1
|
Li S, Liu Y, Lu S, Xu J, Liu X, Yang D, Yang Y, Hou L, Li N. A crazy trio in Parkinson's disease: metabolism alteration, α-synuclein aggregation, and oxidative stress. Mol Cell Biochem 2025; 480:139-157. [PMID: 38625515 DOI: 10.1007/s11010-024-04985-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 03/06/2024] [Indexed: 04/17/2024]
Abstract
Parkinson's disease (PD) is an aging-associated neurodegenerative disorder, characterized by the progressive loss of dopaminergic neurons in the pars compacta of the substantia nigra and the presence of Lewy bodies containing α-synuclein within these neurons. Oligomeric α-synuclein exerts neurotoxic effects through mitochondrial dysfunction, glial cell inflammatory response, lysosomal dysfunction and so on. α-synuclein aggregation, often accompanied by oxidative stress, is generally considered to be a key factor in PD pathology. At present, emerging evidences suggest that metabolism alteration is closely associated with α-synuclein aggregation and PD progression, and improvement of key molecules in metabolism might be potentially beneficial in PD treatment. In this review, we highlight the tripartite relationship among metabolic changes, α-synuclein aggregation, and oxidative stress in PD, and offer updated insights into the treatments of PD, aiming to deepen our understanding of PD pathogenesis and explore new therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Sheng Li
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yanbing Liu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Sen Lu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jiayi Xu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiaokun Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Di Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Yuxuan Yang
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Lin Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Ning Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
2
|
Shukla A, Meena K, Gupta A, Sandhir R. 1H NMR-Based Metabolomic Signatures in Rodent Models of Sporadic Alzheimer's Disease and Metabolic Disorders. ACS Chem Neurosci 2024; 15:4478-4499. [PMID: 39629865 DOI: 10.1021/acschemneuro.4c00510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024] Open
Abstract
Alzheimer's disease (AD) is a chronic neurological disorder that impacts the elderly population all over the globe. Evidence suggests association between AD and metabolic disorders such as diabetes mellitus (DM) and obesity (OB). The present study is an attempt to evaluate metabolic alterations in the serum and brain through NMR spectroscopy with the aim to identify shared metabolic signatures. AD was induced in rats by stereotactic intracerebroventricular injection of oligomerized Aβ-42 peptide into the brain. DM and OB were induced by intraperitoneal injection of streptozotocin and feeding rats on a high-fat diet, respectively. The metabolic alterations obtained through 1H NMR spectroscopy were further subjected to multivariate analysis by principal component analysis and partial least-squares discrimination for identification of metabolic signatures. In the serum, the levels of lactate and betaine were increased in AD, DM, and OB rats. On the other hand, the metabolite profile of brain indicated increase in the levels of lactate, N-acetylaspartate, and creatinine in AD, DM, and OB rats. Additionally, the concentration of neurochemicals such as glutamate, GABA, N-acetylglutamate, and myo-inositol were also elevated. The alterations in neurotransmitters and cerebral energy metabolism were accompanied by deficits in cognition assessed by Morris water maze in AD, DM, and OB rats. The perturbed metabolic profiles were accompanied by the presence of pathogenic amyloid deposits visualized by Congo red stain in the brains of AD, DM, and OB rats. Overall, the study identifies common metabolic signatures in AD, DM, and OB that may be involved in etiopathogenesis and also suggests linkages between these three conditions.
Collapse
Affiliation(s)
- Ananya Shukla
- Department of Biochemistry, Hargobind Khorana Block (BMS Block II), Panjab University, Sector-25, Chandigarh 160014, India
| | - Khushbhu Meena
- Centre of Bio-Medical Research, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS) Campus, Lucknow, Uttar Pradesh 226014, India
| | - Ashish Gupta
- Centre of Bio-Medical Research, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS) Campus, Lucknow, Uttar Pradesh 226014, India
| | - Rajat Sandhir
- Department of Biochemistry, Hargobind Khorana Block (BMS Block II), Panjab University, Sector-25, Chandigarh 160014, India
| |
Collapse
|
3
|
Morais LH, Boktor JC, MahmoudianDehkordi S, Kaddurah-Daouk R, Mazmanian SK. α-synuclein overexpression and the microbiome shape the gut and brain metabolome in mice. NPJ Parkinsons Dis 2024; 10:208. [PMID: 39477976 PMCID: PMC11525669 DOI: 10.1038/s41531-024-00816-w] [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: 06/07/2024] [Accepted: 10/10/2024] [Indexed: 11/02/2024] Open
Abstract
Pathological forms of α-synuclein contribute to synucleinopathies, including Parkinson's disease (PD). Most cases of PD arise from gene-environment interactions. Microbiome composition is altered in PD, and gut bacteria are causal to symptoms in animal models. We quantitatively profiled nearly 630 metabolites in the gut, plasma, and brain of α-synuclein-overexpressing (ASO) mice, compared to wild-type (WT) animals, and comparing germ-free (GF) to specific pathogen-free (SPF) animals (n = 5 WT-SPF; n = 6 ASO-SPF; n = 6 WT-GF; n = 6 ASO-GF). Many differentially expressed metabolites in ASO mice are also dysregulated in human PD patients, including amine oxides, bile acids and indoles. The microbial metabolite trimethylamine N-oxide (TMAO) strongly correlates from the gut to the plasma to the brain in mice, notable since TMAO is elevated in the blood and cerebrospinal fluid of PD patients. These findings uncover broad metabolomic changes that are influenced by the intersection of host genetics and microbiome in a mouse model of PD.
Collapse
Affiliation(s)
- Livia H Morais
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Joseph C Boktor
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | | | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA.
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA.
- Department of Medicine, Duke University, Durham, NC, USA.
| | - Sarkis K Mazmanian
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
4
|
Pocivavsek A, Schwarcz R, Erhardt S. Neuroactive Kynurenines as Pharmacological Targets: New Experimental Tools and Exciting Therapeutic Opportunities. Pharmacol Rev 2024; 76:978-1008. [PMID: 39304346 PMCID: PMC11549936 DOI: 10.1124/pharmrev.124.000239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
Both preclinical and clinical studies implicate functional impairments of several neuroactive metabolites of the kynurenine pathway (KP), the major degradative cascade of the essential amino acid tryptophan in mammals, in the pathophysiology of neurologic and psychiatric diseases. A number of KP enzymes, such as tryptophan 2,3-dioxygenase (TDO2), indoleamine 2,3-dioxygenases (IDO1 and IDO2), kynurenine aminotransferases (KATs), kynurenine 3-monooxygenase (KMO), 3-hydroxyanthranilic acid oxygenase (3-HAO), and quinolinic acid phosphoribosyltransferase (QPRT), control brain KP metabolism in health and disease and are therefore increasingly considered to be promising targets for the treatment of disorders of the nervous system. Understanding the distribution, cellular expression, and regulation of KP enzymes and KP metabolites in the brain is therefore critical for the conceptualization and implementation of successful therapeutic strategies. SIGNIFICANCE STATEMENT: Studies have implicated the kynurenine pathway of tryptophan in the pathophysiology of neurologic and psychiatric diseases. Key enzymes of the kynurenine pathway regulate brain metabolism in both health and disease, making them promising targets for treating these disorders. Therefore, understanding the distribution, cellular expression, and regulation of these enzymes and metabolites in the brain is critical for developing effective therapeutic strategies. This review endeavors to describe these processes in detail.
Collapse
Affiliation(s)
- Ana Pocivavsek
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina (A.P.); Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland (R.S.); and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.E.)
| | - Robert Schwarcz
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina (A.P.); Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland (R.S.); and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.E.)
| | - Sophie Erhardt
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina (A.P.); Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland (R.S.); and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.E.)
| |
Collapse
|
5
|
Lu S, Ji N, Wang W, Lin X, Gao D, Geng D. Salidroside improves cognitive function in Parkinson's disease via Braf-mediated mitogen‑activated protein kinase signaling pathway. Biomed Pharmacother 2024; 177:116968. [PMID: 38901199 DOI: 10.1016/j.biopha.2024.116968] [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: 01/24/2024] [Revised: 05/31/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024] Open
Abstract
OBJECTIVE To delve into the underlying mechanism of Salidroside (Sal) on the improvement of cognitive function in Parkinson's Disease (PD). METHODS The experimental mice were divided into Control group, Model group [injected with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)], and Model+Sal (low concentration, high concentration) group. Mouse hippocampal tissues were extracted for RNA sequencing to obtain the core pathway and core gene. Mouse plasma was prepared and analyzed by LC-MS to obtain differential metabolites. In vitro experiments were verified by immunofluorescence and lentiviral transduction. RESULTS ELISA signaled that Sal facilitated the reduction of neuronal damage and inflammatory reaction in mice. MPTP_Sal_Low and MPTP_Sal_High groups had high levels of glial cell derived neurotrophie factor (GDNF) expression. Differentially expressed genes (DEGs) in control group, MPTP group and MPTP_Sal_High group were identified by transcriptomic, which were classified to the mitogen-activated protein kinase (MAPK) signaling pathway, and the core gene Braf was obtained. Metabolomics manifested that the differential metabolites involved DL-tyrosine, adenosine, phosphoenolpyruvate, and L-tryptophan. In vitro experiments verified that Sal treatment inhibited the up-regulation of p-p38, p-c-Jun N-terminal kinase (JNK), and p-extracellular signal-regulated kinase (ERK) expression, and growth of neuronal protrusions. The OE-Braf group showed a significant up-regulation of the GDNF expression, a decrease in the expression of p-p38, p-JNK, and p-ERK, and a significant growth of neuronal protrusions. CONCLUSION Sal may exert its effects in PD through the Braf-mediated MAPK signaling pathway, which can increase GDNF expression and promote neuronal protrusion growth for the protection of neurological function and the improvement of cognitive function.
Collapse
Affiliation(s)
- Shujin Lu
- Nanjing Medical University, Nanjing, Jiangsu Province 211166, China; Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, China
| | - Niu Ji
- Nanjing Medical University, Nanjing, Jiangsu Province 211166, China; Department of Neurology, Lianyungang First People's Hospital, Lianyungang, Jiangsu Province 222000, China
| | - Wei Wang
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, China
| | - Xiaoqian Lin
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, China
| | - Dianshuai Gao
- Nanjing Medical University, Nanjing, Jiangsu Province 211166, China; School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China.
| | - Deqin Geng
- Nanjing Medical University, Nanjing, Jiangsu Province 211166, China; Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, China.
| |
Collapse
|
6
|
Hu W, Wang W, Liao H, Bulloch G, Zhang X, Shang X, Huang Y, Hu Y, Yu H, Yang X, He M, Zhu Z. Metabolic profiling reveals circulating biomarkers associated with incident and prevalent Parkinson's disease. NPJ Parkinsons Dis 2024; 10:130. [PMID: 38982064 PMCID: PMC11233508 DOI: 10.1038/s41531-024-00713-2] [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: 09/22/2023] [Accepted: 04/19/2024] [Indexed: 07/11/2024] Open
Abstract
The metabolic profile predating the onset of Parkinson's disease (PD) remains unclear. We aim to investigate the metabolites associated with incident and prevalent PD and their predictive values in the UK Biobank participants with metabolomics and genetic data at the baseline. A panel of 249 metabolites was quantified using a nuclear magnetic resonance analytical platform. PD was ascertained by self-reported history, hospital admission records and death registers. Cox proportional hazard models and logistic regression models were used to investigate the associations between metabolites and incident and prevalent PD, respectively. Area under receiver operating characteristics curves (AUC) were used to estimate the predictive values of models for future PD. Among 109,790 participants without PD at the baseline, 639 (0.58%) individuals developed PD after one year from the baseline during a median follow-up period of 12.2 years. Sixty-eight metabolites were associated with incident PD at nominal significance (P < 0.05), spanning lipids, lipid constituent of lipoprotein subclasses and ratios of lipid constituents. After multiple testing corrections (P < 9 × 10-4), polyunsaturated fatty acids (PUFA) and omega-6 fatty acids remained significantly associated with incident PD, and PUFA was shared by incident and prevalent PD. Additionally, 14 metabolites were exclusively associated with prevalent PD, including amino acids, fatty acids, several lipoprotein subclasses and ratios of lipids. Adding these metabolites to the conventional risk factors yielded a comparable predictive performance to the risk-factor-based model (AUC = 0.766 vs AUC = 0.768, P = 0.145). Our findings suggested metabolic profiles provided additional knowledge to understand different pathways related to PD before and after its onset.
Collapse
Affiliation(s)
- Wenyi Hu
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Centre for Eye Research Australia; Ophthalmology, University of Melbourne, Melbourne, VIC, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, VIC, Australia
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Huan Liao
- Neural Regeneration Group, Institute of Reconstructive Neurobiology, University of Bonn, Bonn, Germany
| | - Gabriella Bulloch
- Centre for Eye Research Australia; Ophthalmology, University of Melbourne, Melbourne, VIC, Australia
| | - Xiayin Zhang
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xianwen Shang
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Centre for Eye Research Australia; Ophthalmology, University of Melbourne, Melbourne, VIC, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, VIC, Australia
| | - Yu Huang
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yijun Hu
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Honghua Yu
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiaohong Yang
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Mingguang He
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, VIC, Australia.
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong SAR, China.
- Research Centre for SHARP Vision, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China.
| | - Zhuoting Zhu
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Centre for Eye Research Australia; Ophthalmology, University of Melbourne, Melbourne, VIC, Australia.
