1
|
Zhang Y, Zhang CY, Yuan J, Jiang H, Sun P, Hui L, Xu L, Yu L, Guo Z, Wang L, Yang Y, Li M, Li SW, Yang J, Li W, Teng Z, Xiao X. Human mood disorder risk gene Synaptotagmin-14 contributes to mania-like behaviors in mice. Mol Psychiatry 2025:10.1038/s41380-025-02933-1. [PMID: 39966626 DOI: 10.1038/s41380-025-02933-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/30/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
Bipolar disorder (BD) and major depressive disorder (MDD) are the most prevalent mood disorders and cause considerable burden worldwide. Compelling evidence suggests a pronounced overlap between these two disorders in clinical symptoms, treatment strategies, and genetic etiology. Here we leverage a BD GWAS (1822 cases and 4650 controls) and a MDD GWAS (5303 cases and 5337 controls), followed by independent replications, to investigate their shared genetic basis among Han Chinese. We have herein identified a lead SNP rs126277 at the 1q32.2 locus, which also exhibited nominal associations with mood disorders and several relevant sub-clinical phenotypes (e.g., mania) in European populations. Bulk tissue and single-cell eQTL analyses suggest that the risk G-allele of rs126277 predicted lower SYT14 mRNA expression in human brains. We generated mice lacking Syt14 (Syt14-/-) and mice with insufficient expression of Syt14 in the hippocampus (Syt14-KD), and found that depletion of Syt14 resulted in mania-like behaviors including hyperactivity and anti-depressive behaviors, resembling aspects of mood disorders. We also confirmed that deficiency of this gene in the hippocampus was sufficient to induce hyperactivity in mice. RNA-sequencing analyses of the hippocampus of Syt14-/- mice revealed significant upregulation of Per1 as well as downregulation of Slc7a11 and Ptprb. Ultrastructural analyses showed significant alteration of the number of vesicles within 50 nm to the active zone and the width of synaptic cleft in the ventral hippocampus of Syt14-/- mice compared with the control mice. Overall, we have identified a novel mood disorder risk gene SYT14, and confirmed its impact on mania-like behaviors. While the current study identifies an essential mood disorder risk gene, further investigations elucidating the detailed mechanisms by which SYT14 contributes to the pathogenesis of the illnesses are needed.
Collapse
Affiliation(s)
- Yue Zhang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chu-Yi Zhang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jing Yuan
- Department of Psychiatry, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hongyan Jiang
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ping Sun
- Qingdao Mental Health Center, Qingdao, Shandong, China
| | - Li Hui
- Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Li Xu
- Department of Psychiatry, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ling Yu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zeyi Guo
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Lu Wang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yi Yang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Ming Li
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Shi-Wu Li
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jianzhong Yang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, Zhejiang, China
| | - Wei Li
- Department of Blood Transfusion, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhaowei Teng
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xiao Xiao
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
2
|
Yun CS, Hwang YH, Yeon J, Baek HM, Kim DY, Han BS. Effects of Acute Stress on Metabolic Interactions Related to the Tricarboxylic Acid (TCA) Cycle in the Left Hippocampus of Mice. Metabolites 2024; 14:699. [PMID: 39728480 DOI: 10.3390/metabo14120699] [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: 10/18/2024] [Revised: 11/22/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES The acute stress response affects brain metabolites closely linked to the tricarboxylic acid (TCA) cycle. This response involves time-dependent changes in hormones and neurotransmitters, which contribute to resilience and the ability to adapt to acute stress while maintaining homeostasis. This physiological mechanism of metabolic dynamics, combined with time-series analysis, has prompted the development of new methods to observe the relationship between TCA cycle-related brain metabolites. This study aimed to observe the acute stress response through metabolic interactions using time-series proton magnetic resonance spectroscopy (1H-MRS) in the left hippocampus of mice. METHODS In this study, 4-week-old male C57BL/6N mice (n = 24) were divided into control (n = 12) and acute stress groups (n = 12). Acute stress was induced through a 2 h restraint protocol. Time-series 1H-MRS data were obtained on the left hippocampus of both groups using a 9.4 T 1H-MRS scanner. Time-series MRS data were quantified using LCModel, and significant metabolic interactions were identified through Spearman correlation analysis, a one-tailed sign test, and false discovery rate correction. RESULTS No significant metabolic correlation coefficient was observed in the control group. However, in the acute stress group, glutathione (GSH) and N-acetyl aspartate (NAA) showed a significant positive correlation over time, with a high correlation coefficient exceeding 0.5. CONCLUSIONS Temporal measurement of GSH and NAA, combined with correlation analysis, offers a comprehensive understanding for the metabolic dynamics during acute stress. This approach emphasizes their distinct roles and interdependence in the progression of oxidative stress, mitochondrial function, and the maintenance of physiological homeostasis.
Collapse
Affiliation(s)
- Chang-Soo Yun
- Department of Radiation Convergence Engineering, College of Software and Digital Healthcare Convergence, Yonsei University, 1, Yeonsedae-gil, Heungeop-myeon, Wonju 26493, Republic of Korea
| | - Yoon Ho Hwang
- Institute for Human Genomic Study, College of Medicine, Korea University, Seoul 15355, Republic of Korea
| | - Jehyeong Yeon
- Department of Radiation Convergence Engineering, College of Software and Digital Healthcare Convergence, Yonsei University, 1, Yeonsedae-gil, Heungeop-myeon, Wonju 26493, Republic of Korea
| | - Hyeon-Man Baek
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| | - Dong Youn Kim
- Department of Biomedical Engineering, College of Software and Digital Healthcare Convergence, Yonsei University, 1, Yeonsedae-gil, Heungeop-myeon, Wonju 26493, Republic of Korea
| | - Bong Soo Han
- Department of Radiation Convergence Engineering, College of Software and Digital Healthcare Convergence, Yonsei University, 1, Yeonsedae-gil, Heungeop-myeon, Wonju 26493, Republic of Korea
| |
Collapse
|
3
|
Jha S, Torres-Carmona E, Iwata Y, Ma C, Graff-Guerrero A, Fischer CE, Mulsant B, Pollock BG, Rajji TK, Kumar S. Neuronal viability/astrocyte activity ratio in the dorsolateral prefrontal cortex as a biomarker of Alzheimer's dementia: a proton magnetic resonance spectroscopy study. Cereb Cortex 2024; 34:bhae465. [PMID: 39587372 DOI: 10.1093/cercor/bhae465] [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: 06/10/2024] [Revised: 11/04/2024] [Accepted: 11/09/2024] [Indexed: 11/27/2024] Open
Abstract
N-acetyl-aspartate (NAA) and myo-inositol (mI) are neurometabolites reflecting neuronal viability and astrocyte activity, respectively. These are quantified using proton magnetic resonance spectroscopy (1H-MRS) and may be biomarkers for Alzheimer's disease dementia (AD). Our objectives were: 1) Compare dorsolateral prefrontal cortex (DLPFC) NAA and mI levels between AD and cognitively healthy control participants (HC) 2) assess if NAA/mI ratio can distinguish groups, and 3) explore the relationship between metabolites and cognition. The study included 64 participants over 55, 41 with AD. Bilateral DLPFC NAA and mI levels were quantified using 3 T 1H-MRS and normalized to H2O. NAA and NAA/mI ratio were lower in AD vs. HC. mI was unchanged. The NAA/mI ratio at a cut-off value of 1.69 showed 59% sensitivity and 87% specificity at distinguishing AD from HC. NAA was associated positively with cognition. In conclusion, DLPFC metabolite changes suggest altered mitochondrial function in AD. NAA/mI ratio shows good specificity in distinguishing AD from HC, suggesting its role in complementing other biomarkers. Future studies should evaluate NAA/mI ratio with other disease specific biomarkers.
Collapse
Affiliation(s)
- Shreya Jha
- Temerty Faculty of Medicine, University of Toronto, 1 King's College Cir, Toronto, Ontario M5S 1A8, Canada
- Adult Neurodevelopment and Geriatric Psychiatry Division, Centre for Addiction and Mental Health, 1000 Queen St W, Toronto, Ontario M6J 1H4, Canada
| | - Edgardo Torres-Carmona
- Research Imaging Centre, Centre for Addiction and Mental Health, 1000 Queen St W, Toronto, Ontario M6J 1H4, Canada
| | - Yusuke Iwata
- Research Imaging Centre, Centre for Addiction and Mental Health, 1000 Queen St W, Toronto, Ontario M6J 1H4, Canada
| | - Clement Ma
- Adult Neurodevelopment and Geriatric Psychiatry Division, Centre for Addiction and Mental Health, 1000 Queen St W, Toronto, Ontario M6J 1H4, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, 155 College St Room 500, Toronto, Ontario, M5T 3M7, Canada
| | - Ariel Graff-Guerrero
- Adult Neurodevelopment and Geriatric Psychiatry Division, Centre for Addiction and Mental Health, 1000 Queen St W, Toronto, Ontario M6J 1H4, Canada
- Research Imaging Centre, Centre for Addiction and Mental Health, 1000 Queen St W, Toronto, Ontario M6J 1H4, Canada
| | - Corinne E Fischer
- Temerty Faculty of Medicine, University of Toronto, 1 King's College Cir, Toronto, Ontario M5S 1A8, Canada
- Department of Psychiatry, St. Michaels Hospital, 36 Queen St E, Toronto, Ontario M5B 1W8, Canada
| | - Benoit Mulsant
- Temerty Faculty of Medicine, University of Toronto, 1 King's College Cir, Toronto, Ontario M5S 1A8, Canada
- Adult Neurodevelopment and Geriatric Psychiatry Division, Centre for Addiction and Mental Health, 1000 Queen St W, Toronto, Ontario M6J 1H4, Canada
| | - Bruce G Pollock
- Campbell Family Mental Health Research Institute, Division of Geriatric Psychiatry, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario M5T 1R8, Canada
| | - Tarek K Rajji
- Temerty Faculty of Medicine, University of Toronto, 1 King's College Cir, Toronto, Ontario M5S 1A8, Canada
- Adult Neurodevelopment and Geriatric Psychiatry Division, Centre for Addiction and Mental Health, 1000 Queen St W, Toronto, Ontario M6J 1H4, Canada
- Department of Psychiatry, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, United States
| | - Sanjeev Kumar
- Temerty Faculty of Medicine, University of Toronto, 1 King's College Cir, Toronto, Ontario M5S 1A8, Canada
- Adult Neurodevelopment and Geriatric Psychiatry Division, Centre for Addiction and Mental Health, 1000 Queen St W, Toronto, Ontario M6J 1H4, Canada
| |
Collapse
|
4
|
De Felice M, Szkudlarek HJ, Uzuneser TC, Rodríguez-Ruiz M, Sarikahya MH, Pusparajah M, Galindo Lazo JP, Whitehead SN, Yeung KKC, Rushlow WJ, Laviolette SR. The Impacts of Adolescent Cannabinoid Exposure on Striatal Anxiety- and Depressive-Like Pathophysiology Are Prevented by the Antioxidant N-Acetylcysteine. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100361. [PMID: 39257692 PMCID: PMC11381987 DOI: 10.1016/j.bpsgos.2024.100361] [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: 04/05/2024] [Revised: 06/18/2024] [Accepted: 07/09/2024] [Indexed: 09/12/2024] Open
Abstract
Background Exposure to Δ9-tetrahydrocannabinol (THC) is an established risk factor for later-life neuropsychiatric vulnerability, including mood- and anxiety-related symptoms. The psychotropic effects of THC on affect and anxiogenic behavioral phenomena are known to target the striatal network, particularly the nucleus accumbens, a neural region linked to mood and anxiety disorder pathophysiology. THC may increase neuroinflammatory responses via the redox system and dysregulate inhibitory and excitatory neural balance in various brain circuits, including the striatum. Thus, interventions that can induce antioxidant effects may counteract the neurodevelopmental impacts of THC exposure. Methods In the current study, we used an established preclinical adolescent rat model to examine the impacts of adolescent THC exposure on various behavioral, molecular, and neuronal biomarkers associated with increased mood and anxiety disorder vulnerability. Moreover, we investigated the protective properties of the antioxidant N-acetylcysteine against THC-related pathology. Results We demonstrated that adolescent THC exposure induced long-lasting anxiety- and depressive-like phenotypes concomitant with differential neuronal and molecular abnormalities in the two subregions of the nucleus accumbens, the shell and the core. In addition, we report for the first time that N-acetylcysteine can prevent THC-induced accumbal pathophysiology and associated behavioral abnormalities. Conclusions The preventive effects of this antioxidant intervention highlight the critical role of redox mechanisms underlying cannabinoid-induced neurodevelopmental pathology and identify a potential intervention strategy for the prevention and/or reversal of these pathophysiological sequelae.
Collapse
Affiliation(s)
- Marta De Felice
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Hanna J Szkudlarek
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Taygun C Uzuneser
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Mar Rodríguez-Ruiz
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Mohammed H Sarikahya
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | | | | | - Shawn N Whitehead
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Ken K-C Yeung
- Department of Chemistry, Western University, London, Ontario, Canada
- Department of Biochemistry, Western University, London, Ontario, Canada
| | - Walter J Rushlow
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Steven R Laviolette
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- Division of Maternal, Fetal and Newborn Health, Children's Health Research Institute (CHRI), London, Ontario, Canada
| |
Collapse
|
5
|
Hong S, Tomar JS, Shen J. Metabolic coupling between glutamate and N-acetylaspartate in the human brain. J Cereb Blood Flow Metab 2024; 44:1608-1617. [PMID: 38483126 PMCID: PMC11418672 DOI: 10.1177/0271678x241239783] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 09/06/2024]
Abstract
A metabolic coupling between glutamate and N-acetylaspartate measured by in vivo magnetic resonance spectroscopy has been recently reported in the literature with inconsistent findings. In this study, confounders originating from Pearson's spurious correlation of ratios and spectral correlation due to overlapping magnetic resonance spectroscopy signals of glutamate and N-acetylaspartate were practically eliminated to facilitate the determination of any metabolic link between glutamate and N-acetylaspartate in the human brain using in vivo magnetic resonance spectroscopy. In both occipital and medial prefrontal cortices of healthy individuals, correlations between glutamate and N-acetylaspartate were found to be insignificant. Our results do not lend support to a recent hypothesis that N-acetylaspartate serves as a significant reservoir for the rapid replenishment of glutamate during signaling or stress.
Collapse
Affiliation(s)
- Sungtak Hong
- Section on Magnetic Resonance Spectroscopy, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Jyoti Singh Tomar
- Section on Magnetic Resonance Spectroscopy, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Jun Shen
- Section on Magnetic Resonance Spectroscopy, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
6
|
Grønbæk-Thygesen M, Hartmann-Petersen R. Cellular and molecular mechanisms of aspartoacylase and its role in Canavan disease. Cell Biosci 2024; 14:45. [PMID: 38582917 PMCID: PMC10998430 DOI: 10.1186/s13578-024-01224-6] [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: 12/15/2023] [Accepted: 03/24/2024] [Indexed: 04/08/2024] Open
Abstract
Canavan disease is an autosomal recessive and lethal neurological disorder, characterized by the spongy degeneration of the white matter in the brain. The disease is caused by a deficiency of the cytosolic aspartoacylase (ASPA) enzyme, which catalyzes the hydrolysis of N-acetyl-aspartate (NAA), an abundant brain metabolite, into aspartate and acetate. On the physiological level, the mechanism of pathogenicity remains somewhat obscure, with multiple, not mutually exclusive, suggested hypotheses. At the molecular level, recent studies have shown that most disease linked ASPA gene variants lead to a structural destabilization and subsequent proteasomal degradation of the ASPA protein variants, and accordingly Canavan disease should in general be considered a protein misfolding disorder. Here, we comprehensively summarize the molecular and cell biology of ASPA, with a particular focus on disease-linked gene variants and the pathophysiology of Canavan disease. We highlight the importance of high-throughput technologies and computational prediction tools for making genotype-phenotype predictions as we await the results of ongoing trials with gene therapy for Canavan disease.
