will be both valuable in its own right and complementary to pharmacoproteomics and PGx.
Collapse
Affiliation(s)
- Jeremy R Everett
- Medway Metabonomics Research Group, School of Science, University of Greenwich, Chatham Maritime, UK
| | | | | |
Collapse
|
50
|
Motsinger-Reif AA, Zhu H, Kling MA, Matson W, Sharma S, Fiehn O, Reif DM, Appleby DH, Doraiswamy PM, Trojanowski JQ, Kaddurah-Daouk R, Arnold SE. Comparing metabolomic and pathologic biomarkers alone and in combination for discriminating Alzheimer's disease from normal cognitive aging. Acta Neuropathol Commun 2013; 1:28. [PMID: 24252434 PMCID: PMC3893491 DOI: 10.1186/2051-5960-1-28] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 06/07/2013] [Indexed: 01/24/2023] Open
Abstract
Background A critical and as-yet unmet need in Alzheimer disease (AD) research is the development of novel markers that can identify individuals at risk for cognitive decline due to AD. This would aid intervention trials designed to slow the progression of AD by increasing diagnostic certainty, and provide new pathophysiologic clues and potential drug targets. Results We used two metabolomics platforms (gas chromatography-time of flight mass spectrometry [GC-TOF] and liquid chromatography LC-ECA array [LC-ECA]) to measure a number of metabolites in cerebrospinal fluid (CSF) from patients with AD dementia and from cognitively normal controls. We used stepwise logistic regression models with cross-validation to assess the ability of metabolite markers to discriminate between clinically diagnosed AD participants and cognitively normal controls and we compared these data with traditional CSF Luminex immunoassay amyloid-β and tau biomarkers. Aβ and tau biomarkers had high accuracy to discriminate cases and controls (testing area under the curve: 0.92). The accuracy of GC-TOF metabolites and LC-ECA metabolites by themselves to discriminate clinical AD participants from controls was high (testing area under the curve: 0.70 and 0.96, respectively). Conclusions Our study identified several CSF small-molecule metabolites that discriminated especially well between clinically diagnosed AD and control groups. They appear to be suitable for further confirmatory and validation studies, and show the potential to provide predictive performance for AD.
Collapse
|