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
7
|
Morais LH, Boktor JC, MahmoudianDehkordi S, Kaddurah-Daouk R, Mazmanian SK. α-Synuclein Overexpression and the Microbiome Shape the Gut and Brain Metabolome in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.597975. [PMID: 38915679 PMCID: PMC11195096 DOI: 10.1101/2024.06.07.597975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Pathological forms of the protein α-synuclein contribute to a family of disorders termed synucleinopathies, which includes Parkinson's disease (PD). Most cases of PD are believed to arise from gene-environment interactions. Microbiome composition is altered in PD, and gut bacteria are causal to symptoms and pathology in animal models. To explore how the microbiome may impact PD-associated genetic risks, we quantitatively profiled nearly 630 metabolites from 26 biochemical classes in the gut, plasma, and brain of α-synuclein-overexpressing (ASO) mice with or without microbiota. We observe tissue-specific changes driven by genotype, microbiome, and their interaction. Many differentially expressed metabolites in ASO mice are also dysregulated in human PD patients, including amine oxides, bile acids and indoles. Notably, levels of the microbial metabolite trimethylamine N-oxide (TMAO) strongly correlate from the gut to the plasma to the brain, identifying a product of gene-environment interactions that may influence PD-like outcomes in mice. TMAO is elevated in the blood and cerebral spinal fluid of PD patients. These findings uncover broad metabolomic changes that are influenced by the intersection of host genetics and the microbiome in a mouse model of PD.
Collapse
Affiliation(s)
- Livia H. Morais
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Joseph C. Boktor
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | | | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Sarkis K. Mazmanian
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| |
Collapse
|
8
|
Han L, Chen X, Wang Y, Zhang R, Zhao T, Pu L, Huang Y, Sun H. A machine learning algorithm based on circulating metabolic biomarkers offers improved predictions of neurological diseases. Clin Chim Acta 2024; 558:119671. [PMID: 38621587 DOI: 10.1016/j.cca.2024.119671] [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: 01/04/2024] [Revised: 04/01/2024] [Accepted: 04/10/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND AND AIMS A machine learning algorithm based on circulating metabolic biomarkers for the predictions of neurological diseases (NLDs) is lacking. To develop a machine learning algorithm to compare the performance of a metabolic biomarker-based model with that of a clinical model based on conventional risk factors for predicting three NLDs: dementia, Parkinson's disease (PD), and Alzheimer's disease (AD). MATERIALS AND METHODS The eXtreme Gradient Boosting (XGBoost) algorithm was used to construct a metabolic biomarker-based model (metabolic model), a clinical risk factor-based model (clinical model), and a combined model for the prediction of the three NLDs. Risk discrimination (c-statistic), net reclassification improvement (NRI) index, and integrated discrimination improvement (IDI) index values were determined for each model. RESULTS The results indicate that incorporation of metabolic biomarkers into the clinical model afforded a model with improved performance in the prediction of dementia, AD, and PD, as demonstrated by NRI values of 0.159 (0.039-0.279), 0.113 (0.005-0.176), and 0.201 (-0.021-0.423), respectively; and IDI values of 0.098 (0.073-0.122), 0.070 (0.049-0.090), and 0.085 (0.068-0.101), respectively. CONCLUSION The performance of the model based on circulating NMR spectroscopy-detected metabolic biomarkers was better than that of the clinical model in the prediction of dementia, AD, and PD.
Collapse
Affiliation(s)
- Liyuan Han
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo No 2 Hospital, Ningbo 315000, China; Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo 315000, China
| | - Xi Chen
- Department of Economics, Yale University, USA; Yale Alzheimer's Disease Research Center, Yale University, USA
| | - Yue Wang
- School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu province, China
| | - Ruijie Zhang
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo No 2 Hospital, Ningbo 315000, China; Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo 315000, China
| | - Tian Zhao
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo No 2 Hospital, Ningbo 315000, China; Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo 315000, China
| | - Liyuan Pu
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo No 2 Hospital, Ningbo 315000, China; Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo 315000, China
| | - Yi Huang
- Laboratory of Neurological Diseases and Brain Function, Department of Neurosur-gery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China; Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang 315010, China.
| | - Hongpeng Sun
- School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu province, China.
| |
Collapse
|
9
|
Trimigno A, Holderman NR, Dong C, Boardman KD, Zhao J, O’Day EM. NMR Precision Metabolomics: Dynamic Peak Sum Thresholding and Navigators for Highly Standardized and Reproducible Metabolite Profiling of Clinical Urine Samples. Metabolites 2024; 14:275. [PMID: 38786752 PMCID: PMC11122845 DOI: 10.3390/metabo14050275] [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: 04/09/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Metabolomics, especially urine-based studies, offers incredible promise for the discovery and development of clinically impactful biomarkers. However, due to the unique challenges of urine, a highly precise and reproducible workflow for NMR-based urine metabolomics is lacking. Using 1D and 2D non-uniform sampled (NUS) 1H-13C NMR spectroscopy, we systematically explored how changes in hydration or specific gravity (SG) and pH can impact biomarker discovery. Further, we examined additional sources of error in metabolomics studies and identified Navigator molecules that could monitor for those biases. Adjustment of SG to 1.002-1.02 coupled with a dynamic sum-based peak thresholding eliminates false positives associated with urine hydration and reduces variation in chemical shift. We identified Navigator molecules that can effectively monitor for inconsistencies in sample processing, SG, protein contamination, and pH. The workflow described provides quality assurance and quality control tools to generate high-quality urine metabolomics data, which is the first step in biomarker discovery.
Collapse
|
10
|
Li S, Lin Y, Jones D, Walker DI, Duarte Folle A, Del Rosario I, Yu Y, Zhang K, Keener AM, Bronstein J, Ritz B, Paul KC. Untargeted serum metabolic profiling of diabetes mellitus among Parkinson's disease patients. NPJ Parkinsons Dis 2024; 10:100. [PMID: 38730245 PMCID: PMC11087477 DOI: 10.1038/s41531-024-00711-4] [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: 04/17/2023] [Accepted: 04/16/2024] [Indexed: 05/12/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a common comorbidity among Parkinson's disease (PD) patients. Yet, little is known about dysregulated pathways that are unique in PD patients with T2DM. We applied high-resolution metabolomic profiling in serum samples of 636 PD and 253 non-PD participants recruited from Central California. We conducted an initial discovery metabolome-wide association and pathway enrichment analysis. After adjusting for multiple testing, in positive (or negative) ion mode, 30 (25) metabolic features were associated with T2DM in both PD and non-PD participants, 162 (108) only in PD participants, and 32 (7) only in non-PD participants. Pathway enrichment analysis identified 17 enriched pathways associated with T2DM in both the PD and non-PD participants, 26 pathways only in PD participants, and 5 pathways only in non-PD participants. Several amino acid, nucleic acids, and fatty acid metabolisms were associated with T2DM only in the PD patient group suggesting a possible link between PD and T2DM.
Collapse
Affiliation(s)
- Shiwen Li
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Yuyuan Lin
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Dean Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, USA
| | - Douglas I Walker
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Aline Duarte Folle
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Irish Del Rosario
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Yu Yu
- Center for Health Policy Research, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Keren Zhang
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Adrienne M Keener
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Jeff Bronstein
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Beate Ritz
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Kimberly C Paul
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
11
|
Wang X, Dong T, Li X, Yu W, Jia Z, Liu Y, Yang J. Global biomarker trends in Parkinson's disease research: A bibliometric analysis. Heliyon 2024; 10:e27437. [PMID: 38501016 PMCID: PMC10945172 DOI: 10.1016/j.heliyon.2024.e27437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/11/2023] [Accepted: 02/28/2024] [Indexed: 03/20/2024] Open
Abstract
As the second most common neurodegenerative disease globally, Parkinson's disease (PD) affects millions of people worldwide. In recent years, the scientific publications related to PD biomarker research have exploded, reflecting the growing interest in unraveling the complex pathophysiology of PD. In this study, we aim to use various bibliometric tools to identify key scientific concepts, detect emerging trends, and analyze the global trends and development of PD biomarker research.The research encompasses various stages of biomarker development, including exploration, identification, and multi-modal research. MOVEMENT DISORDERS emerged as the leading journal in terms of publications and citations. Key authors such as Mollenhauer and Salem were identified, while the University of Pennsylvania and USA stood out in collaboration and research output. NEUROSCIENCES emerged as the most important research direction. Key biomarker categories include α-synuclein-related markers, neurotransmitter-related markers, inflammation and immune system-related markers, oxidative stress and mitochondrial function-related markers, and brain imaging-related markers. Furthermore, future trends in PD biomarker research focus on exosomes and plasma biomarkers, miRNA, cerebrospinal fluid biomarkers, machine learning applications, and animal models of PD. These trends contribute to early diagnosis, disease progression monitoring, and understanding the pathological mechanisms of PD.
Collapse
Affiliation(s)
- Xingxin Wang
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Tiantian Dong
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xuhao Li
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Wenyan Yu
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zhixia Jia
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yuanxiang Liu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jiguo Yang
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| |
Collapse
|
12
|
Wang X, Dong T, Li X, Yu W, Jia Z, Liu Y, Yang J. Global biomarker trends in Parkinson's disease research: A bibliometric analysis. Heliyon 2024; 10:e27437. [PMID: 38501016 DOI: 10.1016/j.heliyon.2024.e27437if:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/11/2023] [Accepted: 02/28/2024] [Indexed: 07/25/2024] Open
Abstract
As the second most common neurodegenerative disease globally, Parkinson's disease (PD) affects millions of people worldwide. In recent years, the scientific publications related to PD biomarker research have exploded, reflecting the growing interest in unraveling the complex pathophysiology of PD. In this study, we aim to use various bibliometric tools to identify key scientific concepts, detect emerging trends, and analyze the global trends and development of PD biomarker research.The research encompasses various stages of biomarker development, including exploration, identification, and multi-modal research. MOVEMENT DISORDERS emerged as the leading journal in terms of publications and citations. Key authors such as Mollenhauer and Salem were identified, while the University of Pennsylvania and USA stood out in collaboration and research output. NEUROSCIENCES emerged as the most important research direction. Key biomarker categories include α-synuclein-related markers, neurotransmitter-related markers, inflammation and immune system-related markers, oxidative stress and mitochondrial function-related markers, and brain imaging-related markers. Furthermore, future trends in PD biomarker research focus on exosomes and plasma biomarkers, miRNA, cerebrospinal fluid biomarkers, machine learning applications, and animal models of PD. These trends contribute to early diagnosis, disease progression monitoring, and understanding the pathological mechanisms of PD.
Collapse
Affiliation(s)
- Xingxin Wang
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Tiantian Dong
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xuhao Li
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Wenyan Yu
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zhixia Jia
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yuanxiang Liu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jiguo Yang
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| |
Collapse
|
13
|
Maji S, Samanta J, Natarajan R. Water-Soluble Triazolium Covalent Cages for ATP Sensing. Chemistry 2024; 30:e202303596. [PMID: 38133633 DOI: 10.1002/chem.202303596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/22/2023] [Accepted: 12/22/2023] [Indexed: 12/23/2023]
Abstract
Water-soluble organic cages are attractive targets for their molecular recognition and sensing features of biologically relevant molecules. Here, we have successfully designed and synthesized a pair of water-soluble cationic cages employing click reaction as the fundamental step followed by the N-methylation of the triazole rings. The rigid and shape-persistent 3D hydrophobic cavity, positively charged surface, H-bonding triazolium rings, and excellent water solubility empower both cages to exhibit a superior affinity and selectivity for binding with adenosine-5'-triphosphate (ATP) compared to cyclophanes and other macrocyclic receptors. Both cage molecules (PCC⋅Cl and BCC⋅Cl) can bind a highly emissive dye HPTS (8-hydroxypyrene-1,3,6-trisulfonic acid trisodium salt) to form non-fluorescent complexes. The addition of ATP resulted in the stronger cage⊂ATP complexes with the retention of HPTS emission upon its displacement. The resultant indicator-displacement assay system can efficiently sense and quantify ATP in nanomolar detection limits in buffer solutions and human serum matrix. Spectroscopic and theoretical studies revealed the synergistic effect of π⋅⋅⋅π stacking interaction between the aromatic moiety of the cationic cages and the adenine moiety of ATP, as well as the electrostatic and hydrogen bonding interaction between the phosphate anion of ATP and triazole protons of cages, played the pivotal roles in the sensing process.
Collapse
Affiliation(s)
- Suman Maji
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S C Mullick Road, Kolkata, 700032, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Jayanta Samanta
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S C Mullick Road, Kolkata, 700032, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ramalingam Natarajan
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S C Mullick Road, Kolkata, 700032, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
14
|
Li H, Zeng F, Huang C, Pu Q, Thomas ER, Chen Y, Li X. The potential role of glucose metabolism, lipid metabolism, and amino acid metabolism in the treatment of Parkinson's disease. CNS Neurosci Ther 2024; 30:e14411. [PMID: 37577934 PMCID: PMC10848100 DOI: 10.1111/cns.14411] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/15/2023] Open
Abstract
PURPOSE OF REVIEW Parkinson's disease (PD) is a common neurodegenerative disease, which can cause progressive deterioration of motor function causing muscle stiffness, tremor, and bradykinesia. In this review, we hope to describe approaches that can improve the life of PD patients through modifications of energy metabolism. RECENT FINDINGS The main pathological features of PD are the progressive loss of nigrostriatal dopaminergic neurons and the production of Lewy bodies. Abnormal aggregation of α-synuclein (α-Syn) leading to the formation of Lewy bodies is closely associated with neuronal dysfunction and degeneration. The main causes of PD are said to be mitochondrial damage, oxidative stress, inflammation, and abnormal protein aggregation. Presence of abnormal energy metabolism is another cause of PD. Many studies have found significant differences between neurodegenerative diseases and metabolic decompensation, which has become a biological hallmark of neurodegenerative diseases. SUMMARY In this review, we highlight the relationship between abnormal energy metabolism (Glucose metabolism, lipid metabolism, and amino acid metabolism) and PD. Improvement of key molecules in glucose metabolism, fat metabolism, and amino acid metabolism (e.g., glucose-6-phosphate dehydrogenase, triglycerides, and levodopa) might be potentially beneficial in PD. Some of these metabolic indicators may serve well during the diagnosis of PD. In addition, modulation of these metabolic pathways may be a potential target for the treatment and prevention of PD.