Collapse
Affiliation(s)
- Martin Grønbæk-Thygesen
- The Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, 2200N, Copenhagen, Denmark.
| | - Rasmus Hartmann-Petersen
- The Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, 2200N, Copenhagen, Denmark.
| |
Collapse
|
7
|
Khan T, Waseem R, Shahid M, Ansari J, Hassan MI, Shamsi A, Islam A. Understanding the Modulation of α-Synuclein Fibrillation by N-Acetyl Aspartate: A Brain Metabolite. ACS OMEGA 2024; 9:12262-12271. [PMID: 38496993 PMCID: PMC10938311 DOI: 10.1021/acsomega.4c00595] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/09/2024] [Accepted: 02/19/2024] [Indexed: 03/19/2024]
Abstract
α-Synuclein (α-Syn) fibrillation is a prominent contributor to neuronal deterioration and plays a significant role in the advancement of Parkinson's Disease (PD). Considering this, the exploration of novel compounds that can inhibit or modulate the aggregation of α-Syn is a topic of significant research. This study, for the first time, elucidated the effect of N-acetyl aspartate (NAA), a brain osmolyte, on α-Syn aggregation using spectroscopic and microscopic approaches. Thioflavin T (ThT) assay revealed that a lower concentration of NAA inhibits α-Syn aggregation, whereas higher concentrations of NAA accelerate the aggregation. Further, this paradoxical effect of NAA was complemented by ANS, RLS, and the turbidity assay. The secondary structure transition was more pronounced at higher concentrations of NAA by circular dichroism, corroborating the fluorescence spectroscopic observations. Confocal microscopy also confirmed the paradoxical effect of NAA on α-Syn aggregation. Interaction studies including fluorescence quenching and molecular docking were employed to determine the binding affinity and critical residues involved in the α-Syn-NAA interaction. The explanation for this paradoxical nature of NAA could be a solvophobic effect. The results offer a profound understanding of the modulatory mechanism of α-Syn aggregation by NAA, thereby suggesting the potential role of NAA at lower concentrations in therapeutics against α-Syn aggregation-related disorders.
Collapse
Affiliation(s)
- Tanzeel Khan
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Rashid Waseem
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Mohammad Shahid
- Department
of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Jaoud Ansari
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Md. Imtaiyaz Hassan
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Anas Shamsi
- Centre
of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman346, United Arab
Emirates
| | - Asimul Islam
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| |
Collapse
|
8
|
Gliozzi M, Coppoletta AR, Cardamone A, Musolino V, Carresi C, Nucera S, Ruga S, Scarano F, Bosco F, Guarnieri L, Macrì R, Mollace R, Belzung C, Mollace V. The dangerous "West Coast Swing" by hyperglycaemia and chronic stress in the mouse hippocampus: Role of kynurenine catabolism. Pharmacol Res 2024; 201:107087. [PMID: 38301816 DOI: 10.1016/j.phrs.2024.107087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/03/2024]
Abstract
Growing epidemiological studies highlight a bi-directional relationship between depressive symptoms and diabetes mellitus. However, the detrimental impact of their co-existence on mental health suggests the need to treat this comorbidity as a separate entity rather than the two different pathologies. Herein, we characterized the peculiar mechanisms activated in mouse hippocampus from the concurrent development of hyperglycaemia, characterizing the different diabetes subtypes, and chronic stress, recognized as a possible factor predisposing to major depression. Our work demonstrates that kynurenine overproduction, leading to apoptosis in the hippocampus, is triggered in a different way depending on hyperglycaemia or chronic stress. Indeed, in the former, kynurenine appears produced by infiltered macrophages whereas, in the latter, peripheral kynurenine preferentially promotes resident microglia activation. In this scenario, QA, derived from kynurenine catabolism, appears a key mediator causing glutamatergic synapse dysfunction and apoptosis, thus contributing to brain atrophy. We demonstrated that the coexistence of hyperglycaemia and chronic stress worsened hippocampal damage through alternative mechanisms, such as GLUT-4 and BDNF down-expression, denoting mitochondrial dysfunction and apoptosis on one hand and evoking the compromission of neurogenesis on the other. Overall, in the degeneration of neurovascular unit, hyperglycaemia and chronic stress interacted each other as the partners of a "West Coast Swing" in which the leading role can be assumed alternatively by each partner of the dance. The comprehension of these mechanisms can open novel perspectives in the management of diabetic/depressed patients, but also in the understanding the pathogenesis of other neurodegenerative disease characterized by the compromission of hippocampal function.
Collapse
Affiliation(s)
- Micaela Gliozzi
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy.
| | - Anna Rita Coppoletta
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy
| | - Antonio Cardamone
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy
| | - Vincenzo Musolino
- Laboratory of Pharmaceutical Biology, Department of Health Sciences, Institute of Research for Food Safety & Health IRC-FSH, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Cristina Carresi
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Saverio Nucera
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy
| | - Stefano Ruga
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy
| | - Federica Scarano
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy
| | - Francesca Bosco
- Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Lorenza Guarnieri
- Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Roberta Macrì
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy
| | - Rocco Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Catherine Belzung
- UMR 1253, iBrain, Inserm, Université de Tours, CEDEX 1, 37032 Tours, France
| | - Vincenzo Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
9
|
Annunziato M, Bashirova N, Eeza MNH, Lawson A, Fernandez-Lima F, Tose LV, Matysik J, Alia A, Berry JP. An Integrated Metabolomics-Based Model, and Identification of Potential Biomarkers, of Perfluorooctane Sulfonic Acid Toxicity in Zebrafish Embryos. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2024. [PMID: 38411227 DOI: 10.1002/etc.5824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/28/2023] [Accepted: 01/08/2024] [Indexed: 02/28/2024]
Abstract
Known for their high stability and surfactant properties, per- and polyfluoroalkyl substances (PFAS) have been widely used in a range of manufactured products. Despite being largely phased out due to concerns regarding their persistence, bioaccumulation, and toxicity, legacy PFAS such as perfluorooctanesulfonic acid (PFOS) and perfluorooctanoic acid continue to persist at high levels in the environment, posing risks to aquatic organisms. We used high-resolution magic angle spinning nuclear magnetic resonance spectroscopy in intact zebrafish (Danio rerio) embryos to investigate the metabolic pathways altered by PFOS both before and after hatching (i.e., 24 and 72 h post fertilization [hpf], respectively). Assessment of embryotoxicity found embryo lethality in the parts-per-million range with no significant difference in mortality between the 24- and 72-hpf exposure groups. Metabolic profiling revealed mostly consistent changes between the two exposure groups, with altered metabolites generally associated with oxidative stress, lipid metabolism, energy production, and mitochondrial function, as well as specific targeting of the liver and central nervous system as key systems. These metabolic changes were further supported by analyses of tissue-specific production of reactive oxygen species, as well as nontargeted mass spectrometric lipid profiling. Our findings suggest that PFOS-induced metabolic changes in zebrafish embryos may be mediated through previously described interactions with regulatory and transcription factors leading to disruption of mitochondrial function and energy metabolism. The present study proposes a systems-level model of PFOS toxicity in early life stages of zebrafish, and also identifies potential biomarkers of effect and exposure for improved environmental biomonitoring. Environ Toxicol Chem 2024;00:1-19. © 2024 SETAC.
Collapse
Affiliation(s)
- Mark Annunziato
- Institute of Environment, Florida International University, Miami, Florida, USA
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
- Biomolecular Science Institute, Florida International University, Miami, Florida, USA
| | - Narmin Bashirova
- Institute for Analytical Chemistry, University of Leipzig, Leipzig, Germany
| | - Muhamed N H Eeza
- Institute for Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
| | - Ariel Lawson
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
| | - Francisco Fernandez-Lima
- Institute of Environment, Florida International University, Miami, Florida, USA
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
- Biomolecular Science Institute, Florida International University, Miami, Florida, USA
| | - Lilian V Tose
- Institute of Environment, Florida International University, Miami, Florida, USA
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
- Biomolecular Science Institute, Florida International University, Miami, Florida, USA
| | - Jörg Matysik
- Institute for Analytical Chemistry, University of Leipzig, Leipzig, Germany
| | - A Alia
- Institute for Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - John P Berry
- Institute of Environment, Florida International University, Miami, Florida, USA
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
- Biomolecular Science Institute, Florida International University, Miami, Florida, USA
| |
Collapse
|
10
|
Huang H, Zhang M, Zhao Y, Li Y, Jin W, Guo R, Liu W, Cai B, Li J, Yuan S, Huang X, Lin X, Liang ZP, Li B, Luo J. Simultaneous high-resolution whole-brain MR spectroscopy and [ 18F]FDG PET for temporal lobe epilepsy. Eur J Nucl Med Mol Imaging 2024; 51:721-733. [PMID: 37823910 DOI: 10.1007/s00259-023-06465-0] [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: 07/23/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023]
Abstract
PURPOSE Precise lateralizing the epileptogenic zone in patients with drug-resistant mesial temporal lobe epilepsy (mTLE) remains challenging, particularly when routine MRI scans are inconclusive (MRI-negative). This study aimed to investigate the synergy of fast, high-resolution, whole-brain MRSI in conjunction with simultaneous [18F]FDG PET for the lateralization of mTLE. METHODS Forty-eight drug-resistant mTLE patients (M/F 31/17, age 12-58) underwent MRSI and [18F]FDG PET on a hybrid PET/MR scanner. Lateralization of mTLE was evaluated by visual inspection and statistical classifiers of metabolic mappings against routine MRI. Additionally, this study explored how disease status influences the associations between altered N-acetyl aspartate (NAA) and FDG uptake using hierarchical moderated multiple regression. RESULTS The high-resolution whole-brain MRSI data offers metabolite maps at comparable resolution to [18F]FDG PET. Visual examinations of combined MRSI and [18F]FDG PET showed an mTLE lateralization accuracy rate of 91.7% in a 48-patient cohort, surpassing routine MRI (52.1%). Notably, out of 23 MRI-negative mTLE, combined MRSI and [18F]FDG PET helped detect 19 cases. Logistical regression models combining hippocampal NAA level and FDG uptake improved lateralization performance (AUC=0.856), while further incorporating extrahippocampal regions such as amygdala, thalamus, and superior temporal gyrus increased the AUC to 0.939. Concurrent MRSI/PET revealed a moderating influence of disease duration and hippocampal atrophy on the association between hippocampal NAA and glucose uptake, providing significant new insights into the disease's trajectory. CONCLUSION This paper reports the first metabolic imaging study using simultaneous high-resolution MRSI and [18F]FDG PET, which help visualize MRI-unidentifiable lesions and may thus advance diagnostic tools and management strategies for drug-resistant mTLE.
Collapse
Affiliation(s)
- Hui Huang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Miao Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yibo Zhao
- Beckman Institute for Advanced Sciences and Technology, University of Illinois at Urbana Champaign, Urbana, IL, 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana Champaign, Urbana, IL, 61801, USA
| | - Yudu Li
- Beckman Institute for Advanced Sciences and Technology, University of Illinois at Urbana Champaign, Urbana, IL, 61801, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana Champaign, Urbana, IL, 61801, USA
| | - Wen Jin
- Beckman Institute for Advanced Sciences and Technology, University of Illinois at Urbana Champaign, Urbana, IL, 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana Champaign, Urbana, IL, 61801, USA
| | - Rong Guo
- Beckman Institute for Advanced Sciences and Technology, University of Illinois at Urbana Champaign, Urbana, IL, 61801, USA
- Siemens Medical Solutions USA, Inc, Urbana, IL, 61801, USA
| | - Wei Liu
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Bingyang Cai
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jiwei Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Siyu Yuan
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xinyun Huang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaozhu Lin
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhi-Pei Liang
- Beckman Institute for Advanced Sciences and Technology, University of Illinois at Urbana Champaign, Urbana, IL, 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana Champaign, Urbana, IL, 61801, USA
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jie Luo
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
11
|
Gonsalves MA, White TL, Barredo J, DeMayo MM, DeLuca E, Harris AD, Carpenter LL. Cortical glutamate, Glx, and total N-acetylaspartate: potential biomarkers of repetitive transcranial magnetic stimulation treatment response and outcomes in major depression. Transl Psychiatry 2024; 14:5. [PMID: 38184652 PMCID: PMC10771455 DOI: 10.1038/s41398-023-02715-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 01/08/2024] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is an effective treatment for individuals with major depressive disorder (MDD) who have not improved with standard therapies. However, only 30-45% of patients respond to rTMS. Predicting response to rTMS will benefit both patients and providers in terms of prescribing and targeting treatment for maximum efficacy and directing resources, as individuals with lower likelihood of response could be redirected to more suitable treatment alternatives. In this exploratory study, our goal was to use proton magnetic resonance spectroscopy to examine how glutamate (Glu), Glx, and total N-acetylaspartate (tNAA) predict post-rTMS changes in overall MDD severity and symptoms, and treatment response. Metabolites were measured in a right dorsal anterior cingulate cortex voxel prior to a standard course of 10 Hz rTMS to the left DLPFC in 25 individuals with MDD. MDD severity and symptoms were evaluated via the Inventory of Depression Symptomatology Self-Report (IDS-SR). rTMS response was defined as ≥50% change in full-scale IDS-SR scores post treatment. Percent change in IDS-SR symptom domains were evaluated using principal component analysis and established subscales. Generalized linear and logistic regression models were used to evaluate the relationship between baseline Glu, Glx, and tNAA and outcomes while controlling for age and sex. Participants with baseline Glu and Glx levels in the lower range had greater percent change in full scale IDS-SR scores post-treatment (p < 0.001), as did tNAA (p = 0.007). Low glutamatergic metabolite levels also predicted greater percent change in mood/cognition symptoms (p ≤ 0.001). Low-range Glu, Glx, and tNAA were associated with greater improvement on the immuno-metabolic subscale (p ≤ 0.003). Baseline Glu predicted rTMS responder status (p = 0.025) and had an area under the receiving operating characteristic curve of 0.81 (p = 0.009), demonstrating excellent discriminative ability. Baseline Glu, Glx, and tNAA significantly predicted MDD improvement after rTMS; preliminary evidence also demonstrates metabolite association with symptom subdomain improvement post-rTMS. This work provides feasibility for a personalized medicine approach to rTMS treatment selection, with individuals with Glu levels in the lower range potentially being the best candidates.
Collapse
Affiliation(s)
- Meghan A Gonsalves
- Neuroscience Graduate Program, Brown University, Providence, RI, USA.
- Butler Hospital Neuromodulation Research Facility, Providence, RI, USA.
- Center of Biomedical Research Excellence (COBRE) for Neuromodulation, Butler Hospital, Providence, RI, USA.
| | - Tara L White
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA
- Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI, USA
- Carney Institute for Brain Sciences, Brown University, Providence, RI, USA
| | - Jennifer Barredo
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, RI, USA
- Providence VA Medical Center, Providence, RI, USA
- Clinical Neuroimaging Research Core, Brown University, Providence, RI, USA
| | - Marilena M DeMayo
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Emily DeLuca
- Clinical Neuroimaging Research Core, Brown University, Providence, RI, USA
| | - Ashley D Harris
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Linda L Carpenter
- Butler Hospital Neuromodulation Research Facility, Providence, RI, USA
- Center of Biomedical Research Excellence (COBRE) for Neuromodulation, Butler Hospital, Providence, RI, USA
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, RI, USA
| |
Collapse
|
12
|
Just N, Chevillard PM, Batailler M, Dubois JP, Vaudin P, Pillon D, Migaud M. Multiparametric MR Evaluation of the Photoperiodic Regulation of Hypothalamic Structures in Sheep. Neuroscience 2023; 535:142-157. [PMID: 37913859 DOI: 10.1016/j.neuroscience.2023.10.022] [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: 08/25/2023] [Revised: 10/16/2023] [Accepted: 10/24/2023] [Indexed: 11/03/2023]
Abstract
Most organisms on earth, humans included, have developed strategies to cope with environmental day-night and seasonal cycles to survive. For most of them, their physiological and behavioral functions, including the reproductive function, are synchronized with the annual changes of day length, to ensure winter survival and subsequent reproductive success in the following spring. Sheep are sensitive to photoperiod, which also regulates natural adult neurogenesis in their hypothalamus. We postulate that the ovine model represents a good alternative to study the functional and metabolic changes occurring in response to photoperiodic changes in hypothalamic structures of the brain. Here, the impact of the photoperiod on the neurovascular coupling and the metabolism of the hypothalamic structures was investigated at 3T using BOLD fMRI, perfusion-MRI and proton magnetic resonance spectroscopy (1H-MRS). A longitudinal study involving 8 ewes was conducted during long days (LD) and short days (SD) revealing significant BOLD, rCBV and metabolic changes in hypothalamic structures of the ewe brain between LD and SD. More specifically, the transition between LD and SD revealed negative BOLD responses to hypercapnia at the beginning of SD period followed by significant increases in BOLD, rCBV, Glx and tNAA concentrations towards the end of the SD period. These observations suggest longitudinal mechanisms promoting the proliferation and differentiation of neural stem cells within the hypothalamic niche of breeding ewes. We conclude that multiparametric MRI studies including 1H-MRS could be promising non-invasive translational techniques to investigate the existence of natural adult neurogenesis in-vivo in gyrencephalic brains.