Collapse
Affiliation(s)
- Hangzhen Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| | - Fancai Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| | - Cancan Huang
- Department of DermatologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Qiqi Pu
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| | | | - Yan Chen
- Department of DermatologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Xiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| |
Collapse
|
15
|
Whittle BJ, Izuogu OG, Lowes H, Deen D, Pyle A, Coxhead J, Lawson RA, Yarnall AJ, Jackson MS, Santibanez-Koref M, Hudson G. Early-stage idiopathic Parkinson's disease is associated with reduced circular RNA expression. NPJ Parkinsons Dis 2024; 10:25. [PMID: 38245550 PMCID: PMC10799891 DOI: 10.1038/s41531-024-00636-y] [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: 07/25/2023] [Accepted: 01/08/2024] [Indexed: 01/22/2024] Open
Abstract
Neurodegeneration in Parkinson's disease (PD) precedes diagnosis by years. Early neurodegeneration may be reflected in RNA levels and measurable as a biomarker. Here, we present the largest quantification of whole blood linear and circular RNAs (circRNA) in early-stage idiopathic PD, using RNA sequencing data from two cohorts (PPMI = 259 PD, 161 Controls; ICICLE-PD = 48 PD, 48 Controls). We identified a replicable increase in TMEM252 and LMNB1 gene expression in PD. We identified novel differences in the expression of circRNAs from ESYT2, BMS1P1 and CCDC9, and replicated trends of previously reported circRNAs. Overall, using circRNA as a diagnostic biomarker in PD did not show any clear improvement over linear RNA, minimising its potential clinical utility. More interestingly, we observed a general reduction in circRNA expression in both PD cohorts, accompanied by an increase in RNASEL expression. This imbalance implicates the activation of an innate antiviral immune response and suggests a previously unknown aspect of circRNA regulation in PD.
Collapse
Affiliation(s)
- Benjamin J Whittle
- Wellcome Centre for Mitochondrial Research, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Osagie G Izuogu
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, UK
| | - Hannah Lowes
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Dasha Deen
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Angela Pyle
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Jon Coxhead
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Rachael A Lawson
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Alison J Yarnall
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Michael S Jackson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Gavin Hudson
- Wellcome Centre for Mitochondrial Research, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
16
|
Abdik E, Çakır T. Transcriptome-based biomarker prediction for Parkinson's disease using genome-scale metabolic modeling. Sci Rep 2024; 14:585. [PMID: 38182712 PMCID: PMC10770157 DOI: 10.1038/s41598-023-51034-y] [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: 09/15/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease in the world. Identification of PD biomarkers is crucial for early diagnosis and to develop target-based therapeutic agents. Integrative analysis of genome-scale metabolic models (GEMs) and omics data provides a computational approach for the prediction of metabolite biomarkers. Here, we applied the TIMBR (Transcriptionally Inferred Metabolic Biomarker Response) algorithm and two modified versions of TIMBR to investigate potential metabolite biomarkers for PD. To this end, we mapped thirteen post-mortem PD transcriptome datasets from the substantia nigra region onto Human-GEM. We considered a metabolite as a candidate biomarker if its production was predicted to be more efficient by a TIMBR-family algorithm in control or PD case for the majority of the datasets. Different metrics based on well-known PD-related metabolite alterations, PD-associated pathways, and a list of 25 high-confidence PD metabolite biomarkers compiled from the literature were used to compare the prediction performance of the three algorithms tested. The modified algorithm with the highest prediction power based on the metrics was called TAMBOOR, TrAnscriptome-based Metabolite Biomarkers by On-Off Reactions, which was introduced for the first time in this study. TAMBOOR performed better in terms of capturing well-known pathway alterations and metabolite secretion changes in PD. Therefore, our tool has a strong potential to be used for the prediction of novel diagnostic biomarkers for human diseases.
Collapse
Affiliation(s)
- Ecehan Abdik
- Department of Bioengineering, Gebze Technical University, Kocaeli, Turkey
| | - Tunahan Çakır
- Department of Bioengineering, Gebze Technical University, Kocaeli, Turkey.
| |
Collapse
|
17
|
Vallianatou T, Nilsson A, Bjärterot P, Shariatgorji R, Slijkhuis N, Aerts JT, Jansson ET, Svenningsson P, Andrén PE. Rapid Metabolic Profiling of 1 μL Crude Cerebrospinal Fluid by Matrix-Assisted Laser Desorption/Ionization Mass Spectrometry Imaging Can Differentiate De Novo Parkinson's Disease. Anal Chem 2023; 95:18352-18360. [PMID: 38059473 PMCID: PMC10733901 DOI: 10.1021/acs.analchem.3c02900] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 12/08/2023]
Abstract
Parkinson's disease (PD) is a highly prevalent neurodegenerative disorder affecting the motor system. However, the correct diagnosis of PD and atypical parkinsonism may be difficult with high clinical uncertainty. There is an urgent need to identify reliable biomarkers using high-throughput, molecular-specific methods to improve current diagnostics. Here, we present a matrix-assisted laser desorption/ionization mass spectrometry imaging method that requires minimal sample preparation and only 1 μL of crude cerebrospinal fluid (CSF). The method enables analysis of hundreds of samples in a single experiment while simultaneously detecting numerous metabolites with subppm mass accuracy. To test the method, we analyzed CSF samples from 12 de novo PD patients (that is, newly diagnosed and previously untreated) and 12 age-matched controls. Within the identified molecules, we found neurotransmitters and their metabolites such as γ-aminobutyric acid, 3-methoxytyramine, homovanillic acid, serotonin, histamine, amino acids, and metabolic intermediates. Limits of detection were estimated for multiple neurotransmitters with high linearity (R2 > 0.99) and sensitivity (as low as 16 pg/μL). Application of multivariate classification led to a highly significant (P < 0.001) model of PD prediction with a 100% classification rate, which was further thoroughly validated with a permutation test and univariate analysis. Molecules related to the neuromelanin pathway were found to be significantly increased in the PD group, indicated by their elevated relative intensities compared to the control group. Our method enables rapid detection of PD-related biomarkers in low sample volumes and could serve as a valuable tool in the development of robust PD diagnostics.
Collapse
Affiliation(s)
- Theodosia Vallianatou
- Department
of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science
for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| | - Anna Nilsson
- Department
of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science
for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| | - Patrik Bjärterot
- Department
of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science
for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| | - Reza Shariatgorji
- Department
of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science
for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| | - Nuria Slijkhuis
- Department
of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science
for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| | - Jordan T. Aerts
- Department
of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science
for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| | - Erik T. Jansson
- Department
of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science
for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| | - Per Svenningsson
- Department
of Clinical Neuroscience, Karolinska Institute, Stockholm SE-17177, Sweden
| | - Per E. Andrén
- Department
of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science
for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| |
Collapse
|
18
|
Wittung-Stafshede P. Chemical catalysis by biological amyloids. Biochem Soc Trans 2023; 51:1967-1974. [PMID: 37743793 PMCID: PMC10657172 DOI: 10.1042/bst20230617] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/12/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023]
Abstract
Toxic aggregation of proteins and peptides into amyloid fibers is the basis of several human diseases. In each disease, a particular peptide noncovalently assembles into long thin structures with an overall cross-β fold. Amyloids are not only related to disease: functional amyloids are found in many biological systems and artificial peptide amyloids are developed into novel nanomaterials. Amyloid fibers can act as template for the generation of more amyloids but are considered nonreactive in chemical catalysis. The perception of amyloids as chemically inert species was recently challenged by in vitro work on three human amyloid systems. With the use of model substrates, amyloid-β, α-synuclein and glucagon amyloids were found to catalyze biologically relevant chemical reactions. The detected catalytic activity was much less than that of 'real' enzymes, but like that of designed (synthetic) catalytic amyloids. I here describe the current knowledge around this new activity of natural amyloids and the putative connection to metabolic changes in amyloid diseases. These pioneering studies imply that catalytic activity is an unexplored gain-of-function activity of disease amyloids. In fact, all biological amyloids may harbor intrinsic catalytic activity, tuned by each amyloid's particular fold, that await discovery.
Collapse
|
19
|
Wei P, Han G, Wang Y. Effects of dopamine D2 receptor antagonists on retinal pigment epithelial/choroid complex metabolism in form-deprived myopic guinea pigs. Proteomics 2023; 23:e2200325. [PMID: 37491763 DOI: 10.1002/pmic.202200325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 07/27/2023]
Abstract
The retinal pigment epithelial (RPE)/choroid complex regulates myopia development, but the precise pathogenesis of myopia remains unclear. We aimed to investigate the changes in RPE/choroid complex metabolism in a form deprivation myopia model after dopamine D2 receptor (D2R) modulation. Guinea pigs were randomly divided into normal (NC), form deprivation myopia (FDM), and FDM treated with dopamine D2R antagonist groups. Differential metabolites were screened using SIMCA-P software and MetaboAnalyst metabolomics analysis tool. Functions of differential metabolites were analyzed using KEGG enrichment pathways. Relative to the NC group, 38 differential metabolites were identified, comprising 29 increased metabolites (including nicotinic acid, cytosine, and glutamate) and 9 decreased metabolites, of which proline exhibited the largest decrease. Pathway analysis revealed regulation of arginine/proline and aspartate/glutamate metabolism. Intravitreal D2R antagonist injection increased proline concentrations and activated arginine/proline and purine metabolism pathways. In sum, D2R antagonists alleviated the myopia trend of refractive biological parameters in form deprivation myopic guinea pigs, suggesting the involvement of dopamine D2R signaling in myopia pathogenesis. The RPE/choroid may provide glutamate to the retina by activating proline metabolism via metabolic coupling with the retina. Dopamine D2R antagonism may modulate proline/arginine metabolic pathways in the RPE/choroid and regulate metabolism, information presentation, and myopia.
Collapse
Affiliation(s)
- Pinghui Wei
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin, PR China
- Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, PR China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, PR China
| | - Guoge Han
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin, PR China
- Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, PR China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, PR China
| | - Yan Wang
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin, PR China
- Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, PR China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, PR China
| |
Collapse
|
20
|
Horvath I, Mohamed KA, Kumar R, Wittung-Stafshede P. Amyloids of α-Synuclein Promote Chemical Transformations of Neuronal Cell Metabolites. Int J Mol Sci 2023; 24:12849. [PMID: 37629028 PMCID: PMC10454467 DOI: 10.3390/ijms241612849] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/06/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
The assembly of α-synuclein into cross-β structured amyloid fibers results in Lewy body deposits and neuronal degeneration in Parkinson's disease patients. As the cell environment is highly crowded, interactions between the formed amyloid fibers and a range of biomolecules can occur in cells. Although amyloid fibers are considered chemically inert species, recent in vitro work using model substrates has shown α-synuclein amyloids, but not monomers, to catalyze the hydrolysis of ester and phosphoester bonds. To search for putative catalytic activity of α-synuclein amyloids on biologically relevant metabolites, we here incubated α-synuclein amyloids with neuronal SH-SY5Y cell lysates devoid of proteins. LC-MS-based metabolomic (principal component and univariate) analysis unraveled distinct changes in several metabolite levels upon amyloid (but not monomer) incubation. Of 63 metabolites identified, the amounts of four increased (3-hydroxycapric acid, 2-pyrocatechuic acid, adenosine, and NAD), and the amounts of seventeen decreased (including aromatic and apolar amino acids, metabolites in the TCA cycle, keto acids) in the presence of α-synuclein amyloids. Many of these metabolite changes match what has been reported previously in Parkinson's disease patients and animal-model metabolomics studies. Chemical reactivity of α-synuclein amyloids may be a new gain-of-function that alters the metabolite composition in cells and, thereby, modulates disease progression.
Collapse
|
21
|
Liu Q, Zhu D, Li N, Chen S, Hu L, Yu J, Xiong Y. Regulation of LRRK2 mRNA stability by ATIC and its substrate AICAR through ARE-mediated mRNA decay in Parkinson's disease. EMBO J 2023; 42:e113410. [PMID: 37366237 PMCID: PMC10390876 DOI: 10.15252/embj.2022113410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/08/2023] [Accepted: 05/15/2023] [Indexed: 06/28/2023] Open
Abstract
Mutations in LRRK2 are the most common genetic causes of Parkinson's disease (PD). While the enzymatic activity of LRRK2 has been linked to PD, previous work has also provided support for an important role of elevated LRRK2 protein levels, independent of enzymatic activity, in PD pathogenesis. However, the mechanisms underlying the regulation of LRRK2 protein levels remain unclear. Here, we identify a role for the purine biosynthesis pathway enzyme ATIC in the regulation of LRRK2 levels and toxicity. AICAr, the precursor of ATIC substrate, regulates LRRK2 levels in a cell-type-specific manner in vitro and in mouse tissue. AICAr regulates LRRK2 levels through AUF1-mediated mRNA decay. Upon AICAr treatment, the RNA binding protein AUF1 is recruited to the AU-rich elements (ARE) of LRRK2 mRNA leading to the recruitment of the decapping enzyme complex DCP1/2 and decay of LRRK2 mRNA. AICAr suppresses LRRK2 expression and rescues LRRK2-induced dopaminergic neurodegeneration and neuroinflammation in PD Drosophila and mouse models. Together, this study provides insight into a novel regulatory mechanism of LRRK2 protein levels and function via LRRK2 mRNA decay that is distinct from LRRK2 enzymatic functions.