Collapse
Affiliation(s)
- Nathalie Just
- INRAE Centre Val de Loire, UMR Physiologie de la Reproduction et des Comportements CNRS, IFCE, INRAE, Université de Tours, 37380 Nouzilly France; Danish Research Centre for Magnetic Resonance (DRCMR), Hvidovre, Denmark.
| | - Pierre Marie Chevillard
- INRAE Centre Val de Loire, UMR Physiologie de la Reproduction et des Comportements CNRS, IFCE, INRAE, Université de Tours, 37380 Nouzilly France
| | - Martine Batailler
- INRAE Centre Val de Loire, UMR Physiologie de la Reproduction et des Comportements CNRS, IFCE, INRAE, Université de Tours, 37380 Nouzilly France
| | - Jean-Philippe Dubois
- INRAE Centre Val de Loire, UMR Physiologie de la Reproduction et des Comportements CNRS, IFCE, INRAE, Université de Tours, 37380 Nouzilly France
| | - Pascal Vaudin
- INRAE Centre Val de Loire, UMR Physiologie de la Reproduction et des Comportements CNRS, IFCE, INRAE, Université de Tours, 37380 Nouzilly France
| | - Delphine Pillon
- INRAE Centre Val de Loire, UMR Physiologie de la Reproduction et des Comportements CNRS, IFCE, INRAE, Université de Tours, 37380 Nouzilly France
| | - Martine Migaud
- INRAE Centre Val de Loire, UMR Physiologie de la Reproduction et des Comportements CNRS, IFCE, INRAE, Université de Tours, 37380 Nouzilly France
| |
Collapse
|
13
|
Baranovicova E, Kalenska D, Kaplan P, Kovalska M, Tatarkova Z, Lehotsky J. Blood and Brain Metabolites after Cerebral Ischemia. Int J Mol Sci 2023; 24:17302. [PMID: 38139131 PMCID: PMC10743907 DOI: 10.3390/ijms242417302] [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: 10/27/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
The study of an organism's response to cerebral ischemia at different levels is essential to understanding the mechanism of the injury and protection. A great interest is devoted to finding the links between quantitative metabolic changes and post-ischemic damage. This work aims to summarize the outcomes of the most studied metabolites in brain tissue-lactate, glutamine, GABA (4-aminobutyric acid), glutamate, and NAA (N-acetyl aspartate)-regarding their biological function in physiological conditions and their role after cerebral ischemia/reperfusion. We focused on ischemic damage and post-ischemic recovery in both experimental-including our results-as well as clinical studies. We discuss the role of blood glucose in view of the diverse impact of hyperglycemia, whether experimentally induced, caused by insulin resistance, or developed as a stress response to the cerebral ischemic event. Additionally, based on our and other studies, we analyze and critically discuss post-ischemic alterations in energy metabolites and the elevation of blood ketone bodies observed in the studies on rodents. To complete the schema, we discuss alterations in blood plasma circulating amino acids after cerebral ischemia. So far, no fundamental brain or blood metabolite(s) has been recognized as a relevant biological marker with the feasibility to determine the post-ischemic outcome or extent of ischemic damage. However, studies from our group on rats subjected to protective ischemic preconditioning showed that these animals did not develop post-ischemic hyperglycemia and manifested a decreased metabolic infringement and faster metabolomic recovery. The metabolomic approach is an additional tool for understanding damaging and/or restorative processes within the affected brain region reflected in the blood to uncover the response of the whole organism via interorgan metabolic communications to the stressful cerebral ischemic challenge.
Collapse
Affiliation(s)
- Eva Baranovicova
- Biomedical Center BioMed, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia;
| | - Dagmar Kalenska
- Department of Anatomy, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia
| | - Peter Kaplan
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia (Z.T.)
| | - Maria Kovalska
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia
| | - Zuzana Tatarkova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia (Z.T.)
| | - Jan Lehotsky
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia (Z.T.)
| |
Collapse
|
14
|
Ernst T, Ryan MC, Liang HJ, Wang JP, Cunningham E, Saleh MG, Kottilil S, Chang L. Neuronal and Glial Metabolite Abnormalities in Participants With Persistent Neuropsychiatric Symptoms After COVID-19: A Brain Proton Magnetic Resonance Spectroscopy Study. J Infect Dis 2023; 228:1559-1570. [PMID: 37540098 PMCID: PMC10681871 DOI: 10.1093/infdis/jiad309] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 06/01/2023] [Accepted: 08/02/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND The aim of this study was to determine whether neurometabolite abnormalities indicating neuroinflammation and neuronal injury are detectable in individuals post-coronavirus disease 2019 (COVID-19) with persistent neuropsychiatric symptoms. METHODS All participants were studied with proton magnetic resonance spectroscopy at 3 T to assess neurometabolite concentrations (point-resolved spectroscopy, relaxation time/echo time = 3000/30 ms) in frontal white matter (FWM) and anterior cingulate cortex-gray matter (ACC-GM). Participants also completed the National Institutes of Health Toolbox cognition and motor batteries and selected modules from the Patient-Reported Outcomes Measurement Information System. RESULTS Fifty-four participants were evaluated: 29 post-COVID-19 (mean ± SD age, 42.4 ± 12.3 years; approximately 8 months from COVID-19 diagnosis; 19 women) and 25 controls (age, 44.1 ± 12.3 years; 14 women). When compared with controls, the post-COVID-19 group had lower total N-acetyl compounds (tNAA; ACC-GM: -5.0%, P = .015; FWM: -4.4%, P = .13), FWM glutamate + glutamine (-9.5%, P = .001), and ACC-GM myo-inositol (-6.2%, P = .024). Additionally, only hospitalized patients post-COVID-19 showed age-related increases in myo-inositol, choline compounds, and total creatine (interaction P = .029 to <.001). Across all participants, lower FWM tNAA and higher ACC-GM myo-inositol predicted poorer performance on several cognitive measures (P = .001-.009), while lower ACC-GM tNAA predicted lower endurance on the 2-minute walk (P = .005). CONCLUSIONS In participants post-COVID-19 with persistent neuropsychiatric symptoms, the lower-than-normal tNAA and glutamate + glutamine indicate neuronal injury, while the lower-than-normal myo-inositol reflects glial dysfunction, possibly related to mitochondrial dysfunction and oxidative stress in Post-COVID participants with persistent neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Thomas Ernst
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland
- Department of Neurology, School of Medicine, Johns Hopkins University
| | - Meghann C Ryan
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland
- Program in Neuroscience, School of Medicine, University of Maryland
| | - Hua-Jun Liang
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland
| | - Justin P Wang
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland
| | - Eric Cunningham
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland
| | - Muhammad G Saleh
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland
| | - Shyamasundaran Kottilil
- Institute of Human Virology, Division of Infectious Disease, Department of Medicine, School of Medicine, University of Maryland
| | - Linda Chang
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland
- Department of Neurology, School of Medicine, Johns Hopkins University
- Department of Neurology, School of Medicine, University of Maryland, Baltimore
| |
Collapse
|
15
|
Ryberg M, Boraxbekk CJ, Kjaer M, Demnitz N. Effects of acute physical activity on brain metabolites as measured by magnetic resonance spectroscopy ( 1H-MRS) in humans: A systematic review. Heliyon 2023; 9:e20534. [PMID: 37818016 PMCID: PMC10560775 DOI: 10.1016/j.heliyon.2023.e20534] [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: 05/03/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023] Open
Abstract
Physical activity (PA) promotes brain health in a variety of domains including cognition, mood, and neuroplasticity. At the neurochemical level, the mechanisms underlying these effects in the brain are not fully understood. With proton Magnetic Resonance Spectroscopy (1H-MRS), it is possible to non-invasively quantify metabolite concentrations, enabling studies to obtain measures of exercise-induced neurochemical changes. This systematic review aimed to examine the existing literature on acute effects of PA on brain metabolites as measured by 1H-MRS. Four databases (Cochrane Central Register of Controlled Trials, PubMed, Embase, and PsycINFO) were searched, identifying 2965 studies, of which 9 met the inclusion criteria. Across studies, Gamma-AminoButyric Acid (GABA) and lactate tended to increase after exercise, while no significant changes in choline were reported. For glutamine/glutamate (Glx), studies were inconclusive. Conclusions were limited by the lack of consensus on 1H-MRS data processing and exercise protocols. To reduce inter-study differences, future studies are recommended to (1): apply a standardized exercise index (2), consider the onset time of MRS scans, and (3) follow standardized MRS quantification methods.
Collapse
Affiliation(s)
- Mathias Ryberg
- Institute of Sports Medicine Copenhagen (ISMC), Copenhagen University Hospital – Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400 Copenhagen NV, Denmark
| | - Carl-Johan Boraxbekk
- Institute of Sports Medicine Copenhagen (ISMC), Copenhagen University Hospital – Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400 Copenhagen NV, Denmark
- Danish Research Center for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital – Amager and Hvidovre, Kettegård Allé 30, 2650 Hvidovre, Denmark
- Department of Neurology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Institute for Clinical Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Kjaer
- Institute of Sports Medicine Copenhagen (ISMC), Copenhagen University Hospital – Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400 Copenhagen NV, Denmark
- Institute for Clinical Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Healthy Aging, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Naiara Demnitz
- Danish Research Center for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital – Amager and Hvidovre, Kettegård Allé 30, 2650 Hvidovre, Denmark
| |
Collapse
|
16
|
McKiernan E, Su L, O'Brien J. MRS in neurodegenerative dementias, prodromal syndromes and at-risk states: A systematic review of the literature. NMR IN BIOMEDICINE 2023; 36:e4896. [PMID: 36624067 DOI: 10.1002/nbm.4896] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND In recent years, MRS has benefited from increased MRI field strengths, new acquisition protocols and new processing techniques. This review aims to determine how this has altered our understanding of MRS neurometabolic markers in neurodegenerative dementias. METHODS Our systematic review of human in vivo MRS literature since 2002 pertains to Alzheimer's disease (AD), dementia with Lewy bodies (DLB), Parkinson's disease dementia, frontotemporal dementia (FTD), prodromal and 'at-risk' states. Studies using field strengths of 3 T or more were included. RESULTS Of 85 studies, AD and/or mild cognitive impairment (MCI) were the most common conditions of interest (58 papers, 68%). Only 14 (16%) studies included other dementia syndromes and 13 (15%) investigated 'at-risk' cohorts. Earlier findings of lower N-acetylaspartate and higher myo-inositol were confirmed. Additionally, lower choline and creatine in AD and MCI were reported, though inconsistently. Previously challenging-to-measure metabolites (glutathione, glutamate and gamma-aminobutyric acid) were reportedly lower in AD, FTD and DLB compared with controls. DISCUSSION Increasing field strength alongside targeted acquisition protocols has revealed additional metabolite changes. Most studies were small and regional metabolite differences between dementia types may not have been captured due to the predominant placement of voxels in the posterior cingulate cortex. The standard of data collection, quality control and analysis is improving due to greater consensus regarding acquisition and processing techniques. Ongoing harmonization of techniques, creation of larger and longitudinal cohorts, and placement of MRS voxels in more diverse regions will strengthen future research.
Collapse
Affiliation(s)
- Elizabeth McKiernan
- Department of Psychiatry, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Li Su
- Department of Psychiatry, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - John O'Brien
- Department of Psychiatry, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| |
Collapse
|
17
|
Kumari M, Arora P, Sharma P, Hasija Y, Rana P, D'souza MM, Chandra N, Trivedi R. Acute metabolic alterations in the hippocampus are associated with decreased acetylation after blast induced TBI. Metabolomics 2023; 19:5. [PMID: 36635559 DOI: 10.1007/s11306-022-01970-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 12/21/2022] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Blast induced Traumatic brain injury (BI-TBI) is common among military personnels as well as war affected civilians. In the war zone, people can also encounter repeated exposure of blast wave, which may affect their cognition and metabolic alterations. OBJECTIVE In this study we assess the metabolic and histological changes in the hippocampus of rats at 24 h post injury. METHOD Rats were divided into four groups: (i) Sham; (ii) Mild TBI (mi); (iii) Moderate TBI (mo); and (iv) Repetitive mild TBI (rm TBI) and then subjected to different intensities of blast exposure. Hippocampal tissues were collected after 24 h of injury for proton nuclear magnetic resonance spectroscopy (1H NMR spectroscopy) and immunohistochemical (IHC) analysis. RESULTS The metabolic alterations were found in the hippocampal tissue samples and these alterations showed significant change in glutamate, N-Acetylaspartic acid (NAA), acetate, creatine, phosphoethanolamine (PE), ethanolamine and PC/choline concentrations in rmTBI rats only. IHC studies revealed that AH3 (Acetyl histone) positive cells were decreased in rm TBI tissue samples in comparison to other TBI groups and sham rats. This might reflect an epigenetic alteration due to repeated blast exposure at 24 h post injury. Additionally, astrogliosis was observed in miTBI and moTBI hippocampal tissue while no change was observed in rmTBI tissues. CONCLUSION The present study reports altered acetylation in the presence of altered metabolism in hippocampal tissue of blast induced rmTBI at 24 h post injury. Mechanistic understanding of these intertwined processes may help in the development of better therapeutic pathways and agents for blast induced TBI in near future.
Collapse
Affiliation(s)
- Megha Kumari
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Science (INMAS), DRDO, Delhi, India
- Department of Biotechnology, Delhi Technological University, Delhi, 110042, India
| | - Palkin Arora
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Science (INMAS), DRDO, Delhi, India
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | - Priyanka Sharma
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Science (INMAS), DRDO, Delhi, India
| | - Yasha Hasija
- Department of Biotechnology, Delhi Technological University, Delhi, 110042, India
| | - Poonam Rana
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Science (INMAS), DRDO, Delhi, India
| | - Maria M D'souza
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Science (INMAS), DRDO, Delhi, India
| | - Namas Chandra
- Center for Injury Biomechanics, New Jersey Institute of Technology, 323 Dr Martin Luther King Jr Blvd, Newark, NJ, 07102, USA
| | - Richa Trivedi
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Science (INMAS), DRDO, Delhi, India.
| |
Collapse
|
18
|
Bonnekoh LM, Seidenbecher S, Knigge K, Hünecke AK, Metzger CD, Tempelmann C, Kanowski M, Kaufmann J, Meyer-Lotz G, Schlaaff K, Dobrowolny H, Tozzi L, Gescher DM, Steiner J, Kirschbaum C, Frodl T. Long-term cortisol stress response in depression and comorbid anxiety is linked with reduced N-acetylaspartate in the anterior cingulate cortex. World J Biol Psychiatry 2023; 24:34-45. [PMID: 35332851 DOI: 10.1080/15622975.2022.2058084] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVES Major Depression (MDD) and anxiety disorders are stress-related disorders that share pathophysiological mechanisms. There is evidence for alterations of glutamate-glutamine, N-acetylaspartate (NAA) and GABA in the anterior cingulate cortex (ACC), a stress-sensitive region affected by hypothalamic-pituitary-adrenal axis (HPA). The aim was to investigate metabolic alterations in the ACC and whether hair cortisol, current stress or early life adversity predict them. METHODS We investigated 22 patients with MDD and comorbid anxiety disorder and 23 healthy controls. Proton magnetic resonance spectroscopy was performed with voxels placed in pregenual (pg) and dorsal (d) ACC in 3 T. Analysis of hair cortisol was performed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS The N-acetylaspartate/Creatin ratio (NAA/Cr) was reduced in patients in both pgACC (p = .040) and dACC (p = .016). A significant interactive effect of diagnosis and cortisol on both pg-NAA/Cr (F = 5.00, p = .033) and d-NAA/Cr (F = 7.86, p = .009) was detected, whereby in controls cortisol was positively correlated with d-NAA/Cr (r = 0.61, p = .004). CONCLUSIONS Our results suggest a relationship between NAA metabolism in ACC and HPA axis activity as represented by long-term cortisol output.