Collapse
Affiliation(s)
- Qinfang Liu
- Department of NeuroscienceUniversity of Connecticut School of MedicineFarmingtonCTUSA
| | - Dong Zhu
- Department of NeuroscienceUniversity of Connecticut School of MedicineFarmingtonCTUSA
| | - Naren Li
- Department of Physiology & NeurobiologyUniversity of ConnecticutStorrsCTUSA
| | - Shifan Chen
- Department of NeuroscienceUniversity of Connecticut School of MedicineFarmingtonCTUSA
| | - Liang Hu
- Department of Physiology & NeurobiologyUniversity of ConnecticutStorrsCTUSA
| | - Jianzhong Yu
- Department of Physiology & NeurobiologyUniversity of ConnecticutStorrsCTUSA
| | - Yulan Xiong
- Department of NeuroscienceUniversity of Connecticut School of MedicineFarmingtonCTUSA
| |
Collapse
|
22
|
Salaramoli S, Joshaghani HR, Hashemy SI. Salivary Biomarkers: Noninvasive Ways for Diagnosis of Parkinson's Disease. Neurol Res Int 2023; 2023:3555418. [PMID: 37434876 PMCID: PMC10332915 DOI: 10.1155/2023/3555418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 06/25/2023] [Accepted: 06/27/2023] [Indexed: 07/13/2023] Open
Abstract
Finding reliable biomarkers has a crucial role in Parkinson's disease (PD) assessments. Saliva is a bodily fluid, which might be used as a source of biomarkers for PD. Our article has reviewed several publications on salivary proteins in PD patients and their potential as biomarkers. We find out that α-Syn's proportion in oligomeric form is higher in PD patients' saliva, which is potent to use as a biomarker for PD. The salivary concentration of DJ-1 and alpha-amylase is lower in PD patients. Also, substance P level is more moderate in PD patients. Although salivary flow rate is decreased in PD patients, high levels of heme oxygenase and acetylcholinesterase might be used as noninvasive biomarkers. Salivary miRNAs (miR-153, miR-223, miR-874, and miR-145-3p) are novel diagnostic biomarkers that should be given more attention.
Collapse
Affiliation(s)
- Sanaz Salaramoli
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Joshaghani
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
23
|
Cocco C, Manai AL, Manca E, Noli B. Brain-Biomarker Changes in Body Fluids of Patients with Parkinson's Disease. Int J Mol Sci 2023; 24:10932. [PMID: 37446110 DOI: 10.3390/ijms241310932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Parkinson's disease (PD) is an incurable neurodegenerative disease that is rarely diagnosed at an early stage. Although the understanding of PD-related mechanisms has greatly improved over the last decade, the diagnosis of PD is still based on neurological examination through the identification of motor symptoms, including bradykinesia, rigidity, postural instability, and resting tremor. The early phase of PD is characterized by subtle symptoms with a misdiagnosis rate of approximately 16-20%. The difficulty in recognizing early PD has implications for the potential use of novel therapeutic approaches. For this reason, it is important to discover PD brain biomarkers that can indicate early dopaminergic dysfunction through their changes in body fluids, such as saliva, urine, blood, or cerebrospinal fluid (CSF). For the CFS-based test, the invasiveness of sampling is a major limitation, whereas the other body fluids are easier to obtain and could also allow population screening. Following the identification of the crucial role of alpha-synuclein (α-syn) in the pathology of PD, a very large number of studies have summarized its changes in body fluids. However, methodological problems have led to the poor diagnostic/prognostic value of this protein and alternative biomarkers are currently being investigated. The aim of this paper is therefore to summarize studies on protein biomarkers that are alternatives to α-syn, particularly those that change in nigrostriatal areas and in biofluids, with a focus on blood, and, eventually, saliva and urine.
Collapse
Affiliation(s)
- Cristina Cocco
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Antonio Luigi Manai
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Elias Manca
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Barbara Noli
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| |
Collapse
|
24
|
Tassone A, Meringolo M, Ponterio G, Bonsi P, Schirinzi T, Martella G. Mitochondrial Bioenergy in Neurodegenerative Disease: Huntington and Parkinson. Int J Mol Sci 2023; 24:ijms24087221. [PMID: 37108382 PMCID: PMC10138549 DOI: 10.3390/ijms24087221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Strong evidence suggests a correlation between degeneration and mitochondrial deficiency. Typical cases of degeneration can be observed in physiological phenomena (i.e., ageing) as well as in neurological neurodegenerative diseases and cancer. All these pathologies have the dyshomeostasis of mitochondrial bioenergy as a common denominator. Neurodegenerative diseases show bioenergetic imbalances in their pathogenesis or progression. Huntington's chorea and Parkinson's disease are both neurodegenerative diseases, but while Huntington's disease is genetic and progressive with early manifestation and severe penetrance, Parkinson's disease is a pathology with multifactorial aspects. Indeed, there are different types of Parkinson/Parkinsonism. Many forms are early-onset diseases linked to gene mutations, while others could be idiopathic, appear in young adults, or be post-injury senescence conditions. Although Huntington's is defined as a hyperkinetic disorder, Parkinson's is a hypokinetic disorder. However, they both share a lot of similarities, such as neuronal excitability, the loss of striatal function, psychiatric comorbidity, etc. In this review, we will describe the start and development of both diseases in relation to mitochondrial dysfunction. These dysfunctions act on energy metabolism and reduce the vitality of neurons in many different brain areas.
Collapse
Affiliation(s)
- Annalisa Tassone
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Saint Camillus International University of Health and Medical Sciences, 00131 Rome, Italy
| | - Giulia Ponterio
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| | - Tommaso Schirinzi
- Unit of Neurology, Department of Systems Medicine, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| |
Collapse
|
25
|
Impaired Extracellular Proteostasis in Patients with Heart Failure. Arch Med Res 2023; 54:211-222. [PMID: 36797157 DOI: 10.1016/j.arcmed.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 01/11/2023] [Accepted: 02/02/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Proteostasis impairment and the consequent increase of amyloid burden in the myocardium have been associated with heart failure (HF) development and poor prognosis. A better knowledge of the protein aggregation process in biofluids could assist the development and monitoring of tailored interventions. AIM To compare the proteostasis status and protein's secondary structures in plasma samples of patients with HF with preserved ejection fraction (HFpEF), patients with HF with reduced ejection fraction (HFrEF), and age-matched individuals. METHODS A total of 42 participants were enrolled in 3 groups: 14 patients with HFpEF, 14 patients with HFrEF, and 14 age-matched individuals. Proteostasis-related markers were analyzed by immunoblotting techniques. Fourier Transform Infrared (FTIR) Spectroscopy in Attenuated Total Reflectance (ATR) was applied to assess changes in the protein's conformational profile. RESULTS Patients with HFrEF showed an elevated concentration of oligomeric proteic species and reduced clusterin levels. ATR-FTIR spectroscopy coupled with multivariate analysis allowed the discrimination of HF patients from age-matched individuals in the protein amide I absorption region (1700-1600 cm-1), reflecting changes in protein conformation, with a sensitivity of 73 and a specificity of 81%. Further analysis of FTIR spectra showed significantly reduced random coils levels in both HF phenotypes. Also, compared to the age-matched group, the levels of structures related to fibril formation were significantly increased in patients with HFrEF, whereas the β-turns were significantly increased in patients with HFpEF. CONCLUSION Both HF phenotypes showed a compromised extracellular proteostasis and different protein conformational changes, suggesting a less efficient protein quality control system.
Collapse
|
26
|
LeWitt PA, Li J, Wu KH, Lu M. Diagnostic metabolomic profiling of Parkinson's disease biospecimens. Neurobiol Dis 2023; 177:105962. [PMID: 36563791 DOI: 10.1016/j.nbd.2022.105962] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Reliable and sensitive biomarkers are needed for enhancing and predicting Parkinson's disease (PD) diagnosis. OBJECTIVE To investigate comprehensive metabolomic profiling of biochemicals in CSF and serum for determining diagnostic biomarkers of PD. METHODS Fifty subjects, symptomatic with PD for ≥5 years, were matched to 50 healthy controls (HCs). We used ultrahigh-performance liquid chromatography linked to tandem mass spectrometry (UHPLC-MS/MS) for measuring relative concentrations of ≤1.5 kDalton biochemicals. A reference library created from authentic standards facilitated chemical identifications. Analytes underwent univariate analysis for PD association, with false discovery rate-adjusted p-value (≤0.05) determinations. Multivariate analysis (for identifying a panel of biochemicals discriminating PD from HCs) used several biostatistical methods, including logistic LASSO regression. RESULTS Comparing PD and HCs, strong differentiation was achieved from CSF but not serum specimens. With univariate analysis, 21 CSF compounds exhibited significant differential concentrations. Logistic LASSO regression led to selection of 23 biochemicals (11 shared with those determined by the univariate analysis). The selected compounds, as a group, distinguished PD from HCs, with Area-Under-the-Receiver-Operating-Characteristic (ROC) curve of 0.897. With optimal cutoff, logistic LASSO achieved 100% sensitivity and 96% specificity (and positive and negative predictive values of 96% and 100%). Ten-fold cross-validation gave 84% sensitivity and 82% specificity (and 82% positive and 84% negative predictive values). From the logistic LASSO-chosen regression model, 2 polyamine metabolites (N-acetylcadaverine and N-acetylputrescine) were chosen and had the highest fold-changes in comparing PD to HCs. Another chosen biochemical, acisoga (N-(3-acetamidopropyl)pyrrolidine-2-one), also is a polyamine metabolism derivative. CONCLUSIONS UHPLC-MS/MS assays provided a metabolomic signature highly predictive of PD. These findings provide further evidence for involvement of polyamine pathways in the neurodegeneration of PD.
Collapse
Affiliation(s)
- Peter A LeWitt
- Departments of Neurology, Henry Ford Hospital, West Bloomfield, MI, USA; Wayne State University School of Medicine, West Bloomfield, MI, USA.
| | - Jia Li
- The Department of Public Health Science, Henry Ford Health System, Detroit, MI, USA
| | - Kuan-Han Wu
- The Department of Public Health Science, Henry Ford Health System, Detroit, MI, USA
| | - Mei Lu
- The Department of Public Health Science, Henry Ford Health System, Detroit, MI, USA
| |
Collapse
|
27
|
Urine biomarkers discovery by metabolomics and machine learning for Parkinson's disease diagnoses. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
|
28
|
Miyamoto K, Saiki S, Matsumoto H, Suzuki A, Yamashita Y, Iseki T, Ueno SI, Shiina K, Kataura T, Kamagata K, Imamichi Y, Sasazawa Y, Fujimaki M, Akamatsu W, Hattori N. Systemic Metabolic Alteration Dependent on the Thyroid-Liver Axis in Early PD. Ann Neurol 2023; 93:303-316. [PMID: 36128871 PMCID: PMC10092289 DOI: 10.1002/ana.26510] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 01/31/2023]
Abstract
OBJECTIVE Parkinson's disease (PD) is a common neurodegenerative disease characterized by initial involvement of the olfactory bulb/amygdala or autonomic nerves followed by nigral degeneration. Although autonomic innervation strictly regulates multiorgan systems, including endocrine functions, circulation, and digestion, how dysautonomia in PD affects systemic metabolism has not been identified. In this study, we tried to estimate the pathogenic linkage of PD by nuclear medicine techniques, trans-omic analysis of blood samples, and cultured cell experiments. METHODS Thyroid mediastinum ratio of 123 I-metaiodobenzylguanidine (MIBG) scintigraphy was measured in 1,158 patients with PD. Furthermore, serum exosome miRNA transcriptome analysis and plasma metabolome analysis followed by trans-omic analysis were performed in patients with de novo PD and age-matched healthy control persons. Additionally, thyroid hormone was administered to skeletal muscle and liver derived cells to evaluate the effect of hypothyroidism for these organs. RESULTS Sympathetic denervation of thyroid correlating with its cardiac denervation was confirmed in 1,158 patients with PD by MIBG scintigraphy. Among patients with drug-naïve PD, comprehensive metabolome analysis revealed decreased levels of thyroxine and insufficient fatty acid β-oxidation, which positively correlate with one another. Likewise, both plasma metabolome data and transcriptome data of circulating exosomal miRNAs, revealed specific enrichment of the peroxisome proliferator-activated receptor (PPARα) axis. Finally, association of thyroid hormone with PPARα-dependent β-oxidation regulation was confirmed by in vitro experiments. INTERPRETATION Our findings suggest that interorgan communications between the thyroid and liver are disorganized in the early stage of PD, which would be a sensitive diagnostic biomarker for PD. ANN NEUROL 2023;93:303-316.
Collapse
Affiliation(s)
- Kengo Miyamoto
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shinji Saiki
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hirotaka Matsumoto
- School of Information and Data Sciences, Nagasaki University, Nagasaki, Japan.,Laboratory for Bioinformatics Research, RIKEN Center for Biosystems Dynamics Research, Saitama, Japan
| | - Ayami Suzuki
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuri Yamashita
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Aging Biology in Health and Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tatou Iseki
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shin-Ichi Ueno
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kenta Shiina
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tetsushi Kataura
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Koji Kamagata
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoko Imamichi
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yukiko Sasazawa
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Motoki Fujimaki
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Wado Akamatsu
- Center for Genomic and Regenerative Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
29
|
Qiu J, Peng G, Tang Y, Li S, Liu Z, Zheng J, Wang Y, Liu H, Wei L, Su Y, Lin Y, Dai W, Zhang Z, Chen X, Ding L, Guo W, Zhu X, Xu P, Mo M. Lipid profiles in the cerebrospinal fluid of rats with 6-hydroxydopamine-induced lesions as a model of Parkinson's disease. Front Aging Neurosci 2023; 14:1077738. [PMID: 36742201 PMCID: PMC9895836 DOI: 10.3389/fnagi.2022.1077738] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 12/30/2022] [Indexed: 01/21/2023] Open
Abstract
Background Parkinson's disease (PD) is a progressive neurodegenerative disease with characteristic pathological abnormalities, including the loss of dopaminergic (DA) neurons, a dopamine-depleted striatum, and microglial activation. Lipid accumulation exhibits a close relationship with these pathologies in PD. Methods Here, 6-hydroxydopamine (6-OHDA) was used to construct a rat model of PD, and the lipid profile in cerebrospinal fluid (CSF) obtained from model rats was analyzed using lipidomic approaches. Results Establishment of this PD model was confirmed by apomorphine-induced rotation behaviors, loss of DA neurons, depletion of dopamine in the striatum, and microglial activation after 6-OHDA-induced lesion generation. Unsupervised and supervised methods were employed for lipid analysis. A total of 172 lipid species were identified in CSF and subsequently classified into 18 lipid families. Lipid families, including eicosanoids, triglyceride (TG), cholesterol ester (CE), and free fatty acid (FFA), and 11 lipid species exhibited significantly altered profiles 2 weeks after 6-OHDA administration, and significant changes in eicosanoids, TG, CE, CAR, and three lipid species were noted 5 weeks after 6-OHDA administration. During the period of 6-OHDA-induced lesion formation, the lipid families and species showed concentration fluctuations related to the recovery of behavior and nigrostriatal abnormalities. Correlation analysis showed that the levels of eicosanoids, CE, TG families, and TG (16:0_20:0_18:1) exhibited positive relationships with apomorphine-induced rotation behaviors and negative relationships with tyrosine hydroxylase (TH) expression in the midbrain. Conclusion These results revealed that non-progressive nigrostriatal degeneration induced by 6-OHDA promotes the expression of an impairment-related lipidomic signature in CSF, and the level of eicosanoids, CE, TG families, and TG (16:0_20:0_18:1) in CSF may reveal pathological changes in the midbrain after 6-OHDA insult.
Collapse
Affiliation(s)
- Jiewen Qiu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guoyou Peng
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuting Tang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shiyin Li
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zengfu Liu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiayun Zheng
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yunxin Wang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hanqun Liu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lijian Wei
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yilin Su
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuwan Lin
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Dai
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiling Zhang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiang Chen
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liuyan Ding
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenyuan Guo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoqin Zhu
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingshu Mo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Internal Medicine, Huilai People’s Hospital, Jieyang, China
| |
Collapse
|
30
|
Zhang Y, Liang F, Zhang D, Qi S, Liu Y. Metabolites as extracellular vesicle cargo in health, cancer, pleural effusion, and cardiovascular diseases: An emerging field of study to diagnostic and therapeutic purposes. Biomed Pharmacother 2023; 157:114046. [PMID: 36469967 DOI: 10.1016/j.biopha.2022.114046] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/19/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Extracellular vesicles (EVs) are highly diverse nanoscale membrane-bound structures released from different cell types into the extracellular environment. They play essential functions in cell signaling by transporting their cargo, such as proteins, RNA, DNA, lipids, metabolites, and small molecules, to recipient cells. It has recently been shown that EVs might modulate carcinogenesis by delivering cargo to recipient cells. Furthermore, recent discoveries revealed that changes in plasma-derived EV levels and cargo in subjects with metabolic diseases were documented by many researchers, suggesting that EVs might be a promising source of disease biomarkers. One of the cargos of EVs that has recently attracted the most attention is metabolites. The metabolome of these vesicles introduces a plethora of disease indicators; hence, examining the metabolomics of EVs detected in human biofluids would be an effective approach. On the other hand, metabolites have various roles in biological systems, including the production of energies, synthesizing macromolecules, and serving as signaling molecules and hormones. Metabolome rewiring in cancer and stromal cells is a characteristic of malignancy, but the current understanding of how this affects the metabolite composition and activity of tumor-derived EVs remains in its infancy. Since new findings and studies in the field of exosome biology and metabolism are constantly being published, it is likely that diagnostic and treatment techniques, including the use of exosome metabolites, will be launched in the coming years. Recent years have seen increased interest in the EV metabolome as a possible source for biomarker development. However, our understanding of the role of these molecules in health and disease is still immature. In this work, we have provided the latest findings regarding the role of metabolites as EV cargoes in the pathophysiology of diseases, including cancer, pleural effusion (PE), and cardiovascular disease (CVD). We also discussed the significance of metabolites as EV cargoes of microbiota and their role in host-microbe interaction. In addition, the latest findings on metabolites in the form of EV cargoes as biomarkers for disease diagnosis and treatment are presented in this study.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China
| | - Feng Liang
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China
| | - DuoDuo Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin Province 130021, People's Republic of China
| | - Shuang Qi
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China.
| | - Yan Liu
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China.
| |
Collapse
|
31
|
Luigetti M, Guglielmino V, Romano A, Sciarrone MA, Vitali F, Sabino A, Gervasoni J, Primiano A, Santucci L, Moroni R, Primiano G. A Metabolic Signature of Hereditary Transthyretin Amyloidosis: A Pilot Study. Int J Mol Sci 2022; 23:ijms232416133. [PMID: 36555770 PMCID: PMC9783933 DOI: 10.3390/ijms232416133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Hereditary transthyretin amyloidosis is the most common form of hereditary amyloidosis, with an autosomal dominant inheritance and a variable penetrance. ATTRv amyloidosis can present as a progressive, axonal sensory autonomic and motor neuropathy or as an infiltrative cardiomyopathy. The definition of biomarkers for the early diagnosis of ATTRv is particularly important in the current era of emerging treatments. In this sense, metabolomics could be an instrument able to provide metabolic profiles with their related metabolic pathways, and we would propose them as possible fluid biomarkers. The aim of this study is to identify altered metabolites (free fatty acids and amino acids) in subjects with a confirmed pathogenic TTR variant. Out of the studied total free fatty acids and amino acids, the serum values of palmitic acid are significantly lower in the ATTRv patients compared to the recruited healthy subjects. The metabolic remodeling identified in this neurogenetic disorder could be the manifestation of pathophysiological processes of the disease, such as mitochondrial dysfunction and neuroinflammation, and contribute to explaining some of its clinical manifestations.
Collapse
Affiliation(s)
- Marco Luigetti
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Correspondence: ; Tel.: +39-06-3015-4435
| | | | - Angela Romano
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | | | - Andrea Sabino
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Jacopo Gervasoni
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Aniello Primiano
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Lavinia Santucci
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Rossana Moroni
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Guido Primiano
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
32
|
Liu C, Liu Z, Fang Y, Du Z, Yan Z, Yuan X, Dai L, Yu T, Xiong M, Tian Y, Li H, Li F, Zhang J, Meng L, Wang Z, Jiang H, Zhang Z. Exposure to the environmentally toxic pesticide maneb induces Parkinson's disease-like neurotoxicity in mice: A combined proteomic and metabolomic analysis. CHEMOSPHERE 2022; 308:136344. [PMID: 36087732 DOI: 10.1016/j.chemosphere.2022.136344] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 08/03/2022] [Accepted: 09/02/2022] [Indexed: 06/15/2023]
Abstract
Maneb is a typical dithiocarbamate fungicide that has been extensively used worldwide. Epidemiological evidence shows that exposure to maneb is an environmental risk factor for Parkinson's disease (PD). However, the mechanisms underlying maneb-induced neurotoxicity have yet to be elucidated. In this study, we exposed SH-SY5Y cells to maneb at environmentally relevant concentrations (0, 0.1, 5, 10 mg/L) and found that maneb dose-dependently decreased the cell viability. Furthermore, maneb (60 mg/kg) induced PD-like motor impairment in α-synuclein A53T transgenic mice. The results of tandem mass tag (TMT) proteomics and metabolomics studies of mouse brain and serum revealed significant changes in proteins and metabolites in the pathways involved in the neurotransmitter system. The omics results were verified by targeted metabolomics and Western blot analysis, which demonstrated that maneb induced disturbance of the PD-related pathways, including the phenylalanine and tryptophan metabolism pathways, dopaminergic synapse, synaptic vesicle cycle, mitochondrial dysfunction, and oxidative stress. In addition, the PD-like phenotype induced by maneb was attenuated by the asparagine endopeptidase (AEP) inhibitor compound #11 (CP11) (10 mg/kg), indicating that AEP may play a role in maneb-induced neurotoxicity. To the best of our knowledge, this is the first study to investigate the molecular mechanisms underlying maneb-induced PD-like phenotypes using multiomics analysis, which identified novel therapeutic targets for PD associated with pesticides and other environmental pollutants.
Collapse
Affiliation(s)
- Chaoyang Liu
- Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan, 430073, China; Department of Environmental Engineering, Zhongnan University of Economics and Law, Wuhan, 430073, China; Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zehua Liu
- Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan, 430073, China; Department of Environmental Engineering, Zhongnan University of Economics and Law, Wuhan, 430073, China
| | - Yanyan Fang
- Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan, 430073, China; Department of Environmental Engineering, Zhongnan University of Economics and Law, Wuhan, 430073, China
| | - Zhen Du
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China
| | - Zhi Yan
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China
| | - Xin Yuan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lijun Dai
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting Yu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Min Xiong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ye Tian
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Honghu Li
- Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan, 430073, China; Department of Environmental Engineering, Zhongnan University of Economics and Law, Wuhan, 430073, China
| | - Fei Li
- Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan, 430073, China; Department of Environmental Engineering, Zhongnan University of Economics and Law, Wuhan, 430073, China
| | - Jingdong Zhang
- Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan, 430073, China; Department of Environmental Engineering, Zhongnan University of Economics and Law, Wuhan, 430073, China
| | - Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhihao Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Haiqiang Jiang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
33
|
Gątarek P, Sekulska-Nalewajko J, Bobrowska-Korczaka B, Pawełczyk M, Jastrzębski K, Głąbiński A, Kałużna-Czaplińska J. Plasma Metabolic Disturbances in Parkinson's Disease Patients. Biomedicines 2022; 10:biomedicines10123005. [PMID: 36551761 PMCID: PMC9775245 DOI: 10.3390/biomedicines10123005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Plasma from patients with Parkinson's disease (PD) is a valuable source of information indicating altered metabolites associated with the risk or progression of the disease. Neurotoxicity of dopaminergic neurons, which is triggered by aggregation of α-synuclein, is the main pathogenic feature of PD. However, a growing body of scientific reports indicates that metabolic changes may precede and directly contribute to neurodegeneration. Identification and characterization of the abnormal metabolic pattern in patients' plasma are therefore crucial for the search for potential PD biomarkers. The aims of the present study were (1) to identify metabolic alterations in plasma metabolome in subjects with PD as compared with the controls; (2) to find new potential markers, some correlations among them; (3) to identify metabolic pathways relevant to the pathophysiology of PD. Plasma samples from patients with PD (n = 25) and control group (n = 12) were collected and the gas chromatography-time-of-flight-mass spectrometry GC-TOFMS-based metabolomics approach was used to evaluate the metabolic changes based on the identified 14 metabolites with significantly altered levels using univariate and multivariate statistical analysis. The panel, including 6 metabolites (L-3-methoxytyrosine, aconitic acid, L-methionine, 13-docosenamide, hippuric acid, 9,12-octadecadienoic acid), was identified to discriminate PD from controls with the area under the curve (AUC) of 0.975, with an accuracy of 92%. We also used statistical criteria to identify the significantly altered level of metabolites. The metabolic pathways involved were associated with linoleic acid metabolism, mitochondrial electron transport chain, glycerolipid metabolism, and bile acid biosynthesis. These abnormal metabolic changes in the plasma of patients with PD were mainly related to the amino acid metabolism, TCA cycle metabolism, and mitochondrial function.