Collapse
Affiliation(s)
- Linda M Bonnekoh
- Department of Psychiatry and Psychotherapy, Otto von Guericke University, Magdeburg, Germany.,Department of Psychiatry and Psychotherapy, University of Münster, Münster, Germany.,Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Münster, Münster, Germany
| | - Stephanie Seidenbecher
- Department of Psychiatry and Psychotherapy, Otto von Guericke University, Magdeburg, Germany
| | - Katrin Knigge
- Department of Psychiatry and Psychotherapy, Otto von Guericke University, Magdeburg, Germany
| | - Anne-Kathrin Hünecke
- Department of Psychiatry and Psychotherapy, Otto von Guericke University, Magdeburg, Germany
| | - Coraline D Metzger
- Department of Psychiatry and Psychotherapy, Otto von Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Claus Tempelmann
- Department of Neurology, Otto von Guericke University, Magdeburg, Germany.,Center of Behavioral Brain Sciences (CBBS), Otto von Guericke Universität Magdeburg, Magdeburg, Germany
| | - Martin Kanowski
- Department of Neurology, Otto von Guericke University, Magdeburg, Germany
| | - Jörn Kaufmann
- Department of Neurology, Otto von Guericke University, Magdeburg, Germany
| | - Gabriela Meyer-Lotz
- Department of Psychiatry and Psychotherapy, Otto von Guericke University, Magdeburg, Germany
| | - Konstantin Schlaaff
- Department of Psychiatry and Psychotherapy, Otto von Guericke University, Magdeburg, Germany.,Translational Psychiatry Laboratory, Otto von Guericke University, Magdeburg, Germany
| | - Henrik Dobrowolny
- Department of Psychiatry and Psychotherapy, Otto von Guericke University, Magdeburg, Germany
| | - Leonardo Tozzi
- Department of Psychiatry and Psychotherapy, Otto von Guericke University, Magdeburg, Germany.,Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Dorothee M Gescher
- Department of Psychiatry and Psychotherapy, Otto von Guericke University, Magdeburg, Germany.,Department of Psychiatry and Psychotherapy, University RWTH, Aachen, Germany
| | - Johann Steiner
- Department of Psychiatry and Psychotherapy, Otto von Guericke University, Magdeburg, Germany.,Translational Psychiatry Laboratory, Otto von Guericke University, Magdeburg, Germany
| | - Clemens Kirschbaum
- Department of Psychology, Dresden University of Technology, Dresden, Germany
| | - Thomas Frodl
- Department of Psychiatry and Psychotherapy, Otto von Guericke University, Magdeburg, Germany.,Center of Behavioral Brain Sciences (CBBS), Otto von Guericke Universität Magdeburg, Magdeburg, Germany.,Department of Psychiatry and Psychotherapy, University RWTH, Aachen, Germany
| |
Collapse
|
19
|
Chabert J, Allauze E, Pereira B, Chassain C, De Chazeron I, Rotgé JY, Fossati P, Llorca PM, Samalin L. Glutamatergic and N-Acetylaspartate Metabolites in Bipolar Disorder: A Systematic Review and Meta-Analysis of Proton Magnetic Resonance Spectroscopy Studies. Int J Mol Sci 2022; 23:ijms23168974. [PMID: 36012234 PMCID: PMC9409038 DOI: 10.3390/ijms23168974] [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: 07/08/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/26/2022] Open
Abstract
The exact neurobiological mechanisms of bipolar disorder (BD) remain unknown. However, some neurometabolites could be implicated, including Glutamate (Glu), Glutamine (Gln), Glx, and N-acetylaspartate (NAA). Proton Magnetic Resonance Spectroscopy (1H-MRS) allows one to quantify these metabolites in the human brain. Thus, we conducted a systematic review and meta-analysis of the literature to compare their levels between BD patients and healthy controls (HC). The main inclusion criteria for inclusion were 1H-MRS studies comparing levels of Glu, Gln, Glx, and NAA in the prefrontal cortex (PFC), anterior cingulate cortex (ACC), and hippocampi between patients with BD in clinical remission or a major depressive episode and HC. Thirty-three studies were included. NAA levels were significantly lower in the left white matter PFC (wmPFC) of depressive and remitted BD patients compared to controls and were also significantly higher in the left dorsolateral PFC (dlPFC) of depressive BD patients compared to HC. Gln levels were significantly higher in the ACC of remitted BD patients compared to in HC. The decreased levels of NAA of BD patients may be related to the alterations in neuroplasticity and synaptic plasticity found in BD patients and may explain the deep white matter hyperintensities frequently observed via magnetic resonance imagery.
Collapse
Affiliation(s)
- Jonathan Chabert
- Service de Psychiatrie Adulte, CHU Clermont-Ferrand, CNRS, Institut Pascal, Université Clermont Auvergne, 58 Rue Montalembert, 63003 Clermont-Ferrand, France
- Correspondence: (J.C.); (L.S.); Tel.: +33-4-73-752-124 (J.C. & L.S.)
| | - Etienne Allauze
- Service de Psychiatrie Adulte, CHU Clermont-Ferrand, CNRS, Institut Pascal, Université Clermont Auvergne, 58 Rue Montalembert, 63003 Clermont-Ferrand, France
| | - Bruno Pereira
- Biostatistics Unit (DRCI), CHU Clermont-Ferrand, Université Clermont Auvergne, 7 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Carine Chassain
- Imaging Department, CHU Clermont-Ferrand, CNRS, Institut Pascal, Université Clermont Auvergne, Clermont Auvergne INP, 58 Rue Montalembert, 63003 Clermont-Ferrand, France
| | - Ingrid De Chazeron
- Service de Psychiatrie Adulte, CHU Clermont-Ferrand, CNRS, Institut Pascal, Université Clermont Auvergne, 58 Rue Montalembert, 63003 Clermont-Ferrand, France
| | - Jean-Yves Rotgé
- Service de Psychiatrie Adulte, Pitié-Salpêtrière Hospital, CNRS UMR 7593, 47-83 Bd de l’Hôpital, 75651 Paris, France
| | - Philippe Fossati
- Service de Psychiatrie Adulte, Pitié-Salpêtrière Hospital, CNRS UMR 7593, 47-83 Bd de l’Hôpital, 75651 Paris, France
| | - Pierre-Michel Llorca
- Service de Psychiatrie Adulte, CHU Clermont-Ferrand, CNRS, Institut Pascal, Université Clermont Auvergne, 58 Rue Montalembert, 63003 Clermont-Ferrand, France
| | - Ludovic Samalin
- Service de Psychiatrie Adulte, CHU Clermont-Ferrand, CNRS, Institut Pascal, Université Clermont Auvergne, 58 Rue Montalembert, 63003 Clermont-Ferrand, France
- Correspondence: (J.C.); (L.S.); Tel.: +33-4-73-752-124 (J.C. & L.S.)
| |
Collapse
|
20
|
Moxon-Emre I, Croarkin PE, Daskalakis ZJ, Blumberger DM, Lyon RE, Tani H, Truong P, Lai MC, Desarkar P, Sailasuta N, Szatmari P, Ameis SH. NAA/Glu Ratio Associated with Suicidal Ideation in Pilot Sample of Autistic Youth and Young Adults. Brain Sci 2022; 12:785. [PMID: 35741670 PMCID: PMC9220790 DOI: 10.3390/brainsci12060785] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 02/01/2023] Open
Abstract
Suicidality is increased in autism spectrum disorder (ASD), yet effective interventions are lacking. Developing biologically based approaches for preventing and treating suicidality in ASD hinges on the identification of biomarkers of suicidal ideation (SI). Here, we assessed magnetic resonance spectroscopy (MRS) markers of glutamatergic neurotransmission in ASD youth and young adults. Twenty-eight ASD participants (16-33 years) underwent 1H-MRS, and metabolites were quantified using LCModel. N-acetylaspartate (NAA), glutamate (Glu), and the NAA/Glu ratio from the left dorsolateral prefrontal cortex were compared between ASD SI+ (n = 13) and ASD SI- (n = 15) participants. We found that ASD SI+ participants had a higher NAA/Glu ratio compared ASD SI- participants. The NAA/Glu ratio also predicted SI and significantly discriminated between ASD SI+/SI- participants. All analyses including NAA and Glu alone were non-significant. Here, we provide preliminary evidence for the importance of NAA/Glu in ASD with SI, with implications for biomarker discovery. Further mechanistic research into risk and interventional approaches to address SI in ASD are needed.
Collapse
Affiliation(s)
- Iska Moxon-Emre
- Cundill Centre for Child and Youth Depression, The Margaret and Wallace McCain Centre for Child, Youth & Family Mental Health, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (I.M.-E.); (R.E.L.); (H.T.); (M.-C.L.); (P.S.)
| | - Paul E. Croarkin
- Department of Psychiatry and Psychology, Division of Child and Adolescent Psychiatry, Mayo Clinic, Rochester, MN 55905, USA;
| | - Zafiris J. Daskalakis
- Temerty Centre for Therapeutic Brain Intervention, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M6J 1H1, Canada; (Z.J.D.); (D.M.B.); (P.D.)
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Daniel M. Blumberger
- Temerty Centre for Therapeutic Brain Intervention, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M6J 1H1, Canada; (Z.J.D.); (D.M.B.); (P.D.)
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Rachael E. Lyon
- Cundill Centre for Child and Youth Depression, The Margaret and Wallace McCain Centre for Child, Youth & Family Mental Health, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (I.M.-E.); (R.E.L.); (H.T.); (M.-C.L.); (P.S.)
| | - Hideaki Tani
- Cundill Centre for Child and Youth Depression, The Margaret and Wallace McCain Centre for Child, Youth & Family Mental Health, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (I.M.-E.); (R.E.L.); (H.T.); (M.-C.L.); (P.S.)
| | - Peter Truong
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (P.T.); (N.S.)
| | - Meng-Chuan Lai
- Cundill Centre for Child and Youth Depression, The Margaret and Wallace McCain Centre for Child, Youth & Family Mental Health, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (I.M.-E.); (R.E.L.); (H.T.); (M.-C.L.); (P.S.)
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1R8, Canada
- Research Institute, Department of Psychiatry, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Psychology, University of Toronto, Toronto, ON M5S 3G3, Canada
| | - Pushpal Desarkar
- Temerty Centre for Therapeutic Brain Intervention, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M6J 1H1, Canada; (Z.J.D.); (D.M.B.); (P.D.)
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Napapon Sailasuta
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (P.T.); (N.S.)
| | - Peter Szatmari
- Cundill Centre for Child and Youth Depression, The Margaret and Wallace McCain Centre for Child, Youth & Family Mental Health, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (I.M.-E.); (R.E.L.); (H.T.); (M.-C.L.); (P.S.)
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1R8, Canada
- Research Institute, Department of Psychiatry, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Stephanie H. Ameis
- Cundill Centre for Child and Youth Depression, The Margaret and Wallace McCain Centre for Child, Youth & Family Mental Health, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (I.M.-E.); (R.E.L.); (H.T.); (M.-C.L.); (P.S.)
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1R8, Canada
- Research Institute, Department of Psychiatry, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
21
|
Bissonnette JN, Francis AM, MacNeil S, Crocker CE, Tibbo PG, Fisher DJ. Glutamate and N-Acetylaspartate Alterations Observed in Early Phase Psychosis: A Systematic Review of Proton Magnetic Resonance Spectroscopy Studies. Psychiatry Res Neuroimaging 2022; 321:111459. [PMID: 35183897 DOI: 10.1016/j.pscychresns.2022.111459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 11/27/2022]
Abstract
Glutamate and N-acetylaspartate have been investigated in the neuropathology of chronic schizophrenia, with fewer studies focusing on early phase psychosis. Additionally, there has been little review and synthesis of the literature focused on multiple brain regions. This systematic review aims to provide a clear report of the current state of research on glutamate and n-acetylaspartate concentrations in early phase psychosis (defined as the first five years following psychosis onset) in multiple brain regions. Existing literature was searched systematically to compile reports of glutamate/glutamate+glutamine (Glx) and n-acetylaspartate absolute levels and ratios in both male and female individuals with early phase psychosis. Reports on glutamate/Glx concentrations in the medial prefrontal region and thalamus were varied, but the majority of reports suggested no alterations in EPP. No studies reported glutamate alterations in the hippocampus or cerebellum. There was no evidence for n-acetylaspartate alterations in the caudate, basal ganglia, and medial prefrontal cortex, and minimal evidence for NAA reductions in the thalamus, anterior cingulate cortex, and hippocampus. Future research should focus on the regions that are less commonly reported, and should aim to explore possible confounds, such as medication status and substance use.
Collapse
Affiliation(s)
- J N Bissonnette
- Department of Psychiatry, Dalhousie University, Halifax, NS.
| | - A M Francis
- Department of Psychology, Saint Mary's University, Halifax, NS.
| | - S MacNeil
- Department of Psychology, Mount Saint Vincent University, Halifax, NS.
| | - C E Crocker
- Department of Psychiatry, Dalhousie University, Halifax, NS; Nova Scotia Early Psychosis Program, Halifax, NS; Department of Diagnostic Imaging, Dalhousie University, Halifax, NS.
| | - P G Tibbo
- Department of Psychiatry, Dalhousie University, Halifax, NS; Nova Scotia Early Psychosis Program, Halifax, NS.
| | - D J Fisher
- Department of Psychiatry, Dalhousie University, Halifax, NS; Department of Psychology, Saint Mary's University, Halifax, NS; Department of Psychology, Mount Saint Vincent University, Halifax, NS.
| |
Collapse
|
22
|
Lee MH, Hwang YH, Yun CS, Han BS, Kim DY. Altered small-world property of a dynamic metabolic network in murine left hippocampus after exposure to acute stress. Sci Rep 2022; 12:3885. [PMID: 35273207 PMCID: PMC8913833 DOI: 10.1038/s41598-022-07586-6] [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: 09/30/2021] [Accepted: 02/18/2022] [Indexed: 11/09/2022] Open
Abstract
The acute stress response is a natural and fundamental reaction that balances the physiological conditions of the brain. To maintain homeostasis in the brain, the response is based on changes over time in hormones and neurotransmitters, which are related to resilience and can adapt successfully to acute stress. This increases the need for dynamic analysis over time, and new approaches to examine the relationship between metabolites have emerged. This study investigates whether the constructed metabolic network is a realistic or a random network and is affected by acute stress. While the metabolic network in the control group met the criteria for small-worldness at all time points, the metabolic network in the stress group did not at some time points, and the small-worldness had resilience after the fifth time point. The backbone metabolic network only met the criteria for small-worldness in the control group. Additionally, creatine had lower local efficiency in the stress group than the control group, and for the backbone metabolic network, creatine and glutamate were lower and higher in the stress group than the control group, respectively. These findings provide evidence of metabolic imbalance that may be a pre-stage of alterations to brain structure due to acute stress.
Collapse
Affiliation(s)
- Min-Hee Lee
- Institute of Human Genomic Study, College of Medicine, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Yoon Ho Hwang
- Department of Biomedical Engineering, Yonsei University, Wonju, Republic of Korea
| | - Chang-Soo Yun
- Department of Radiation Convergence Engineering, Yonsei University, Wonju, Republic of Korea
| | - Bong Soo Han
- Department of Radiation Convergence Engineering, Yonsei University, Wonju, Republic of Korea
| | - Dong Youn Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, Republic of Korea.
| |
Collapse
|
23
|
Matthews DC, Mao X, Dowd K, Tsakanikas D, Jiang CS, Meuser C, Andrews RD, Lukic AS, Lee J, Hampilos N, Shafiian N, Sano M, David Mozley P, Fillit H, McEwen BS, Shungu DC, Pereira AC. Riluzole, a glutamate modulator, slows cerebral glucose metabolism decline in patients with Alzheimer's disease. Brain 2021; 144:3742-3755. [PMID: 34145880 PMCID: PMC8719848 DOI: 10.1093/brain/awab222] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/07/2021] [Accepted: 05/22/2021] [Indexed: 11/14/2022] Open
Abstract
Dysregulation of glutamatergic neural circuits has been implicated in a cycle of toxicity, believed among the neurobiological underpinning of Alzheimer's disease. Previously, we reported preclinical evidence that the glutamate modulator riluzole, which is FDA approved for the treatment of amyotrophic lateral sclerosis, has potential benefits on cognition, structural and molecular markers of ageing and Alzheimer's disease. The objective of this study was to evaluate in a pilot clinical trial, using neuroimaging biomarkers, the potential efficacy and safety of riluzole in patients with Alzheimer's disease as compared to placebo. A 6-month phase 2 double-blind, randomized, placebo-controlled study was conducted at two sites. Participants consisted of males and females, 50 to 95 years of age, with a clinical diagnosis of probable Alzheimer's disease, and Mini-Mental State Examination between 19 and 27. Ninety-four participants were screened, 50 participants who met inclusion criteria were randomly assigned to receive 50 mg riluzole (n = 26) or placebo (n = 24) twice a day. Twenty-two riluzole-treated and 20 placebo participants completed the study. Primary end points were baseline to 6 months changes in (i) cerebral glucose metabolism as measured with fluorodeoxyglucose-PET in prespecified regions of interest (hippocampus, posterior cingulate, precuneus, lateral temporal, inferior parietal, frontal); and (ii) changes in posterior cingulate levels of the neuronal viability marker N-acetylaspartate as measured with in vivo proton magnetic resonance spectroscopy. Secondary outcome measures were neuropsychological testing for correlation with neuroimaging biomarkers and in vivo measures of glutamate in posterior cingulate measured with magnetic resonance spectroscopy as a potential marker of target engagement. Measures of cerebral glucose metabolism, a well-established Alzheimer's disease biomarker and predictor of disease progression, declined significantly less in several prespecified regions of interest with the most robust effect in posterior cingulate, and effects in precuneus, lateral temporal, right hippocampus and frontal cortex in riluzole-treated participants in comparison to the placebo group. No group effect was found in measures of N-acetylaspartate levels. A positive correlation was observed between cognitive measures and regional cerebral glucose metabolism. A group × visit interaction was observed in glutamate levels in posterior cingulate, potentially suggesting engagement of glutamatergic system by riluzole. In vivo glutamate levels positively correlated with cognitive performance. These findings support our main primary hypothesis that cerebral glucose metabolism would be better preserved in the riluzole-treated group than in the placebo group and provide a rationale for more powered, longer duration studies of riluzole as a potential intervention for Alzheimer's disease.
Collapse
Affiliation(s)
| | - Xiangling Mao
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | | | | | | | - Caroline Meuser
- Department of Psychiatry, Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Ana S Lukic
- ADM Diagnostics Inc., Northbrook, IL 60062, USA
| | - Jihyun Lee
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Nicholas Hampilos
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Neeva Shafiian
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mary Sano
- Department of Psychiatry, Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - P David Mozley
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Howard Fillit
- Alzheimer's Drug Discovery Foundation, New York, NY 10019, USA
| | | | - Dikoma C Shungu
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ana C Pereira
- The Rockefeller University, New York, NY 10065, USA
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
24
|
Sonmez AI, Lewis CP, Port JD, Athreya AP, Choi DS, Zaccariello MJ, Shekunov J, Blacker CJ, Croarkin PE. A pilot spectroscopy study of adversity in adolescents. Biomark Neuropsychiatry 2021; 5:100043. [PMID: 35783196 PMCID: PMC9248870 DOI: 10.1016/j.bionps.2021.100043] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Background Childhood adversity is a global health problem affecting 25-50% of children worldwide. Few prior studies have examined the underlying neurochemistry of adversity in adolescents. This cross-sectional study examined spectroscopic markers of trauma in a cohort of adolescents with major depressive disorder (MDD) and healthy controls. We hypothesized that historical adversity would have a negative relationship with spectroscopic measures of glutamate metabolites in anterior cingulate cortex. Methods Adolescent participants (aged 13-21) underwent a semi-structured diagnostic interview and clinical assessment, which included the self-report Childhood Trauma Questionnaire (CTQ), a 28-item assessment of childhood adversity. Proton magnetic resonance spectroscopy (1H-MRS) scans at 3 Tesla of an anterior cingulate cortex (ACC) voxel (8 cm3) encompassing both hemispheres were collected using a 2-dimensional J-averaged sequence to assess N-acetylaspartate (NAA), Glx (glutamate+glutamine) and [NAA]/[Glx] concentrations. Generalized linear models assessed the relationships between CTQ scores and metabolite levels in ACC. Results Thirty-nine participants (17 healthy controls, 22 depressed participants) underwent 1H-MRS and completed the CTQ measures. There were decrements in [NAA]/[Glx] ratio in the ACC of participants with childhood adversity while no significant relationship between CTQ total score and any of the ACC metabolites was found in the combined sample. Exploratory results revealed a positive association between Glx levels and CTQ scores in depressed participants. Conversely the [NAA]/[Glx] ratio had a negative association with total CTQ scores in the depressed participants. Emotional Abuse Scale showed a significant negative relationship with [NAA]/[Glx] ratio in the combined sample when adjusted for depression severity. Conclusions Our findings suggest that childhood adversity may impact brain neurochemical profiles. Further longitudinal studies should examine neurochemical correlates of childhood adversity throughout development and in populations with other psychiatric disorders.
Collapse
Affiliation(s)
- A. Irem Sonmez
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Charles P. Lewis
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
| | - John D. Port
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Department of Radiology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Arjun P. Athreya
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN 55905, USA
| | - Doo-Sop Choi
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN 55905, USA
| | - Michael J. Zaccariello
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Julia Shekunov
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Caren J. Blacker
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Paul E. Croarkin
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| |
Collapse
|
25
|
Foret JT, Dekhtyar M, Cole JH, Gourley DD, Caillaud M, Tanaka H, Haley AP. Network Modeling Sex Differences in Brain Integrity and Metabolic Health. Front Aging Neurosci 2021; 13:691691. [PMID: 34267647 PMCID: PMC8275835 DOI: 10.3389/fnagi.2021.691691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/27/2021] [Indexed: 01/14/2023] Open
Abstract
Hypothesis-driven studies have demonstrated that sex moderates many of the relationships between brain health and cardiometabolic disease, which impacts risk for later-life cognitive decline. In the present study, we sought to further our understanding of the associations between multiple markers of brain integrity and cardiovascular risk in a midlife sample of 266 individuals by using network analysis, a technique specifically designed to examine complex associations among multiple systems at once. Separate network models were constructed for male and female participants to investigate sex differences in the biomarkers of interest, selected based on evidence linking them with risk for late-life cognitive decline: all components of metabolic syndrome (obesity, hypertension, dyslipidemia, and hyperglycemia); neuroimaging-derived brain-predicted age minus chronological age; ratio of white matter hyperintensities to whole brain volume; seed-based resting state functional connectivity in the Default Mode Network, and ratios of N-acetyl aspartate, glutamate and myo-inositol to creatine, measured through proton magnetic resonance spectroscopy. Males had a sparse network (87.2% edges = 0) relative to females (69.2% edges = 0), indicating fewer relationships between measures of cardiometabolic risk and brain integrity. The edges in the female network provide meaningful information about potential mechanisms between brain integrity and cardiometabolic health. Additionally, Apolipoprotein ϵ4 (ApoE ϵ4) status and waist circumference emerged as central nodes in the female model. Our study demonstrates that network analysis is a promising technique for examining relationships between risk factors for cognitive decline in a midlife population and that investigating sex differences may help optimize risk prediction and tailor individualized treatments in the future.
Collapse
Affiliation(s)
- Janelle T. Foret
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
| | - Maria Dekhtyar
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
| | - James H. Cole
- Department of Computer Science, Centre for Medical Image Computing, University College London, London, United Kingdom
- Dementia Research Centre, Institute of Neurology, University College London, London, United Kingdom
| | - Drew D. Gourley
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX, United States
| | - Marie Caillaud
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
| | - Hirofumi Tanaka
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX, United States
| | - Andreana P. Haley
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
- Biomedical Imaging Center, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
26
|
Oxidative Dysregulation in Early Life Stress and Posttraumatic Stress Disorder: A Comprehensive Review. Brain Sci 2021; 11:brainsci11060723. [PMID: 34072322 PMCID: PMC8228973 DOI: 10.3390/brainsci11060723] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 12/30/2022] Open
Abstract
Traumatic stress may chronically affect master homeostatic systems at the crossroads of peripheral and central susceptibility pathways and lead to the biological embedment of trauma-related allostatic trajectories through neurobiological alterations even decades later. Lately, there has been an exponential knowledge growth concerning the effect of traumatic stress on oxidative components and redox-state homeostasis. This extensive review encompasses a detailed description of the oxidative cascade components along with their physiological and pathophysiological functions and a systematic presentation of both preclinical and clinical, genetic and epigenetic human findings on trauma-related oxidative stress (OXS), followed by a substantial synthesis of the involved oxidative cascades into specific and functional, trauma-related pathways. The bulk of the evidence suggests an imbalance of pro-/anti-oxidative mechanisms under conditions of traumatic stress, respectively leading to a systemic oxidative dysregulation accompanied by toxic oxidation byproducts. Yet, there is substantial heterogeneity in findings probably relative to confounding, trauma-related parameters, as well as to the equivocal directionality of not only the involved oxidative mechanisms but other homeostatic ones. Accordingly, we also discuss the trauma-related OXS findings within the broader spectrum of systemic interactions with other major influencing systems, such as inflammation, the hypothalamic-pituitary-adrenal axis, and the circadian system. We intend to demonstrate the inherent complexity of all the systems involved, but also put forth associated caveats in the implementation and interpretation of OXS findings in trauma-related research and promote their comprehension within a broader context.
Collapse
|
27
|
Bartnik-Olson BL, Alger JR, Babikian T, Harris AD, Holshouser B, Kirov II, Maudsley AA, Thompson PM, Dennis EL, Tate DF, Wilde EA, Lin A. The clinical utility of proton magnetic resonance spectroscopy in traumatic brain injury: recommendations from the ENIGMA MRS working group. Brain Imaging Behav 2021; 15:504-525. [PMID: 32797399 PMCID: PMC7882010 DOI: 10.1007/s11682-020-00330-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Proton (1H) magnetic resonance spectroscopy provides a non-invasive and quantitative measure of brain metabolites. Traumatic brain injury impacts cerebral metabolism and a number of research groups have successfully used this technique as a biomarker of injury and/or outcome in both pediatric and adult TBI populations. However, this technique is underutilized, with studies being performed primarily at centers with access to MR research support. In this paper we present a technical introduction to the acquisition and analysis of in vivo 1H magnetic resonance spectroscopy and review 1H magnetic resonance spectroscopy findings in different injury populations. In addition, we propose a basic 1H magnetic resonance spectroscopy data acquisition scheme (Supplemental Information) that can be added to any imaging protocol, regardless of clinical magnetic resonance platform. We outline a number of considerations for study design as a way of encouraging the use of 1H magnetic resonance spectroscopy in the study of traumatic brain injury, as well as recommendations to improve data harmonization across groups already using this technique.
Collapse
Affiliation(s)
| | - Jeffry R Alger
- Departments of Neurology and Radiology, University of California Los Angeles, Los Angeles, CA, USA
- NeuroSpectroScopics LLC, Sherman Oaks, Los Angeles, CA, USA
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Talin Babikian
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA
- UCLA Steve Tisch BrainSPORT Program, Los Angeles, CA, USA
| | - Ashley D Harris
- Department of Radiology, University of Calgary, Calgary, Canada
- Child and Adolescent Imaging Research Program, Alberta Children's Hospital Research Institute and the Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Barbara Holshouser
- Department of Radiology, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - Ivan I Kirov
- Bernard and Irene Schwartz Center for Biomedical Imaging, Center for Advanced Imaging Innovation and Research (CAI2R), Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - Andrew A Maudsley
- Department of Radiology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Paul M Thompson
- Imaging Genetics Center, Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of USC, Marina del Rey, Los Angeles, CA, USA
- Departments of Neurology, Pediatrics, Psychiatry, Radiology, Engineering, and Ophthalmology, USC, Los Angeles, CA, USA
| | - Emily L Dennis
- Imaging Genetics Center, Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of USC, Marina del Rey, Los Angeles, CA, USA
- Department of Neurology, University of Utah, Salt Lake City, UT, USA
- Psychiatry Neuroimaging Laboratory, Brigham & Women's Hospital, Boston, MA, USA
| | - David F Tate
- Department of Neurology, University of Utah, Salt Lake City, UT, USA
- George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Elisabeth A Wilde
- Department of Neurology, University of Utah, Salt Lake City, UT, USA
- George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT, USA
- H. Ben Taub Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | - Alexander Lin
- Center for Clinical Spectroscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
28
|
Martens L, Herrmann L, Colic L, Li M, Richter A, Behnisch G, Stork O, Seidenbecher C, Schott BH, Walter M. Met carriers of the BDNF Val66Met polymorphism show reduced Glx/NAA in the pregenual ACC in two independent cohorts. Sci Rep 2021; 11:6742. [PMID: 33762638 PMCID: PMC7990923 DOI: 10.1038/s41598-021-86220-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 03/02/2021] [Indexed: 12/17/2022] Open
Abstract
The Met allele of the Val66Met SNP of the BDNF gene (rs6265) is associated with impaired activity-dependent release of brain-derived neurotrophic factor (BDNF), resulting in reduced synaptic plasticity, impaired glutamatergic neurotransmission, and morphological changes. While previous work has demonstrated Val66Met effects on magnetic resonance spectroscopy (MRS) markers of either glutamatergic metabolism (Glx) or neuronal integrity (NAA), no study has investigated Val66Met effects on these related processes simultaneously. As these metabolites share a metabolic pathway, the Glx/NAA ratio may be a more sensitive marker of changes associated with the Val66Met SNP. This ratio is increased in psychiatric disorders linked to decreased functioning in the anterior cingulate cortex (ACC). In this study, we investigated the correlation of the Val66Met polymorphism of the BDNF gene with Glx/NAA in the pregenual anterior cingulate cortex (pgACC) using MRS at 3 Tesla (T) (n = 30, all males) and 7 T (n = 98, 40 females). In both cohorts, Met carriers had lower Glx/NAA compared to Val homozygotes. Follow-up analyses using absolute quantification revealed that the Met carriers do not show decreased pgACC glutamate or glutamine levels, but instead show increased NAA compared to the Val homozygotes. This finding may in part explain conflicting evidence for Val66Met as a risk factor for developing psychiatric illnesses.