Collapse
Affiliation(s)
- Paulina Gątarek
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, 90-924 Lodz, Poland
- CONEM Poland Chemistry and Nutrition Research Group, Lodz University of Technology, 90-924 Lodz, Poland
- Correspondence: (P.G.); (J.K.-C.); Tel.: +48-426-313-091 (J.K.-C.); Fax: +48-426-313-128 (J.K.-C.)
| | | | | | - Małgorzata Pawełczyk
- Department of Neurology and Stroke, Medical University of Lodz, 90-549 Lodz, Poland
| | - Karol Jastrzębski
- Department of Neurology and Stroke, Medical University of Lodz, 90-549 Lodz, Poland
| | - Andrzej Głąbiński
- Department of Neurology and Stroke, Medical University of Lodz, 90-549 Lodz, Poland
| | - Joanna Kałużna-Czaplińska
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, 90-924 Lodz, Poland
- CONEM Poland Chemistry and Nutrition Research Group, Lodz University of Technology, 90-924 Lodz, Poland
- Correspondence: (P.G.); (J.K.-C.); Tel.: +48-426-313-091 (J.K.-C.); Fax: +48-426-313-128 (J.K.-C.)
| |
Collapse
|
34
|
Goncalves VC, Silva da Fonsêca V, de Paula Faria D, Izidoro MA, Berretta AA, de Almeida ACG, Affonso Fonseca FL, Scorza FA, Scorza CA. Propolis induces cardiac metabolism changes in 6-hydroxydopamine animal model: A dietary intervention as a potential cardioprotective approach in Parkinson’s disease. Front Pharmacol 2022; 13:1013703. [PMID: 36313332 PMCID: PMC9606713 DOI: 10.3389/fphar.2022.1013703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/23/2022] [Indexed: 11/24/2022] Open
Abstract
While there is sustained growth of the older population worldwide, ageing is a consistent risk factor for neurodegenerative diseases, such as Parkinson’s-disease (PD). Considered an emblematic movement disorder, PD comprises a miscellany of non-motor symptoms, for which effective management remains an unfulfilled need in clinical practice. Highlighted are the cardiovascular abnormalities, that cause significant burden in PD patients. Evidence suggests that key biological processes underlying PD pathophysiology can be modulated by diet-derived bioactive compounds, such as green propolis, a natural functional food with biological and pharmacological properties. The effects of propolis on cardiac affection associated to PD have received little coverage. In this study, a metabolomics approach and Positron Emission Tomography (PET) imaging were used to assess the metabolic response to diet supplementation with green propolis on heart outcomes of rats with Parkinsonism induced by 6-hydroxydopamine (6-OHDA rats). Untargeted metabolomics approach revealed four cardiac metabolites (2-hydroxybutyric acid, 3-hydroxybutyric acid, monoacylglycerol and alanine) that were significantly modified between animal groups (6-OHDA, 6-OHDA + Propolis and sham). Propolis-induced changes in the level of these cardiac metabolites suggest beneficial effects of diet intervention. From the metabolites affected, functional analysis identified changes in propanoate metabolism (a key carbohydrate metabolism related metabolic pathway), glucose-alanine cycle, protein and fatty acid biosynthesis, energy metabolism, glutathione metabolism and urea cycle. PET imaging detected higher glucose metabolism in the 17 areas of the left ventricle of all rats treated with propolis, substantially contrasting from those rats that did not consume propolis. Our results bring new insights into cardiac metabolic substrates and pathways involved in the mechanisms of the effects of propolis in experimental PD and provide potential novel targets for research in the quest for future therapeutic strategies.
Collapse
Affiliation(s)
- Valeria C. Goncalves
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- *Correspondence: Valeria C. Goncalves, ; Carla Alessandra Scorza,
| | - Victor Silva da Fonsêca
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Daniele de Paula Faria
- Laboratory of Nuclear Medicine (LIM43), Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, Brazil
| | - Mario Augusto Izidoro
- Laboratório de Espectrometria de Massas—Associação Beneficente de Coleta de Sangue (COLSAN), São Paulo, Brazil
| | | | - Antônio-Carlos G. de Almeida
- Laboratório de Neurociências Experimental e Computacional, Departamento de Engenharia de Biossistemas, Universidade Federal de São João Del-Rei (UFSJ), Minas Gerais, Brazil
| | - Fernando Luiz Affonso Fonseca
- Laboratório de Análises Clínicas da Faculdade de Medicina Do ABC, Santo André, São Paulo, Brazil
- Departamento de Ciencias Farmaceuticas da Universidade Federal de Sao Paulo (UNIFESP), Diadema, Brazil
| | - Fulvio Alexandre Scorza
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Carla Alessandra Scorza
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- *Correspondence: Valeria C. Goncalves, ; Carla Alessandra Scorza,
| |
Collapse
|
35
|
Ahnaou A, Whim D. REM sleep behavior and olfactory dysfunction: improving the utility and translation of animal models in the search for neuroprotective therapies for Parkinson's disease. Neurosci Biobehav Rev 2022; 143:104897. [PMID: 36183864 DOI: 10.1016/j.neubiorev.2022.104897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/19/2022] [Accepted: 09/27/2022] [Indexed: 11/25/2022]
Abstract
Parkinson's disease (PD) is a heterogeneous neurodegenerative disease that belongs to the family of synucleiopathies, varying in age, symptoms and progression. Hallmark of the disease is the accumulation of misfolded α-synuclein protein (α-Syn) in neuronal and non-neuronal brain cells. In past decades, diagnosis and treatment of PD has focused on motor deficits, which for the clinical endpoint, have contributed to the prevalence of deficits in the nigrostriatal dopaminergic system and animal models related to motor behavior to study disease. However, clinical trials have failed to translate results from animal models into effective treatments. PD as a multisystem disorder therefore requires additional assessment of motor and non-motor symptoms. Braak's staging revealed early α-Syn pathology in pontine brainstem and olfactory circuits controlling rapid eye movement sleep behavior disorder (RBD) and olfaction, respectively. Recent converging evidence from multicenter clinical studies supports that RBD is the most important risk factor for prodromal PD and the conduct of neuroprotective therapeutic trials in RBD-enriched cohorts has been recommended. Animal models of RBD and olfaction dysfunction can aid to fill the gap in translational research.
Collapse
Affiliation(s)
- A Ahnaou
- Department of Neuroscience, Janssen Research & Development, a Division of Janssen Pharmaceutica NV. Turnhoutseweg 30, B-2340 Beerse, Belgium.
| | - Drinkenburg Whim
- Department of Neuroscience, Janssen Research & Development, a Division of Janssen Pharmaceutica NV. Turnhoutseweg 30, B-2340 Beerse, Belgium
| |
Collapse
|
36
|
GPBAR1 preserves neurite and synapse of dopaminergic neurons via RAD21-OPCML signaling: Role in preventing Parkinson's disease in mouse model and human patients. Pharmacol Res 2022; 184:106459. [PMID: 36152741 DOI: 10.1016/j.phrs.2022.106459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/20/2022]
Abstract
Parkinson's disease (PD) exhibits systemic impacts on the metabolism, while metabolic alteration contributes to the risk and progression of PD. Bile acids (BA) metabolism disturbance has been linked to PD pathology. Membrane-bound G protein-coupled bile acid receptor 1 (GPBAR1) is expressed in the brain and thought to be neuroprotective; however, the role of GPBAR1 in PD remains unknown. The current study aimed to explore the effect of GPBAR1 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice with dopaminergic (DA) neuron-specific Gpbar1 knockdown or central GPBAR1 activation. The underlying mechanisms were investigated using mesencephalic primary neurons analyzed. Our study found that GPBAR1 was reduced in the substantia nigra of PD patients and MPTP-PD mice, and its expression was negatively correlated with the severity of PD-related features. Genetic downregulation of Gpbar1 in mouse mesencephalic DA neurons exacerbated MPTP-induced neurobehavioral and neuropathological deficits, whereas activation of central GPBAR1 with INT-777 (INT) relieved it. Moreover, in vivo and in vitro experiments showed the neurite- and synapse-protective effects of GPBAR1 activation in PD model. Mechanistically, by promoting the nuclear localization of cohesin subunit RAD21, GPBAR1 activation increased opioid-binding cell adhesion molecule (Opcml) expression, thereby inhibiting neurite and synapse degeneration of DA neurons in PD model. Collectively, our findings demonstrate that GPBAR1 is implicated in PD pathogenesis and activation of central GPBAR1 with INT antagonizes neurodegenerative pathology in PD model. This neuroprotection, at least in part, is attributed to the RAD21-OPCML signaling in neurons. Hence, GPBAR1 may serve as a promising candidate target for PD treatment.
Collapse
|
37
|
Feng H, Xi F. Miltirone Attenuates Reactive Oxygen Species-Dependent Neuronal Apoptosis in MPP +-Induced Cell Model of Parkinson's Disease Through Regulating the PI3K/Akt Pathway. Neurochem Res 2022; 47:3137-3149. [PMID: 35810264 DOI: 10.1007/s11064-022-03669-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022]
Abstract
Miltirone is a phenanthrene-quinone derived from Salvia miltiorrhiza Bunge with anti-inflammatory and anti-oxidant effects. Our study aimed to explore the protective effect of miltirone on 1-methyl-4-phenylpyridinium (MPP+)-induced cell model of Parkinson's disease (PD). PharmMapper database was employed to predict the targets of miltirone. PD-related genes were identified using GeneCards database. The overlapping genes between miltirone and PD were screened out using Venn diagram. KEGG analysis was performed using DAVID and KOBAS databases. Cell viability, reactive oxygen species (ROS) generation, apoptosis, and caspase-3 activity were detected by CCK-8 assay, a ROS assay kit, TUNEL, and caspase-3 activity assay, respectively. Effect of miltirone on the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway was explored by western blot analysis. A total of 214 targets of miltirone and 372 targets related to PD were attained, including 29 overlapping targets. KEGG analysis demonstrated that the 29 overlapping targets were both significantly enriched in the PI3K/Akt pathway. MPP+ stimulation reduced the cell viability in SH-SY5Y cells and neuronal primary cultures derived from human brain. Miltirone or N-acetylcysteine (NAC) attenuated MPP+-induced reduction in cell viability, ROS production, SOD activity reduction, apoptosis, and increase of caspase-3 activity. Additionally, miltirone recuperated MPP+-induced inactivation of the PI3K/Akt pathway. Moreover, treatment with LY294002, an inhibitor of the PI3K/Akt pathway, reversed the inhibitory effect of miltirone on MPP+-induced ROS generation and apoptosis in SH-SY5Y cells and neuronal primary cultures. In conclusion, miltirone attenuated ROS-dependent apoptosis in MPP+-induced cellular model of PD through activating the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Huiqiong Feng
- Department of Neurology, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, 41 Linyin Road, Baotou, 014010, Inner Mongolia, China
| | - Fuqiang Xi
- Department of Neurology, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, 41 Linyin Road, Baotou, 014010, Inner Mongolia, China.
| |
Collapse
|
38
|
Cukier HN, Kim H, Griswold AJ, Codreanu SG, Prince LM, Sherrod SD, McLean JA, Dykxhoorn DM, Ess KC, Hedera P, Bowman AB, Neely MD. Genomic, transcriptomic, and metabolomic profiles of hiPSC-derived dopamine neurons from clinically discordant brothers with identical PRKN deletions. NPJ Parkinsons Dis 2022; 8:84. [PMID: 35768426 PMCID: PMC9243035 DOI: 10.1038/s41531-022-00346-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 05/27/2022] [Indexed: 11/25/2022] Open
Abstract
We previously reported on two brothers who carry identical compound heterozygous PRKN mutations yet present with significantly different Parkinson's Disease (PD) clinical phenotypes. Juvenile cases demonstrate that PD is not necessarily an aging-associated disease. Indeed, evidence for a developmental component to PD pathogenesis is accumulating. Thus, we hypothesized that the presence of additional genetic modifiers, including genetic loci relevant to mesencephalic dopamine neuron development, could potentially contribute to the different clinical manifestations of the two brothers. We differentiated human-induced pluripotent stem cells (hiPSCs) derived from the two brothers into mesencephalic neural precursor cells and early postmitotic dopaminergic neurons and performed wholeexome sequencing and transcriptomic and metabolomic analyses. No significant differences in the expression of canonical dopamine neuron differentiation markers were observed. Yet our transcriptomic analysis revealed a significant downregulation of the expression of three neurodevelopmentally relevant cell adhesion molecules, CNTN6, CNTN4 and CHL1, in the cultures of the more severely affected brother. In addition, several HLA genes, known to play a role in neurodevelopment, were differentially regulated. The expression of EN2, a transcription factor crucial for mesencephalic dopamine neuron development, was also differentially regulated. We further identified differences in cellular processes relevant to dopamine metabolism. Lastly, wholeexome sequencing, transcriptomics and metabolomics data all revealed differences in glutathione (GSH) homeostasis, the dysregulation of which has been previously associated with PD. In summary, we identified genetic differences which could potentially, at least partially, contribute to the discordant clinical PD presentation of the two brothers.
Collapse
Affiliation(s)
- Holly N Cukier
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hyunjin Kim
- School of Health Sciences, Purdue University, West Lafayette, Indiana, IN, USA
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Simona G Codreanu
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Lisa M Prince
- School of Health Sciences, Purdue University, West Lafayette, Indiana, IN, USA
| | - Stacy D Sherrod
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - John A McLean
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Derek M Dykxhoorn
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kevin C Ess
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Peter Hedera
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Neurology, University of Louisville, Louisville, KY, USA
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, Indiana, IN, USA.
| | - M Diana Neely
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
39
|
Horgusluoglu E, Neff R, Song W, Wang M, Wang Q, Arnold M, Krumsiek J, Galindo‐Prieto B, Ming C, Nho K, Kastenmüller G, Han X, Baillie R, Zeng Q, Andrews S, Cheng H, Hao K, Goate A, Bennett DA, Saykin AJ, Kaddurah‐Daouk R, Zhang B. Integrative metabolomics-genomics approach reveals key metabolic pathways and regulators of Alzheimer's disease. Alzheimers Dement 2022; 18:1260-1278. [PMID: 34757660 PMCID: PMC9085975 DOI: 10.1002/alz.12468] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 12/29/2022]
Abstract
Metabolites, the biochemical products of the cellular process, can be used to measure alterations in biochemical pathways related to the pathogenesis of Alzheimer's disease (AD). However, the relationships between systemic abnormalities in metabolism and the pathogenesis of AD are poorly understood. In this study, we aim to identify AD-specific metabolomic changes and their potential upstream genetic and transcriptional regulators through an integrative systems biology framework for analyzing genetic, transcriptomic, metabolomic, and proteomic data in AD. Metabolite co-expression network analysis of the blood metabolomic data in the Alzheimer's Disease Neuroimaging Initiative (ADNI) shows short-chain acylcarnitines/amino acids and medium/long-chain acylcarnitines are most associated with AD clinical outcomes, including episodic memory scores and disease severity. Integration of the gene expression data in both the blood from the ADNI and the brain from the Accelerating Medicines Partnership Alzheimer's Disease (AMP-AD) program reveals ABCA1 and CPT1A are involved in the regulation of acylcarnitines and amino acids in AD. Gene co-expression network analysis of the AMP-AD brain RNA-seq data suggests the CPT1A- and ABCA1-centered subnetworks are associated with neuronal system and immune response, respectively. Increased ABCA1 gene expression and adiponectin protein, a regulator of ABCA1, correspond to decreased short-chain acylcarnitines and amines in AD in the ADNI. In summary, our integrated analysis of large-scale multiomics data in AD systematically identifies novel metabolites and their potential regulators in AD and the findings pave a way for not only developing sensitive and specific diagnostic biomarkers for AD but also identifying novel molecular mechanisms of AD pathogenesis.