Collapse
Affiliation(s)
- Louise Martens
- University Department of Psychiatry and Psychotherapy, Tübingen, Germany.,Graduate Training Center, IMPRS, Tübingen, Germany.,Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Luisa Herrmann
- University Department of Psychiatry and Psychotherapy, Tübingen, Germany.,Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - Lejla Colic
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany.,Clinical Affective Neuroscience Laboratory, Magdeburg, Germany.,Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Meng Li
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany.,Clinical Affective Neuroscience Laboratory, Magdeburg, Germany
| | - Anni Richter
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | | | - Oliver Stork
- Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-Von-Guericke-University, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Constanze Seidenbecher
- Leibniz Institute for Neurobiology, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Björn H Schott
- Leibniz Institute for Neurobiology, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany.,Department of Psychiatry and Psychotherapy, University Medicine Göttingen, Göttingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Martin Walter
- University Department of Psychiatry and Psychotherapy, Tübingen, Germany. .,Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany. .,Clinical Affective Neuroscience Laboratory, Magdeburg, Germany.
| |
Collapse
|
29
|
Metabolomics Insights of the Immunomodulatory Activities of Phlorizin and Phloretin on Human THP-1 Macrophages. Molecules 2021; 26:molecules26040787. [PMID: 33546377 PMCID: PMC7913540 DOI: 10.3390/molecules26040787] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
Dihydrochalcones, phlorizin (PZ) and its aglycone phloretin (PT), have evidenced immunomodulatory effects through several mechanisms. However, the differential metabolic signatures that lead to these properties are largely unknown. Since macrophages play an important role in the immune response, our study aimed to characterise human THP-1 macrophages under PZ and PT exposure. A multiplatform-based untargeted metabolomics approach was used to reveal metabolites associated with the anti-inflammatory mechanisms triggered by the dihydrochalcones in LPS-stimulated macrophages, for the first time. Results showed differential phenotypic response in macrophages for all treatments. Dihydrochalcone treatment in LPS-stimulated macrophages mimics the response under normal conditions, suggesting inhibition of LPS response. Antagonistic effects of dihydrochalcones against LPS was mainly observed in glycerophospholipid and sphingolipid metabolism besides promoting amino acid biosynthesis. Moreover, PT showed greater metabolic activity than PZ. Overall, the findings of this study yielded knowledge about the mechanisms of action PZ and PT at metabolic level in modulating inflammatory response in human cells.
Collapse
|
30
|
Hsu CH, Lin S, Ho AC, Johnson TD, Wang PC, Scafidi J, Tu TW. Comparison of in vivo and in situ detection of hippocampal metabolites in mouse brain using 1 H-MRS. NMR IN BIOMEDICINE 2021; 34:e4451. [PMID: 33258202 PMCID: PMC8214416 DOI: 10.1002/nbm.4451] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 10/04/2020] [Accepted: 11/06/2020] [Indexed: 05/25/2023]
Abstract
The study of cerebral metabolites relies heavily on detection methods and sample preparation. Animal experiments in vivo require anesthetic agents that can alter brain metabolism, whereas ex vivo experiments demand appropriate fixation methods to preserve the tissue from rapid postmortem degradation. In this study, the metabolic profiles of mouse hippocampi using proton magnetic resonance spectroscopy (1 H-MRS) were compared in vivo and in situ with or without focused beam microwave irradiation (FBMI) fixation. Ten major brain metabolites, including lactate (Lac), N-acetylaspartate (NAA), total choline (tCho), myo-inositol (mIns), glutamine (Gln), glutamate (Glu), aminobutyric acid (GABA), glutathione (GSH), total creatine (tCr) and taurine (Tau), were analyzed using LCModel. After FBMI fixation, the concentrations of Lac, tCho and mIns were comparable with those obtained in vivo under isoflurane, whereas other metabolites were significantly lower. Except for a decrease in NAA and an increase in Tau, all the other metabolites remained stable over 41 hours in FBMI-fixed brains. Without FBMI, the concentrations of mIns (before 2 hours), tCho and GABA were close to those measured in vivo. However, higher Lac (P < .01) and lower NAA, Gln, Glu, GSH, tCr and Tau were observed (P < .01). NAA, Gln, Glu, GSH, tCr and Tau exhibited good temporal stability for at least 20 hours in the unfixed brain, whereas a linear increase of tCho, mIns and GABA was observed. Possible mechanisms of postmortem degradation are discussed. Our results indicate that a proper fixation method is required for in situ detection depending on the targeted metabolites of specific interests in the brain.
Collapse
Affiliation(s)
- Chao-Hsiung Hsu
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
| | - Stephen Lin
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
| | - Ai-Chen Ho
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - T. Derek Johnson
- Center for Neuroscience Research, Department of Neurology, Children’s National Hospital, Washington, DC, USA
| | - Paul C. Wang
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
- Department of Electrical Engineering, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Joseph Scafidi
- Center for Neuroscience Research, Department of Neurology, Children’s National Hospital, Washington, DC, USA
| | - Tsang-Wei Tu
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
| |
Collapse
|
31
|
Post-inflammatory behavioural despair in male mice is associated with reduced cortical glutamate-glutamine ratios, and circulating lipid and energy metabolites. Sci Rep 2020; 10:16857. [PMID: 33033375 PMCID: PMC7545201 DOI: 10.1038/s41598-020-74008-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022] Open
Abstract
Post-inflammatory behaviours in rodents are widely used to model human depression and to test the efficacy of novel anti-depressants. Mice injected with lipopolysaccharide (LPS) display a depressive-like phenotype twenty-four hours after endotoxin administration. Despite the widespread use of this model, the mechanisms that underlie the persistent behavioural changes after the transient peripheral inflammatory response remain elusive. The study of the metabolome, the collection of all the small molecule metabolites in a sample, combined with multivariate statistical techniques provides a way of studying biochemical pathways influenced by an LPS challenge. Adult male CD-1 mice received an intraperitoneal injection of either LPS (0.83 mg/kg) or saline, and were assessed for depressive-like behaviour 24 h later. In a separate mouse cohort, pro-inflammatory cytokine gene expression and 1H nuclear magnetic resonance (NMR) metabolomics measurements were made in brain tissue and blood. Statistical analyses included Independent Sample t-tests for gene expression data, and supervised multi-variate analysis using orthogonal partial least squares discriminant analysis for metabolomics. Both plasma and brain metabolites in male mice were altered following a single peripheral LPS challenge that led to depressive-like behaviour in the forced swim test. The plasma metabolites altered by LPS are involved in energy metabolism, including lipoproteins, glucose, creatine, and isoleucine. In the brain, glutamate, serine, and N-acetylaspartate (NAA) were reduced after LPS, whereas glutamine was increased. Serine-modulated glutamatergic signalling and changes in bioenergetics may mediate the behavioural phenotype induced by LPS. In light of other data supporting a central imbalance of glutamate-glutamine cycling in depression, our results suggest that aberrant central glutaminergic signalling may underpin the depressive-like behaviours that result from both inflammation and non-immune pathophysiology. Normalising glutaminergic signalling, rather than seeking to increase serotonergic signalling, might prove to be a more coherent approach to the development of new treatments for mood disorder.
Collapse
|
32
|
Take Advantage of Glutamine Anaplerosis, the Kernel of the Metabolic Rewiring in Malignant Gliomas. Biomolecules 2020; 10:biom10101370. [PMID: 32993063 PMCID: PMC7599606 DOI: 10.3390/biom10101370] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/18/2020] [Accepted: 09/24/2020] [Indexed: 12/11/2022] Open
Abstract
Glutamine is a non-essential amino acid that plays a key role in the metabolism of proliferating cells including neoplastic cells. In the central nervous system (CNS), glutamine metabolism is particularly relevant, because the glutamine-glutamate cycle is a way of controlling the production of glutamate-derived neurotransmitters by tightly regulating the bioavailability of the amino acids in a neuron-astrocyte metabolic symbiosis-dependent manner. Glutamine-related metabolic adjustments have been reported in several CNS malignancies including malignant gliomas that are considered ‘glutamine addicted’. In these tumors, glutamine becomes an essential amino acid preferentially used in energy and biomass production including glutathione (GSH) generation, which is crucial in oxidative stress control. Therefore, in this review, we will highlight the metabolic remodeling that gliomas undergo, focusing on glutamine metabolism. We will address some therapeutic regimens including novel research attempts to target glutamine metabolism and a brief update of diagnosis strategies that take advantage of this altered profile. A better understanding of malignant glioma cell metabolism will help in the identification of new molecular targets and the design of new therapies.
Collapse
|
33
|
Hnilicová P, Štrbák O, Kolisek M, Kurča E, Zeleňák K, Sivák Š, Kantorová E. Current Methods of Magnetic Resonance for Noninvasive Assessment of Molecular Aspects of Pathoetiology in Multiple Sclerosis. Int J Mol Sci 2020; 21:E6117. [PMID: 32854318 PMCID: PMC7504207 DOI: 10.3390/ijms21176117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/13/2020] [Accepted: 08/21/2020] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease with expanding axonal and neuronal degeneration in the central nervous system leading to motoric dysfunctions, psychical disability, and cognitive impairment during MS progression. The exact cascade of pathological processes (inflammation, demyelination, excitotoxicity, diffuse neuro-axonal degeneration, oxidative and metabolic stress, etc.) causing MS onset is still not fully understood, although several accompanying biomarkers are particularly suitable for the detection of early subclinical changes. Magnetic resonance (MR) methods are generally considered to be the most sensitive diagnostic tools. Their advantages include their noninvasive nature and their ability to image tissue in vivo. In particular, MR spectroscopy (proton 1H and phosphorus 31P MRS) is a powerful analytical tool for the detection and analysis of biomedically relevant metabolites, amino acids, and bioelements, and thus for providing information about neuro-axonal degradation, demyelination, reactive gliosis, mitochondrial and neurotransmitter failure, cellular energetic and membrane alternation, and the imbalance of magnesium homeostasis in specific tissues. Furthermore, the MR relaxometry-based detection of accumulated biogenic iron in the brain tissue is useful in disease evaluation. The early description and understanding of the developing pathological process might be critical for establishing clinically effective MS-modifying therapies.
Collapse
Affiliation(s)
- Petra Hnilicová
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (O.Š.); (M.K.)
| | - Oliver Štrbák
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (O.Š.); (M.K.)
| | - Martin Kolisek
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (O.Š.); (M.K.)
| | - Egon Kurča
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (E.K.); (Š.S.); (E.K.)
| | - Kamil Zeleňák
- Clinic of Radiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia;
| | - Štefan Sivák
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (E.K.); (Š.S.); (E.K.)
| | - Ema Kantorová
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (E.K.); (Š.S.); (E.K.)
| |
Collapse
|
34
|
Lewis CP, Port JD, Blacker CJ, Sonmez AI, Seewoo BJ, Leffler JM, Frye MA, Croarkin PE. Altered anterior cingulate glutamatergic metabolism in depressed adolescents with current suicidal ideation. Transl Psychiatry 2020; 10:119. [PMID: 32327639 PMCID: PMC7181616 DOI: 10.1038/s41398-020-0792-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 02/21/2020] [Accepted: 03/25/2020] [Indexed: 01/09/2023] Open
Abstract
The anterior cingulate cortex (ACC) is involved in emotion regulation and salience processing. Prior research has implicated ACC dysfunction in suicidal ideation (SI) and suicidal behavior. This study aimed to quantify ACC glutamatergic concentrations and to examine relationships with SI in a sample of healthy and depressed adolescents. Forty adolescents underwent clinical evaluation and proton magnetic resonance spectroscopy (1H-MRS) at 3 T, utilizing a 2-dimensional J-averaged PRESS sequence sampling a medial pregenual ACC voxel. Cerebrospinal fluid-corrected ACC metabolite concentrations were compared between healthy control (HC, n = 16), depressed without SI (Dep/SI-, n = 13), and depressed with SI (Dep/SI+, n = 11) youth using general linear models covarying for age, sex, and psychotropic medication use. Relationships between ACC metabolites and continuous measures of SI were examined using multiple linear regressions. ROC analysis was used to determine the ability of glutamate+glutamine (Glx) and the N-acetylaspartate (NAA)/Glx ratio to discriminate Dep/SI- and Dep/SI+ adolescents. Dep/SI+ adolescents had higher Glx than Dep/SI- participants (padj = 0.012) and had lower NAA/Glx than both Dep/SI- (padj = 0.002) and HC adolescents (padj = 0.039). There were significant relationships between SI intensity and Glx (pFDR = 0.026), SI severity and NAA/Glx (pFDR = 0.012), and SI intensity and NAA/Glx (pFDR = 0.004). ACC Glx and NAA/Glx discriminated Dep/SI- from Dep/SI+ participants. Uncoupled NAA-glutamatergic metabolism in the ACC may play a role in suicidal ideation and behavior. Longitudinal studies are needed to establish whether aberrant glutamatergic metabolism corresponds to acute or chronic suicide risk. Glutamatergic biomarkers may be promising targets for novel risk assessment and interventional strategies for suicidal ideation and behavior.
Collapse
Affiliation(s)
- Charles P Lewis
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
| | - John D Port
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Caren J Blacker
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - A Irem Sonmez
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Bhedita J Seewoo
- Experimental and Regenerative Neurosciences, School of Biological Sciences, University of Western Australia, Perth, WA, Australia
- Brain Plasticity Group, Perron Institute for Neurological and Translational Research, Perth, WA, Australia
- Centre for Microscopy, Characterisation and Analysis, Research Infrastructure Centres, University of Western Australia, Perth, WA, Australia
| | - Jarrod M Leffler
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Mark A Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Paul E Croarkin
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
35
|
Cecil KM, Naidu P. Advances in Pediatric Neuroimaging. MR Spectroscopy. Semin Pediatr Neurol 2020; 33:100798. [PMID: 32331612 DOI: 10.1016/j.spen.2020.100798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The basic principles of proton magnetic resonance spectroscopy are presented in this work to briefly familiarize the clinician and to distinguish spectroscopy from magnetic resonance imaging. For those knowledgeable about proton magnetic resonance spectroscopy, this article will also provide the reader an update on recent technical and translational developments relevant to pediatric neurologic conditions. These developments were selected for their potential impact towards the clinical care of patients in pediatric-based practices. At this point in time, these new spectroscopic approaches are currently applied to established populations with known diseases. This information will inform our knowledge about diseases and guide therapeutic options for the future.
Collapse
Affiliation(s)
- Kim M Cecil
- Professor of Radiology, Pediatrics, Neuroscience and Environmental Health, Imaging Research Center, Cincinnati Children's Hospital Medical Center, Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, OH.
| | - Padmaja Naidu
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
36
|
Warepam M, Ahmad K, Rahman S, Rahaman H, Kumari K, Singh LR. N-Acetylaspartate Is an Important Brain Osmolyte. Biomolecules 2020; 10:biom10020286. [PMID: 32059525 PMCID: PMC7072545 DOI: 10.3390/biom10020286] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/29/2020] [Accepted: 02/08/2020] [Indexed: 01/11/2023] Open
Abstract
Most of the human diseases related to various proteopathies are confined to the brain, which leads to the development of various forms of neurological disorders. The human brain consists of several osmolytic compounds, such as N-Acetylaspartate (NAA), myo-inositol (mI), glutamate (Glu), glutamine (Gln), creatine (Cr), and choline-containing compounds (Cho). Among these osmolytes, the level of NAA drastically decreases under neurological conditions, and, hence, NAA is considered to be one of the most widely accepted neuronal biomarkers in several human brain disorders. To date, no data are available regarding the effect of NAA on protein stability, and, therefore, the possible effect of NAA under proteopathic conditions has not been fully uncovered. To gain an insight into the effect of NAA on protein stability, thermal denaturation and structural measurements were carried out using two model proteins at different pH values. The results indicate that NAA increases the protein stability with an enhancement of structure formation. We also observed that the stabilizing ability of NAA decreases in a pH-dependent manner. Our study indicates that NAA is an efficient protein stabilizer at a physiological pH.
Collapse
Affiliation(s)
- Marina Warepam
- Department of Biotechnology, Manipur University, Manipur 795003, India; (M.W.); (H.R.)
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea;
| | - Safikur Rahman
- Department of Botany, Munshi Singh College, BR Ambedkar Bihar University, Muzaffarpur, Bihar 845401, India;
| | - Hamidur Rahaman
- Department of Biotechnology, Manipur University, Manipur 795003, India; (M.W.); (H.R.)
| | - Kritika Kumari
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi-110007, India;
| | - Laishram Rajendrakumar Singh
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi-110007, India;
- Correspondence: ; Tel.: +91-9811630757; Fax: +91-11-27666248
| |
Collapse
|
37
|
Shen J, Shenkar D, An L, Tomar JS. Local and Interregional Neurochemical Associations Measured by Magnetic Resonance Spectroscopy for Studying Brain Functions and Psychiatric Disorders. Front Psychiatry 2020; 11:802. [PMID: 32848957 PMCID: PMC7432119 DOI: 10.3389/fpsyt.2020.00802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Magnetic resonance spectroscopy (MRS) studies have found significant correlations among neurometabolites (e.g., between glutamate and GABA) across individual subjects and altered correlations in neuropsychiatric disorders. In this article, we discuss neurochemical associations among several major neurometabolites which underpin these observations by MRS. We also illustrate the role of spectral editing in eliminating unwanted correlations caused by spectral overlapping. Finally, we describe the prospects of mapping macroscopic neurochemical associations across the brain and characterizing excitation-inhibition balance of neural networks using glutamate- and GABA-editing MRS imaging.
Collapse
Affiliation(s)
- Jun Shen
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, United States
| | - Dina Shenkar
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, United States
| | - Li An
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, United States
| | - Jyoti Singh Tomar
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, United States
| |
Collapse
|
38
|
Naylor B, Hesam-Shariati N, McAuley JH, Boag S, Newton-John T, Rae CD, Gustin SM. Reduced Glutamate in the Medial Prefrontal Cortex Is Associated With Emotional and Cognitive Dysregulation in People With Chronic Pain. Front Neurol 2019; 10:1110. [PMID: 31849800 PMCID: PMC6903775 DOI: 10.3389/fneur.2019.01110] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 10/03/2019] [Indexed: 01/03/2023] Open
Abstract
A decrease in glutamate in the medial prefrontal cortex (mPFC) has been extensively found in animal models of chronic pain. Given that the mPFC is implicated in emotional appraisal, cognition and extinction of fear, could a potential decrease in glutamate be associated with increased pessimistic thinking, fear and worry symptoms commonly found in people with chronic pain? To clarify this question, 19 chronic pain subjects and 19 age- and gender-matched control subjects without pain underwent magnetic resonance spectroscopy. Both groups also completed the Temperament and Character, the Beck Depression and the State Anxiety Inventories to measure levels of harm avoidance, depression, and anxiety, respectively. People with chronic pain had significantly higher scores in harm avoidance, depression and anxiety compared to control subjects without pain. High levels of harm avoidance are characterized by excessive worry, pessimism, fear, doubt and fatigue. Individuals with chronic pain showed a significant decrease in mPFC glutamate levels compared to control subjects without pain. In people with chronic pain mPFC glutamate levels were significantly negatively correlated with harm avoidance scores. This means that the lower the concentration of glutamate in the mPFC, the greater the total scores of harm avoidance. High scores are associated with fearfulness, pessimism, and fatigue-proneness. We suggest that chronic pain, particularly the stress-induced release of glucocorticoids, induces changes in glutamate transmission in the mPFC, thereby influencing cognitive, and emotional processing. Thus, in people with chronic pain, regulation of fear, worry, negative thinking and fatigue is impaired.
Collapse
Affiliation(s)
- Brooke Naylor
- Neuroscience Research Australia, Sydney, NSW, Australia.,School of Psychology, Macquarie University, Sydney, NSW, Australia
| | | | - James H McAuley
- Neuroscience Research Australia, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Simon Boag
- School of Psychology, Macquarie University, Sydney, NSW, Australia
| | - Toby Newton-John
- Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
| | | | - Sylvia M Gustin
- Neuroscience Research Australia, Sydney, NSW, Australia.,School of Psychology, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
39
|
Virel A, Dudka I, Laterveer R, Af Bjerkén S. 1H NMR profiling of the 6-OHDA parkinsonian rat brain reveals metabolic alterations and signs of recovery after N-acetylcysteine treatment. Mol Cell Neurosci 2019; 98:131-139. [PMID: 31200101 DOI: 10.1016/j.mcn.2019.06.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/07/2019] [Accepted: 06/11/2019] [Indexed: 11/30/2022] Open
Abstract
Parkinson's disease is the second most common neurodegenerative disease caused by degeneration of dopamine neurons in the substantia nigra. The origin and causes of dopamine neurodegeneration in Parkinson's disease are not well understood but oxidative stress may play an important role in its onset. Much effort has been dedicated to find biomarkers indicative of oxidative stress and neurodegenerative processes in parkinsonian brains. By using proton nuclear magnetic resonance (1H NMR) to identify and quantify key metabolites, it is now possible to elucidate the metabolic pathways affected by pathological conditions like neurodegeneration. The metabolic disturbances in the 6-hydroxydopamine (6-OHDA) hemiparkinsonian rat model were monitored and the nature and size of these metabolic alterations were analyzed. The results indicate that a unilateral injection of 6-OHDA into the striatum causes metabolic changes that not only affect the injected hemisphere but also the contralateral, non-lesioned side. We could clearly identify specific metabolic pathways that were affected, which were mostly related with oxidative stress and neurotransmission. In addition, a partial metabolic recovery by carrying out an antioxidant treatment with N-acetylcysteine (NAC) was observable.
Collapse
Affiliation(s)
- Ana Virel
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden.
| | - Ilona Dudka
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Rutger Laterveer
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Sara Af Bjerkén
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden; Department of Clinical Sciences, Neuroscience, Umeå University, Umeå, Sweden
| |
Collapse
|
40
|
Kim YH, Shim HS, Kim KH, Lee J, Chung BC, Kowall NW, Ryu H, Lee J. Metabolomic Analysis Identifies Alterations of Amino Acid Metabolome Signatures in the Postmortem Brain of Alzheimer's Disease. Exp Neurobiol 2019; 28:376-389. [PMID: 31308797 PMCID: PMC6614073 DOI: 10.5607/en.2019.28.3.376] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 05/19/2019] [Accepted: 05/21/2019] [Indexed: 02/06/2023] Open
Abstract
Despite significant advances in neuroscience research over the past several decades, the exact cause of AD has not yet fully understood. The metabolic hypothesis as well as the amyloid and tau hypotheses have been proposed to be associated with AD pathogenesis. In order to identify metabolome signatures from the postmortem brains of sporadic AD patients and control subjects, we performed ultra performance liquid chromatography coupled with linear ion trap-Orbitrap mass spectrometer (UPLC-LTQ-Orbitrap-MS). Not only our study identified new metabolome signatures but also verified previously known metabolome profiles in the brain. Statistical modeling of the analytical data and validation of the structural assignments discovered metabolic biomarkers associated with the AD pathogenesis. Interestingly, hypotaurin, myo-inositol and oxo-proline levels were markedly elevated in AD while lutamate and N-acetyl-aspartate were decreased in the postmortem brain tissue of AD patients. In addition, neurosteroid level such as cortisol was significantly increased in AD. Together, our data indicate that impaired amino acid metabolism is associated with AD pathogenesis and the altered amino acid signatures can be useful diagnostic biomarkers of AD. Thus, modulation of amino acid metabolism may be a possible therapeutic approach to treat AD.
Collapse
Affiliation(s)
- Yoon Hwan Kim
- MolecularRecognition Research Center, Korea Institute of Science and Technology, Seoul 02792, Korea
- Department of Biotechnology, Graduate School, Korea University, Seoul 02792, Korea
| | - Hyun Soo Shim
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul 02792, Korea
| | - Junghee Lee
- Veteran's Affairs Boston Healthcare System, Boston, MA 02130, USA
- Boston University Alzheimer's Disease Center and Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Bong Chul Chung
- MolecularRecognition Research Center, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Neil W. Kowall
- Veteran's Affairs Boston Healthcare System, Boston, MA 02130, USA
- Boston University Alzheimer's Disease Center and Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Hoon Ryu
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
- Veteran's Affairs Boston Healthcare System, Boston, MA 02130, USA
- Boston University Alzheimer's Disease Center and Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Jeongae Lee
- MolecularRecognition Research Center, Korea Institute of Science and Technology, Seoul 02792, Korea
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, Korea
| |
Collapse
|
41
|
Sivaraman S, Kraguljac NV, White DM, Morgan CJ, Gonzales SS, Lahti AC. Neurometabolic abnormalities in the associative striatum in antipsychotic-naïve first episode psychosis patients. Psychiatry Res Neuroimaging 2018; 281:101-106. [PMID: 30286325 PMCID: PMC7874514 DOI: 10.1016/j.pscychresns.2018.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 06/06/2018] [Accepted: 06/06/2018] [Indexed: 12/23/2022]
Abstract
Schizophrenia is a chronic, often progressive, disorder. Understanding the underlying neurobiology present in the early stages of the illness is as a pivotal step in designing targeted interventions aimed at arresting disease progression. The aim of our study was to examine neurometabolic changes in the dopamine rich associative striatum in medication-naïve first episode psychosis (FEP). We quantified neurometabolites in 14 FEP and 18 healthy controls (HC) matched on key demographic characteristics. Spectra from the voxel in the left associative striatum were acquired using a PRESS sequence (TR/TE = 2000/80 ms; 512 averages). MRS data were quantified in the time domain with AMARES in jMRUI. Choline was significantly elevated in FEP compared to HC. No significant alterations in other metabolites were observed. We did not observe correlations between metabolite levels and clinical characteristics in FEP. Here, we demonstrated elevated choline and a disruption of the relationship between N-acetyl-aspartate and Glx (glutamate + glutamine) in medication-naïve FEP patients in the left striatum indicating possible mitochondrial, membrane and glial dysfunction as an underlying pathological phenomenon. In addition, striatal choline shows promise as a biomarker for FEP that may have utility in clinical trials investigating target engagement in experimental regimens.
Collapse
Affiliation(s)
- Soumya Sivaraman
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nina V Kraguljac
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David M White
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Charity J Morgan
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sara S Gonzales
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Adrienne C Lahti
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
42
|
Increased glutamate and deep brain atrophy can predict the severity of multiple sclerosis. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2018; 163:45-53. [PMID: 30150790 DOI: 10.5507/bp.2018.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 06/19/2018] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE In multiple sclerosis (MS), deep grey matter (DGM) atrophy has been recognised as a crucial component of the disease that presents early and it has been associated with disability. Although the precise mechanism underlying grey matter atrophy is unknown, several hypotheses have been postulated. Our previous research pointed to correlations of hypothalamic metabolic alterations with clinical outcomes of MS, therefore we decided to further test the relationship of these alterations with DGM atrophy. METHODS We used 1H-Magnetic Resonance spectroscopy (1H-MRS) of the hypothalamus to test its metabolites in 26 patients with RRMS and 22 healthy age-matched controls. DGM atrophy was evaluated by simple planimetry of third ventricular width on the hypothalamic level (3VW) in T1 weighted MRI pictures. Metabolite ratios of N-acetyl aspartate (NAA), choline (Cho), glutamate and glutamine (Glx), myo-inositol (mIns) and creatine (Cr) were correlated with Multiple Sclerosis Severity Scale (MSSS) and 3VW. RESULTS Metabolite concentrations were compared between patients and controls using multiple regression models allowing for age, 3VW and metabolites. It revealed that the only relevant predictors of MSSS were 3VW and Glx/NAA. At a significance level of P<0.05, a unit increase of 3VW was associated with a 0.35 increase of MSSS, for a typical value of Glx/NAA; P value 0.0039. A unit increase of Glx/NAA was associated with a 0.93 increase of MSSS, for a typical value of atrophy; P value 0.090. There were significant linear correlations between Glx/Cr and MSSS, Glx/NAA and MSSS, and between mIns/NAA and 3VW. CONCLUSIONS The results suggest that both NAA and Glx are associated with neurodegeneration of hypothalamic DGM and severe disease course. Glx related 1H-MRS parameters seem to be superior to other metabolites in determining disease burden, independently of otherwise powerful 3VW planimetry. Significantly increased mIns/NAA in MS patients compared to controls point to gliosis, which parallels the atrophy of hypothalamic DGM.
Collapse
|
43
|
Sarma SN, Saleem A, Lee JY, Tokumoto M, Hwang GW, Man Chan H, Satoh M. Effects of long-term cadmium exposure on urinary metabolite profiles in mice. J Toxicol Sci 2018; 43:89-100. [PMID: 29479038 DOI: 10.2131/jts.43.89] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Cadmium (Cd) is a common environmental pollutant with known toxic effects on the kidney. Urinary metabolomics is a promising approach to study mechanism by which Cd-induced nephrotoxicity. The aim of this study was to elucidate the mechanism of Cd toxicity and to develop specific biomarkers by identifying urinary metabolic changes after a long-term of Cd exposure and with the critical concentration of Cd in the kidney. Urine samples were collected from wild-type 129/Sv mice after 67 weeks of 300 ppm Cd exposure and analyzed by ultra performance liquid chromatography connected with quadrupole time of flight mass spectrometer (UPLC-QTOF-MS) based metabolomics approach. A total of 40 most differentiated metabolites (9 down-regulated and 31 up-regulated) between the control and Cd-exposed group were identified. The majority of the regulated metabolites are amino acids (glutamine, L-aspartic acid, phenylalanine, tryptophan, and D-proline) indicating that amino acid metabolism pathways are affected by long-term exposure of Cd. However, there are also some nucleotides (guanosine, guanosine monophosphate, cyclic AMP, uridine), amino acid derivatives (homoserine, N-acetyl-L-aspartate, N-acetylglutamine, acetyl-phenylalanine, carboxymethyllysine), and peptides. Results of pathway analysis showed that the arginine and proline metabolism, purine metabolism, alanine, aspartate and glutamate metabolism, and aminoacyl-tRNA biosynthesis were affected compared to the control. This study demonstrates that metabolomics is useful to elucidate the metabolic responses and biological effects induced by Cd-exposure.
Collapse
Affiliation(s)
| | - Ammar Saleem
- Department of Biology, University of Ottawa, Canada
| | - Jin-Yong Lee
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University
| | - Maki Tokumoto
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University
| | - Gi-Wook Hwang
- Laboratory of Molecular Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University
| | | | - Masahiko Satoh
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University
| |
Collapse
|
44
|
Duarte JMN, Xin L. Magnetic Resonance Spectroscopy in Schizophrenia: Evidence for Glutamatergic Dysfunction and Impaired Energy Metabolism. Neurochem Res 2018; 44:102-116. [PMID: 29616444 PMCID: PMC6345729 DOI: 10.1007/s11064-018-2521-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/28/2018] [Accepted: 03/30/2018] [Indexed: 01/03/2023]
Abstract
In the past couple of decades, major efforts were made to increase reliability of metabolic assessments by magnetic resonance methods. Magnetic resonance spectroscopy (MRS) has been valuable for providing in vivo evidence and investigating biomarkers in neuropsychiatric disorders, namely schizophrenia. Alterations of glutamate and glutamine levels in brains of schizophrenia patients relative to healthy subjects are generally interpreted as markers of glutamatergic dysfunction. However, only a small fraction of MRS-detectable glutamate is involved in neurotransmission. Here we review and discuss brain metabolic processes that involve glutamate and that are likely to be implicated in neuropsychiatric disorders.
Collapse
Affiliation(s)
- João M N Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, BMC C11, Sölvegatan 19, 221 84, Lund, Sweden. .,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.
| | - Lijing Xin
- Animal Imaging and Technology Core (AIT), Center for Biomedical Imaging (CIBM), Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
45
|
Posporelis S, Coughlin JM, Marsman A, Pradhan S, Tanaka T, Wang H, Varvaris M, Ward R, Higgs C, Edwards JA, Ford CN, Kim PK, Lloyd AM, Edden RAE, Schretlen DJ, Cascella NG, Barker PB, Sawa A. Decoupling of Brain Temperature and Glutamate in Recent Onset of Schizophrenia: A 7T Proton Magnetic Resonance Spectroscopy Study. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2018; 3:248-254. [PMID: 29486866 PMCID: PMC5836506 DOI: 10.1016/j.bpsc.2017.04.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/23/2017] [Accepted: 04/10/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND Converging evidence suggests that cerebral metabolic and cellular homeostasis is altered in patients with recent onset of schizophrenia. As a possible marker of metabolic changes that might link to altered neurotransmission, we used proton magnetic resonance spectroscopy to estimate brain temperature, and we evaluated its relationship to a relevant metabolite, glutamate, within this study population. METHODS Using proton magnetic resonance spectroscopy at 7T, 20 patients with recent onset (≤24 months after first psychotic symptoms) of schizophrenia and 20 healthy control subjects were studied. We measured levels of N-acetylaspartate and glutamate and estimated brain temperature in a noninvasive manner. RESULTS Healthy control subjects showed a significant negative correlation between glutamate and brain temperature in the anterior cingulate cortex. In contrast, the physiological correlation between glutamate and brain temperature was lost in patients with recent onset of schizophrenia. CONCLUSIONS This study supports the hypothesized disrupted relationship between brain metabolism and neurotransmission in patients with recent onset of schizophrenia. The findings include mechanistic implications that are to be followed up in both preclinical and clinical studies.