Collapse
Affiliation(s)
- Emrin Horgusluoglu
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Ryan Neff
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Won‐Min Song
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Minghui Wang
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Qian Wang
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Matthias Arnold
- Institute of Computational BiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- Department of Psychiatry and Behavioral SciencesDuke UniversityDurhamNorth CarolinaUSA
| | - Jan Krumsiek
- Department of Physiology and BiophysicsWeill Cornell MedicineInstitute for Computational BiomedicineEnglander Institute for Precision MedicineNew YorkNew YorkUSA
| | - Beatriz Galindo‐Prieto
- Department of Physiology and BiophysicsWeill Cornell MedicineInstitute for Computational BiomedicineEnglander Institute for Precision MedicineNew YorkNew YorkUSA
- Helen and Robert Appel Alzheimer's Disease Research InstituteBrain and Mind Research InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Chen Ming
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences; Indiana Alzheimer Disease CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Gabi Kastenmüller
- Institute of Computational BiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
| | - Xianlin Han
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | | | - Qi Zeng
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Shea Andrews
- Department of NeuroscienceRonald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Haoxiang Cheng
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Ke Hao
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Alison Goate
- Department of NeuroscienceRonald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - David A. Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences; Indiana Alzheimer Disease CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Rima Kaddurah‐Daouk
- Department of Psychiatry and Behavioral SciencesDuke UniversityDurhamNorth CarolinaUSA
- Duke Institute of Brain SciencesDuke UniversityDurhamNorth CarolinaUSA
- Department of MedicineDuke UniversityDurhamNorth CarolinaUSA
| | - Bin Zhang
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | | | | |
Collapse
|
40
|
Konjevod M, Sáiz J, Barbas C, Bergareche A, Ardanaz E, Huerta JM, Vinagre-Aragón A, Erro ME, Chirlaque MD, Abilleira E, Ibarluzea JM, Amiano P. A Set of Reliable Samples for the Study of Biomarkers for the Early Diagnosis of Parkinson's Disease. Front Neurol 2022; 13:844841. [PMID: 35707037 PMCID: PMC9189395 DOI: 10.3389/fneur.2022.844841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/19/2022] [Indexed: 11/30/2022] Open
Abstract
Background Parkinson's disease (PD) is a progressive neurodegenerative disorder, diagnosed according to the clinical criteria that occur in already advanced stages of PD. The definition of biomarkers for the early diagnosis of PD represents a challenge that might improve treatment and avoid complications in this disease. Therefore, we propose a set of reliable samples for the identification of altered metabolites to find potential prognostic biomarkers for early PD. Methods This case–control study included plasma samples of 12 patients with PD and 21 control subjects, from the Spanish European Prospective Investigation into Cancer and Nutrition (EPIC)-Navarra cohort, part of the EPIC-Spain study. All the case samples were provided by healthy volunteers who were followed-up for 15.9 (±4.1) years and developed PD disease later on, after the sample collection. Liquid chromatography coupled to tandem mass spectrometry was used for the analysis of samples. Results Out of 40 that were selected and studied due to their involvement in established cases of PD, seven significantly different metabolites between PD cases and healthy control subjects were obtained in this study (benzoic acid, palmitic acid, oleic acid, stearic acid, myo-inositol, sorbitol, and quinolinic acid). These metabolites are related to mitochondrial dysfunction, the oxidative stress, and the mechanisms of energy production. Conclusion We propose the samples from the EPIC study as reliable and invaluable samples for the search of early biomarkers of PD. Likewise, this study might also be a starting point in the establishment of a well-founded panel of metabolites that can be used for the early detection of this disease.
Collapse
Affiliation(s)
- Marcela Konjevod
- Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
- Facultad de Farmacia, Centro de Metabolómica y Bioanálisis, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Jorge Sáiz
- Facultad de Farmacia, Centro de Metabolómica y Bioanálisis, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- *Correspondence: Jorge Sáiz
| | - Coral Barbas
- Facultad de Farmacia, Centro de Metabolómica y Bioanálisis, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Alberto Bergareche
- Department of Neurology, University Hospital Donostia, San Sebastián, Spain
- Neuroscience Area, Biodonostia Health Research Institute, San Sebastián, Spain
- Biomedical Research Networking Centre Consortium for the Area of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Alberto Bergareche
| | - Eva Ardanaz
- Navarra Public Health Institute, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
| | - José Ma Huerta
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| | - Ana Vinagre-Aragón
- Department of Neurology, University Hospital Donostia, San Sebastián, Spain
| | - Ma Elena Erro
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Department of Neurology, Navarra Hospital Complex, Pamplona, Spain
| | - Ma Dolores Chirlaque
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| | - Eunate Abilleira
- Ministry of Health of the Basque Government, Public Health Laboratory in Gipuzkoa, San Sebastián, Spain
- Epidemiology of Chronic and Comunnicable Diseases Area, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Jesús Ma Ibarluzea
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Ministry of Health of the Basque Government, Sub Directorate for Public Health and Addictions of Gipuzkoa, San Sebastián, Spain
- Environmental Epidemiology and Child Development Area, Biodonostia Health Research Institute, San Sebastián, Spain
- Faculty of Psychology, University of the Basque Country UPV/EHU, San Sebastian, Spain
| | - Pilar Amiano
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Ministry of Health of the Basque Government, Public Health Laboratory in Gipuzkoa, San Sebastián, Spain
- Epidemiology of Chronic and Comunnicable Diseases Area, Biodonostia Health Research Institute, San Sebastián, Spain
| |
Collapse
|
41
|
Gramotnev DK, Gramotnev G, Gramotnev A, Summers MJ. Path analysis of biomarkers for cognitive decline in early Parkinson’s disease. PLoS One 2022; 17:e0268379. [PMID: 35560326 PMCID: PMC9106174 DOI: 10.1371/journal.pone.0268379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 04/26/2022] [Indexed: 11/19/2022] Open
Abstract
Clinical and biochemical diversity of Parkinson’s disease (PD) and numerous demographic, clinical, and pathological measures influencing cognitive function and its decline in PD create problems with the determination of effects of individual measures on cognition in PD. This is particularly the case where these measures significantly interrelate with each other producing intricate networks of direct and indirect effects on cognition. Here, we use generalized structural equation modelling (GSEM) to identify and characterize significant paths for direct and indirect effects of 14 baseline measures on global cognition in PD at baseline and at 4 years later. We consider 269 drug-naïve participants from the Parkinson’s Progression Marker Initiative database, diagnosed with idiopathic PD and observed for at least 4 years after baseline. Two GSEM networks are derived, highlighting the possibility of at least two different molecular pathways or two different PD sub-types, with either CSF p-tau181 or amyloid beta (1–42) being the primary protein variables potentially driving progression of cognitive decline. The models provide insights into the interrelations between the 14 baseline variables, and determined their total effects on cognition in early PD. High CSF amyloid concentrations (> 500 pg/ml) are associated with nearly full protection against cognitive decline in early PD in the whole range of baseline age between 40 and 80 years, and irrespectively of whether p-tau181 or amyloid beta (1–42) are considered as the primary protein variables. The total effect of depression on cognition is shown to be strongly amplified by PD, but not at the time of diagnosis or at prodromal stages. CSF p-tau181 protein could not be a reliable indicator of cognitive decline because of its significantly heterogeneous effects on cognition. The outcomes will enable better understanding of the roles of the clinical and pathological measures and their mutual effects on cognition in early PD.
Collapse
Affiliation(s)
| | - Galina Gramotnev
- Research and Data Analysis Centre, Brisbane, Queensland, Australia
| | - Alexandra Gramotnev
- Research and Data Analysis Centre, Brisbane, Queensland, Australia
- Sunshine Coast Mind & Neuroscience – Thompson Institute, University of the Sunshine Coast, Birtinya, Queensland, Australia
| | - Mathew J. Summers
- School of Health and Behavioural Science, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| |
Collapse
|
42
|
Zhang Y, He X, Qian Y, Xu S, Mo C, Yan Z, Yang X, Xiao Q. Plasma branched-chain and aromatic amino acids correlate with the gut microbiota and severity of Parkinson's disease. NPJ Parkinsons Dis 2022; 8:48. [PMID: 35449203 PMCID: PMC9023571 DOI: 10.1038/s41531-022-00312-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
Disturbances of circulating amino acids have been demonstrated in patients with Parkinson’s disease (PD). However, there have been no consistent results for branched-chain amino acids (BCAAs) and aromatic amino acids (AAAs), and related factors have not been explored. We aimed to explore plasma BCAA and AAA profiles in PD patients, and identify their correlations with clinical characteristics and the gut microbiota. Plasma BCAA (leucine, isoleucine, and valine) and AAA (tyrosine and phenylalanine) levels were measured in 106 PD patients and 114 controls. Fecal samples were collected from PD patients for microbiota sequencing and functional analysis. We found that plasma BCAAs and tyrosine were decreased in PD patients. BCAAs and AAAs were correlated with clinical characteristics and microbial taxa, and, in particular, they were negatively correlated with the Hoehn and Yahr stage. Compared with early PD patients, BCAA and AAA levels were even lower, and microbial composition was altered in advanced PD patients. Predictive functional analysis indicated that predicted genes numbers involved in BCAA biosynthesis were lower in advanced PD patients. What’s more, the fecal abundances of critical genes (ilvB, ilvC, ilvD, and ilvN) involved in BCAA biosynthesis were reduced and fecal BCAA concentrations were lower in advanced PD patients. In conclusion, the disturbances of plasma BCAAs and AAAs in PD patients may be related to the gut microbiota and exacerbated with PD severity. The microbial amino acid metabolism may serve as a potential mechanistic link.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoqin He
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwei Qian
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaoqing Xu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengjun Mo
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Yan
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Xiaodong Yang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qin Xiao
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
43
|
Kwon EH, Tennagels S, Gold R, Gerwert K, Beyer L, Tönges L. Update on CSF Biomarkers in Parkinson's Disease. Biomolecules 2022; 12:biom12020329. [PMID: 35204829 PMCID: PMC8869235 DOI: 10.3390/biom12020329] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/02/2022] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
Progress in developing disease-modifying therapies in Parkinson’s disease (PD) can only be achieved through reliable objective markers that help to identify subjects at risk. This includes an early and accurate diagnosis as well as continuous monitoring of disease progression and therapy response. Although PD diagnosis still relies mainly on clinical features, encouragingly, advances in biomarker discovery have been made. The cerebrospinal fluid (CSF) is a biofluid of particular interest to study biomarkers since it is closest to the brain structures and therefore could serve as an ideal source to reflect ongoing pathologic processes. According to the key pathophysiological mechanisms, the CSF status of α-synuclein species, markers of amyloid and tau pathology, neurofilament light chain, lysosomal enzymes and markers of neuroinflammation provide promising preliminary results as candidate biomarkers. Untargeted approaches in the field of metabolomics provide insights into novel and interconnected biological pathways. Markers based on genetic forms of PD can contribute to identifying subgroups suitable for gene-targeted treatment strategies that might also be transferable to sporadic PD. Further validation analyses in large PD cohort studies will identify the CSF biomarker or biomarker combinations with the best value for clinical and research purposes.
Collapse
Affiliation(s)
- Eun Hae Kwon
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany; (E.H.K.); (S.T.); (R.G.)
| | - Sabrina Tennagels
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany; (E.H.K.); (S.T.); (R.G.)
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany; (E.H.K.); (S.T.); (R.G.)
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, D-44801 Bochum, Germany; (K.G.); (L.B.)
| | - Klaus Gerwert
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, D-44801 Bochum, Germany; (K.G.); (L.B.)
- Faculty of Biology and Biotechnology, Department of Biophysics, Ruhr University Bochum, D-44801 Bochum, Germany
| | - Léon Beyer
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, D-44801 Bochum, Germany; (K.G.); (L.B.)
- Faculty of Biology and Biotechnology, Department of Biophysics, Ruhr University Bochum, D-44801 Bochum, Germany
| | - Lars Tönges
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany; (E.H.K.); (S.T.); (R.G.)
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, D-44801 Bochum, Germany; (K.G.); (L.B.)