Collapse
Affiliation(s)
- Sotirios Posporelis
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland; South London and Maudsley National Health Service Foundation Trust, London, United Kingdom
| | - Jennifer M Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland; Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Anouk Marsman
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Subechhya Pradhan
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Teppei Tanaka
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Hongxing Wang
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Mark Varvaris
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Rebecca Ward
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Cecilia Higgs
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Jamie A Edwards
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Candice N Ford
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Pearl K Kim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Ashley M Lloyd
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - David J Schretlen
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Nicola G Cascella
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Peter B Barker
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland.
| |
Collapse
|
46
|
Ribeiro M, Castelhano J, Petrella LI, Sereno J, Rodrigues T, Neves C, Letra L, Baptista FI, Seiça R, Matafome P, Castelo-Branco M. High-fat diet induces a neurometabolic state characterized by changes in glutamate and N-acetylaspartate pools associated with early glucose intolerance: An in vivo multimodal MRI study. J Magn Reson Imaging 2018; 48:757-766. [PMID: 29377412 DOI: 10.1002/jmri.25942] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/18/2017] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Type-2 diabetes mellitus (T2DM) is a metabolic disorder with a broad range of complications in the brain that depend on the conditions that precede its onset, such as obesity and metabolic syndromes. It has been suggested that neurotransmitter and metabolic perturbations may emerge even before the early stages of T2DM and that high-caloric intake could adversely influence the brain in such states. Notwithstanding, evidence for neurochemical and structural alterations in these conditions are still sparse and controversial. PURPOSE To evaluate the influence of high-fat diet in the neurochemical profile and structural integrity of the rodent brain. STUDY TYPE Prospective. SUBJECTS Wistar rats (n = 12/group). FIELD STRENGTH/SEQUENCE A PRESS, ISIS, RARE, and EPI sequences were performed at 9.4T. ASSESSMENT Neurochemical and structural parameters were assessed by magnetic resonance spectroscopy, voxel-based morphometry, volumetry, and diffusion tensor imaging. STATISTICAL TESTS Measurements were compared through Student and Mann-Whitney tests. Pearson correlation was used to assess relationships between parameters. RESULTS Animals submitted to high-caloric intake gained weight (P = 0.003) and developed glucose intolerance (P < 0.001) but not hyperglycemia. In the hippocampus, the diet induced perturbations in glutamatergic metabolites reflected by increased levels of glutamine (P = 0.016) and glutamatergic pool (Glx) (P = 0.036), which were negatively correlated with glucose intolerance (glutamine, r = -0.804, P = 0.029), suggesting a link with neurometabolic dysregulation. At caudate-putamen, high-fat diet led to a surprising increase in the pool of N-acetylaspartate (P = 0.028). A relation with metabolic changes was again suggested by the negative correlation between glucose intolerance and levels of glutamatergic metabolites in this region (glutamate, r = -0.845, P = 0.014; Glx, r = -0.834, P = 0.020). Neither changes in phosphate compounds nor major structural alterations were observed for both regions. DATA CONCLUSION We found evidence that high-fat diet-induced obesity leads to distinct early and region-specific metabolic/neurochemical imbalances in the presence of early glucose intolerance even when structural alterations or T2DM are absent. LEVEL OF EVIDENCE 1 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2018.
Collapse
Affiliation(s)
- Mário Ribeiro
- Center for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Coimbra, Portugal
- CiBIT, Institute of Nuclear Science Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - João Castelhano
- Center for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Coimbra, Portugal
- CiBIT, Institute of Nuclear Science Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - Lorena I Petrella
- Center for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Coimbra, Portugal
- CiBIT, Institute of Nuclear Science Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - José Sereno
- Center for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Coimbra, Portugal
- CiBIT, Institute of Nuclear Science Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - Tiago Rodrigues
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), University of Coimbra, Coimbra, Portugal
| | - Christian Neves
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), University of Coimbra, Coimbra, Portugal
| | - Liliana Letra
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), University of Coimbra, Coimbra, Portugal
| | - Filipa I Baptista
- Center for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Coimbra, Portugal
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), University of Coimbra, Coimbra, Portugal
| | - Raquel Seiça
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), University of Coimbra, Coimbra, Portugal
| | - Paulo Matafome
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), University of Coimbra, Coimbra, Portugal
- Coimbra Health School (ESTeSC), Department of Complementary Sciences, University of Coimbra, Coimbra, Portugal
| | - Miguel Castelo-Branco
- Center for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Coimbra, Portugal
- CiBIT, Institute of Nuclear Science Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
47
|
Rigucci S, Xin L, Klauser P, Baumann PS, Alameda L, Cleusix M, Jenni R, Ferrari C, Pompili M, Gruetter R, Do KQ, Conus P. Cannabis use in early psychosis is associated with reduced glutamate levels in the prefrontal cortex. Psychopharmacology (Berl) 2018; 235:13-22. [PMID: 29075884 DOI: 10.1007/s00213-017-4745-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 08/06/2017] [Accepted: 09/07/2017] [Indexed: 11/26/2022]
Abstract
RATIONALE Recent studies have shown that cannabis may disrupt glutamate (Glu) signaling depressing Glu tone in frequent users. Current evidence have also consistently reported lower Glu-levels in various brain regions, particularly in the medial prefrontal cortex (mPFC) of chronic schizophrenia patients, while findings in early psychosis (EP) are not conclusive. Since cannabis may alter Glu synaptic plasticity and its use is a known risk factor for psychosis, studies focusing on Glu signaling in EP with or without a concomitant cannabis-usage seem crucial. OBJECTIVE We investigate the effect of cannabis use on prefrontal Glu-levels in EP users vs. both EP non-users and healthy controls (HC). METHODS Magnetic resonance spectroscopy was used to measure [GlumPFC] of 35 EP subjects (18 of whom were cannabis users) and 33 HC. For correlative analysis, neuropsychological performances were scored by the MATRICS-consensus cognitive battery. RESULTS [GlumPFC] was lower in EP users comparing to both HC and EP non-users (p < 0.001 and p = 0.01, respectively), while no differences were observed between EP non-users and HC. A greater [GlumPFC]-decline with age was observed in EP users (r = -.46; p = 0.04), but not in EP non-users or HC. Among neuropsychological outcomes, working memory was the only domain that differentiates patients depending on their cannabis use, with users having poorer performances. CONCLUSIONS Cannabis use is associated with reduced prefrontal [GlumPFC] and with a stronger Glu-levels decline with age. Glutamatergic abnormalities might influence the cognitive impairment observed in users and have some relevance for the progression of the disease.
Collapse
Affiliation(s)
- Silvia Rigucci
- TIPP (Treatment and Early Intervention in Psychosis Program); Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Department of Neurosciences, Sensory Organs and Mental Health, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Lijing Xin
- Animal imaging and technology core (AIT), Center for Biomedical Imaging (CIBM), Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Paul Klauser
- TIPP (Treatment and Early Intervention in Psychosis Program); Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Philipp S Baumann
- TIPP (Treatment and Early Intervention in Psychosis Program); Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Luis Alameda
- TIPP (Treatment and Early Intervention in Psychosis Program); Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Martine Cleusix
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Raoul Jenni
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Carina Ferrari
- TIPP (Treatment and Early Intervention in Psychosis Program); Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Maurizio Pompili
- Department of Neurosciences, Sensory Organs and Mental Health, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Rolf Gruetter
- Laboratory of Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Departments of Radiology, University of Lausanne, Lausanne, Switzerland
- Department of Radiology, University of Geneva, Geneva, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland.
| | - Philippe Conus
- TIPP (Treatment and Early Intervention in Psychosis Program); Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
48
|
Kantorová E, Poláček H, Bittšanský M, Baranovičová E, Hnilicová P, Čierny D, Sivák Š, Nosáľ V, Zeleňák K, Kurča E. Hypothalamic damage in multiple sclerosis correlates with disease activity, disability, depression, and fatigue. Neurol Res 2017; 39:323-330. [DOI: 10.1080/01616412.2016.1275460] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- E. Kantorová
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Slovakia
| | - H. Poláček
- Clinic of Nuclear Medicine, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Bratislava, Slovakia
| | - M. Bittšanský
- BioMed Division of Neurosciences, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Bratislava, Slovakia
| | - E. Baranovičová
- BioMed Division of Neurosciences, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Bratislava, Slovakia
| | - P. Hnilicová
- BioMed Division of Neurosciences, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Bratislava, Slovakia
| | - D. Čierny
- Department of Clinical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Bratislava, Slovakia
| | - Š. Sivák
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Slovakia
| | - V. Nosáľ
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Slovakia
| | - K. Zeleňák
- Clinic of Radiodiagnostics, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Bratislava, Slovakia
| | - E. Kurča
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Slovakia
| |
Collapse
|
49
|
Fischer IW, Hansen TM, Lelic D, Brokjaer A, Frøkjær J, Christrup LL, Olesen AE. Objective methods for the assessment of the spinal and supraspinal effects of opioids. Scand J Pain 2016; 14:15-24. [PMID: 28850426 DOI: 10.1016/j.sjpain.2016.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 09/30/2016] [Accepted: 10/03/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND PURPOSE Opioids are potent analgesics. Opioids exert effects after interaction with opioid receptors. Opioid receptors are present in the peripheral- and central nervous system (CNS), but the analgesic effects are primarily mediated via receptors in the CNS. Objective methods for assessment of opioid effects may increase knowledge on the CNS processes responsible for analgesia. The aim of this review was to provide an overview of the most common objective methods for assessment of the spinal and supraspinal effects of opioids and discuss their advantages and limitations. METHOD The literature search was conducted in Pub Med (http://www.ncbi.nlm.nih.gov/pubmed) from November 2014 to June 2016, using free-text terms: "opioid", "morphine" and "oxycodone" combined with the terms "pupillometry," "magnetic resonance spectroscopy," "fMRI," "BOLD," "PET," "pharmaco-EEG", "electroencephalogram", "EEG," "evoked potentials," and "nociceptive reflex". Only original articles published in English were included. RESULTS For assessment of opioid effects at the supraspinal level, the following methods are evaluated: pupillometry, proton magnetic resonance spectroscopy, functional resonance magnetic imaging (fMRI), positron emission tomography (PET), spontaneous electroencephalogram (EEG) and evoked potentials (EPs). Pupillometry is a non-invasive tool used in research as well as in the clinical setting. Proton magnetic resonance spectroscopy has been used for the last decades and it is a non-invasive technique for measurement of in vivo brain metabolite concentrations. fMRI has been a widely used non-invasive method to estimate brain activity, where typically from the blood oxygen level-dependent (BOLD) signal. PET is a nuclear imaging technique based on tracing radio labeled molecules injected into the blood, where receptor distribution, density and activity in the brain can be visualized. Spontaneous EEG is typically quantified in frequency bands, power spectrum and spectral edge frequency. EPs are brain responses (assessed by EEG) to a predefined number of short phasic stimuli. EPs are quantified by their peak latencies and amplitudes, power spectrum, scalp topographies and brain source localization. For assessment of opioid effects at the spinal level, the following methods are evaluated: the nociceptive withdrawal reflex (NWR) and spinal EPs. The nociceptive withdrawal reflex can be recorded from all limbs, but it is standard to record the electromyography signal at the biceps femoris muscle after stimulation of the ipsilateral sural nerve; EPs can be recorded from the spinal cord and are typically recorded after stimulation of the median nerve at the wrist. CONCLUSION AND IMPLICATIONS The presented methods can all be used as objective methods for assessing the centrally mediated effects of opioids. Advantages and limitations should be considered before implementation in drug development, future experimental studies as well as in clinical settings. In conclusion, pupillometry is a sensitive measurement of opioid receptor activation in the CNS and from a practical and economical perspective it may be used as a biomarker for opioid effects in the CNS. However, if more detailed information is needed on opioid effects at different levels of the CNS, then EEG, fMRI, PET and NWR have the potential to be used. Finally, it is conceivable that information from different methods should be considered together for complementary information.
Collapse
Affiliation(s)
- Iben W Fischer
- Mech-Sense, Department of Gastroenterology &Hepatology, Aalborg University Hospital, Mølleparkvej 4, 9000, Aalborg, Denmark.,Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tine M Hansen
- Mech-Sense, Department of Radiology, Aalborg University Hospital, Hobrovej 18-22, 9000, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Dina Lelic
- Mech-Sense, Department of Gastroenterology &Hepatology, Aalborg University Hospital, Mølleparkvej 4, 9000, Aalborg, Denmark
| | - Anne Brokjaer
- Mech-Sense, Department of Gastroenterology &Hepatology, Aalborg University Hospital, Mølleparkvej 4, 9000, Aalborg, Denmark
| | - Jens Frøkjær
- Mech-Sense, Department of Radiology, Aalborg University Hospital, Hobrovej 18-22, 9000, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Lona L Christrup
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne E Olesen
- Mech-Sense, Department of Gastroenterology &Hepatology, Aalborg University Hospital, Mølleparkvej 4, 9000, Aalborg, Denmark.,Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
50
|
Amorini AM, Lazzarino G, Di Pietro V, Signoretti S, Lazzarino G, Belli A, Tavazzi B. Severity of experimental traumatic brain injury modulates changes in concentrations of cerebral free amino acids. J Cell Mol Med 2016; 21:530-542. [PMID: 27696676 PMCID: PMC5323875 DOI: 10.1111/jcmm.12998] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 08/26/2016] [Indexed: 12/29/2022] Open
Abstract
In this study, concentrations of free amino acids (FAA) and amino group containing compounds (AGCC) following graded diffuse traumatic brain injury (mild TBI, mTBI; severe TBI, sTBI) were evaluated. After 6, 12, 24, 48 and 120 hr aspartate (Asp), glutamate (Glu), asparagine (Asn), serine (Ser), glutamine (Gln), histidine (His), glycine (Gly), threonine (Thr), citrulline (Cit), arginine (Arg), alanine (Ala), taurine (Tau), γ‐aminobutyrate (GABA), tyrosine (Tyr), S‐adenosylhomocysteine (SAH), l‐cystathionine (l‐Cystat), valine (Val), methionine (Met), tryptophane (Trp), phenylalanine (Phe), isoleucine (Ile), leucine (Leu), ornithine (Orn), lysine (Lys), plus N‐acetylaspartate (NAA) were determined in whole brain extracts (n = 6 rats at each time for both TBI levels). Sham‐operated animals (n = 6) were used as controls. Results demonstrated that mTBI caused modest, transient changes in NAA, Asp, GABA, Gly, Arg. Following sTBI, animals showed profound, long‐lasting modifications of Glu, Gln, NAA, Asp, GABA, Ser, Gly, Ala, Arg, Citr, Tau, Met, SAH, l‐Cystat, Tyr and Phe. Increase in Glu and Gln, depletion of NAA and Asp increase, suggested a link between NAA hydrolysis and excitotoxicity after sTBI. Additionally, sTBI rats showed net imbalances of the Glu‐Gln/GABA cycle between neurons and astrocytes, and of the methyl‐cycle (demonstrated by decrease in Met, and increase in SAH and l‐Cystat), throughout the post‐injury period. Besides evidencing new potential targets for novel pharmacological treatments, these results suggest that the force acting on the brain tissue at the time of the impact is the main determinant of the reactions ignited and involving amino acid metabolism.
Collapse
Affiliation(s)
- Angela Maria Amorini
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Rome, Rome, Italy
| | - Giacomo Lazzarino
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Rome, Rome, Italy
| | - Valentina Di Pietro
- Neuroscience and Ophthalmology group, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Stefano Signoretti
- Division of Neurosurgery, Department of Neurosciences Head and Neck Surgery, S. Camillo Hospital, Rome, Italy
| | - Giuseppe Lazzarino
- Division of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Antonio Belli
- Neuroscience and Ophthalmology group, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital, Birmingham, UK
| | - Barbara Tavazzi
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Rome, Rome, Italy
| |
Collapse
|