- Correspondence: ; Tel.: +49-234-509-2420; Fax: +49-234-509-2439
| |
Collapse
|
44
|
Plasma Metabolite Signature Classifies Male LRRK2 Parkinson’s Disease Patients. Metabolites 2022; 12:metabo12020149. [PMID: 35208223 PMCID: PMC8876175 DOI: 10.3390/metabo12020149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 02/04/2023] Open
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disease, causing loss of motor and nonmotor function. Diagnosis is based on clinical symptoms that do not develop until late in the disease progression, at which point the majority of the patients’ dopaminergic neurons are already destroyed. While many PD cases are idiopathic, hereditable genetic risks have been identified, including mutations in the gene for LRRK2, a multidomain kinase with roles in autophagy, mitochondrial function, transcription, molecular structural integrity, the endo-lysosomal system, and the immune response. A definitive PD diagnosis can only be made post-mortem, and no noninvasive or blood-based disease biomarkers are currently available. Alterations in metabolites have been identified in PD patients, suggesting that metabolomics may hold promise for PD diagnostic tools. In this study, we sought to identify metabolic markers of PD in plasma. Using a 1H-13C heteronuclear single quantum coherence spectroscopy (HSQC) NMR spectroscopy metabolomics platform coupled with machine learning (ML), we measured plasma metabolites from approximately age/sex-matched PD patients with G2019S LRRK2 mutations and non-PD controls. Based on the differential level of known and unknown metabolites, we were able to build a ML model and develop a Biomarker of Response (BoR) score, which classified male LRRK2 PD patients with 79.7% accuracy, 81.3% sensitivity, and 78.6% specificity. The high accuracy of the BoR score suggests that the metabolomics/ML workflow described here could be further utilized in the development of a confirmatory diagnostic for PD in larger patient cohorts. A diagnostic assay for PD will aid clinicians and their patients to quickly move toward a definitive diagnosis, and ultimately empower future clinical trials and treatment options.
Collapse
|
45
|
Solana-Manrique C, Sanz FJ, Torregrosa I, Palomino-Schätzlein M, Hernández-Oliver C, Pineda-Lucena A, Paricio N. Metabolic Alterations in a Drosophila Model of Parkinson's Disease Based on DJ-1 Deficiency. Cells 2022; 11:cells11030331. [PMID: 35159141 PMCID: PMC8834223 DOI: 10.3390/cells11030331] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/13/2022] Open
Abstract
Parkinson’s disease (PD) is the second-most common neurodegenerative disorder, whose physiopathology is still unclear. Moreover, there is an urgent need to discover new biomarkers and therapeutic targets to facilitate its diagnosis and treatment. Previous studies performed in PD models and samples from PD patients already demonstrated that metabolic alterations are associated with this disease. In this context, the aim of this study is to provide a better understanding of metabolic disturbances underlying PD pathogenesis. To achieve this goal, we used a Drosophila PD model based on inactivation of the DJ-1β gene (ortholog of human DJ-1). Metabolomic analyses were performed in 1-day-old and 15-day-old DJ-1β mutants and control flies using 1H nuclear magnetic resonance spectroscopy, combined with expression and enzymatic activity assays of proteins implicated in altered pathways. Our results showed that the PD model flies exhibited protein metabolism alterations, a shift fromthe tricarboxylic acid cycle to glycolytic pathway to obtain ATP, together with an increase in the expression of some urea cycle enzymes. Thus, these metabolic changes could contribute to PD pathogenesis and might constitute possible therapeutic targets and/or biomarkers for this disease.
Collapse
Affiliation(s)
- Cristina Solana-Manrique
- Departamento de Genética, Facultad CC Biológicas, Instituto Universitario de Biotecnología y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain; (C.S.-M.); (F.J.S.); (I.T.)
| | - Francisco José Sanz
- Departamento de Genética, Facultad CC Biológicas, Instituto Universitario de Biotecnología y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain; (C.S.-M.); (F.J.S.); (I.T.)
| | - Isabel Torregrosa
- Departamento de Genética, Facultad CC Biológicas, Instituto Universitario de Biotecnología y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain; (C.S.-M.); (F.J.S.); (I.T.)
| | | | - Carolina Hernández-Oliver
- Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain; (C.H.-O.); (A.P.-L.)
| | - Antonio Pineda-Lucena
- Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain; (C.H.-O.); (A.P.-L.)
- Programa de Terapias Moleculares, Centro de Investigación Médica Aplicada, Universidad de Navarra, 31008 Pamplona, Spain
| | - Nuria Paricio
- Departamento de Genética, Facultad CC Biológicas, Instituto Universitario de Biotecnología y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain; (C.S.-M.); (F.J.S.); (I.T.)
- Correspondence: ; Tel.: +34-96-354-3005; Fax: +34-96-354-3029
| |
Collapse
|
46
|
Ostapiuk A, Urbanska EM. Kynurenic acid in neurodegenerative disorders-unique neuroprotection or double-edged sword? CNS Neurosci Ther 2022; 28:19-35. [PMID: 34862742 PMCID: PMC8673711 DOI: 10.1111/cns.13768] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 12/24/2022] Open
Abstract
AIMS The family of kynurenine pathway (KP) metabolites includes compounds produced along two arms of the path and acting in clearly opposite ways. The equilibrium between neurotoxic kynurenines, such as 3-hydroxykynurenine (3-HK) or quinolinic acid (QUIN), and neuroprotective kynurenic acid (KYNA) profoundly impacts the function and survival of neurons. This comprehensive review summarizes accumulated evidence on the role of KYNA in Alzheimer's, Parkinson's and Huntington's diseases, and discusses future directions of potential pharmacological manipulations aimed to modulate brain KYNA. DISCUSSION The synthesis of specific KP metabolites is tightly regulated and may considerably vary under physiological and pathological conditions. Experimental data consistently imply that shift of the KP to neurotoxic branch producing 3-HK and QUIN formation, with a relative or absolute deficiency of KYNA, is an important factor contributing to neurodegeneration. Targeting specific brain regions to maintain adequate KYNA levels seems vital; however, it requires the development of precise pharmacological tools, allowing to avoid the potential cognitive adverse effects. CONCLUSIONS Boosting KYNA levels, through interference with the KP enzymes or through application of prodrugs/analogs with high bioavailability and potency, is a promising clinical approach. The use of KYNA, alone or in combination with other compounds precisely influencing specific populations of neurons, is awaiting to become a significant therapy for neurodegenerative disorders.
Collapse
Affiliation(s)
- Aleksandra Ostapiuk
- Laboratory of Cellular and Molecular PharmacologyDepartment of Experimental and Clinical PharmacologyMedical University of LublinLublinPoland
- Present address:
Department of Clinical Digestive OncologyKU LeuvenLeuvenBelgium
| | - Ewa M. Urbanska
- Laboratory of Cellular and Molecular PharmacologyDepartment of Experimental and Clinical PharmacologyMedical University of LublinLublinPoland
| |
Collapse
|
47
|
Huang Y, Zhao M, Chen X, Zhang R, Le A, Hong M, Zhang Y, Jia L, Zang W, Jiang C, Wang J, Fan X, Wang J. Tryptophan Metabolism in Central Nervous System Diseases: Pathophysiology and Potential Therapeutic Strategies. Aging Dis 2022; 14:858-878. [PMID: 37191427 DOI: 10.14336/ad.2022.0916] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 09/16/2022] [Indexed: 11/19/2022] Open
Abstract
The metabolism of L-tryptophan (TRP) regulates homeostasis, immunity, and neuronal function. Altered TRP metabolism has been implicated in the pathophysiology of various diseases of the central nervous system. TRP is metabolized through two main pathways, the kynurenine pathway and the methoxyindole pathway. First, TRP is metabolized to kynurenine, then kynurenic acid, quinolinic acid, anthranilic acid, 3-hydroxykynurenine, and finally 3-hydroxyanthranilic acid along the kynurenine pathway. Second, TRP is metabolized to serotonin and melatonin along the methoxyindole pathway. In this review, we summarize the biological properties of key metabolites and their pathogenic functions in 12 disorders of the central nervous system: schizophrenia, bipolar disorder, major depressive disorder, spinal cord injury, traumatic brain injury, ischemic stroke, intracerebral hemorrhage, multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease. Furthermore, we summarize preclinical and clinical studies, mainly since 2015, that investigated the metabolic pathway of TRP, focusing on changes in biomarkers of these neurologic disorders, their pathogenic implications, and potential therapeutic strategies targeting this metabolic pathway. This critical, comprehensive, and up-to-date review helps identify promising directions for future preclinical, clinical, and translational research on neuropsychiatric disorders.
Collapse
|
48
|
Ostrakhovitch EA, Song ES, Macedo JKA, Gentry MS, Quintero JE, van Horne C, Yamasaki TR. Analysis of circulating metabolites to differentiate Parkinson's disease and essential tremor. Neurosci Lett 2021; 769:136428. [PMID: 34971771 DOI: 10.1016/j.neulet.2021.136428] [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: 08/30/2021] [Revised: 12/24/2021] [Accepted: 12/24/2021] [Indexed: 12/26/2022]
Abstract
Parkinson disease (PD) and essential tremor (ET) are two common adult-onset tremor disorders in which prevalence increases with age. PD is a neurodegenerative condition with progressive disability. In ET, neurodegeneration is not an established etiology. We sought to determine whether an underlying metabolic pattern may differentiate ET from PD. Circulating metabolites in plasma and cerebrospinal fluid were analyzed using gas chromatography-mass spectroscopy. There were several disrupted pathways in PD compared to ET plasma including glycolysis, tyrosine, phenylalanine, tyrosine biosynthesis, purine and glutathione metabolism. Elevated α-synuclein levels in plasma and CSF distinguished PD from ET. The perturbed metabolic state in PD was associated with imbalance in the pentose phosphate pathway, deficits in energy production, and change in NADPH, NADH and nicotinamide phosphoribosyltransferase levels. This work demonstrates significant metabolic differences in plasma and CSF of PD and ET patients.
Collapse
Affiliation(s)
| | - Eun-Suk Song
- Department of Neurology, University of Kentucky, Lexington, KY, 40536, USA
| | - Jessica K A Macedo
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA
| | - Matthew S Gentry
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA
| | - Jorge E Quintero
- Department of Neurosurgery, University of Kentucky, Lexington, KY, 40536, USA
| | - Craig van Horne
- Department of Neurosurgery, University of Kentucky, Lexington, KY, 40536, USA
| | - Tritia R Yamasaki
- Department of Neurology, University of Kentucky, Lexington, KY, 40536, USA; Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA; Veterans Affairs Medical Center, Lexington, KY, 40536, USA
| |
Collapse
|
49
|
Quintero ME, Pontes JGDM, Tasic L. Metabolomics in degenerative brain diseases. Brain Res 2021; 1773:147704. [PMID: 34744014 DOI: 10.1016/j.brainres.2021.147704] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 10/18/2021] [Accepted: 10/23/2021] [Indexed: 12/23/2022]
Abstract
Among the most studied diseases that affect the central nervous system are Parkinson's, Alzheimer's, and Huntington's diseases, but the lack of effective biomarkers, accurate diagnosis, and precise treatment for each of them is currently an issue. Due to the contribution of biomarkers in supporting diagnosis, many recent efforts have focused on their identification and validation at the beginning or during the progression of the mental illness. Metabolome reveals the metabolic processes that result from protein activities under the guided gene expression and environmental factors, either in healthy or pathological conditions. In this context, metabolomics has proven to be a valuable approach. Currently, magnetic resonance spectroscopy (NMR) and mass spectrometry (MS) are the most commonly used bioanalytical techniques for metabolomics. MS-assisted profiling is considered the most versatile technique, and the NMR is the most reproductive. However, each one of them has its drawbacks. In this review, we summarized several alterations in metabolites that have been reported for these three classic brain diseases using MS and NMR-based research, which might suggest some possible biomarkers to support the diagnosis and/or new targets for their treatment.
Collapse
Affiliation(s)
- Melissa Escobar Quintero
- Laboratory of Chemical Biology, Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - João Guilherme de Moraes Pontes
- Laboratory of Chemical Biology, Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Ljubica Tasic
- Laboratory of Chemical Biology, Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
50
|
Klatt S, Doecke JD, Roberts A, Boughton BA, Masters CL, Horne M, Roberts BR. A six-metabolite panel as potential blood-based biomarkers for Parkinson's disease. NPJ Parkinsons Dis 2021; 7:94. [PMID: 34650080 PMCID: PMC8516864 DOI: 10.1038/s41531-021-00239-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 09/13/2021] [Indexed: 12/15/2022] Open
Abstract
Characterisation and diagnosis of idiopathic Parkinson's disease (iPD) is a current challenge that hampers both clinical assessment and clinical trial development with the potential inclusion of non-PD cases. Here, we used a targeted mass spectrometry approach to quantify 38 metabolites extracted from the serum of 231 individuals. This cohort is currently one of the largest metabolomic studies including iPD patients, drug-naïve iPD, healthy controls and patients with Alzheimer's disease as a disease-specific control group. We identified six metabolites (3-hydroxykynurenine, aspartate, beta-alanine, homoserine, ornithine (Orn) and tyrosine) that are significantly altered between iPD patients and control participants. A multivariate model to predict iPD from controls had an area under the curve (AUC) of 0.905, with an accuracy of 86.2%. This panel of metabolites may serve as a potential prognostic or diagnostic assay for clinical trial prescreening, or for aiding in diagnosing pathological disease in the clinic.
Collapse
Affiliation(s)
- Stephan Klatt
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia
- Cooperative Research Centre for Mental Health, Parkville, VIC, 3052, Australia
| | - James D Doecke
- Cooperative Research Centre for Mental Health, Parkville, VIC, 3052, Australia
- Australian e-Health Research Centre, CSIRO, Brisbane, QLD, Australia
| | - Anne Roberts
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Berin A Boughton
- School of Biosciences, The University of Melbourne, Parkville, VIC, 3052, Australia
- Australian National Phenome Centre, Murdoch University, Murdoch, WA, 6150, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia
- Cooperative Research Centre for Mental Health, Parkville, VIC, 3052, Australia
| | - Malcolm Horne
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Blaine R Roberts
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|