1
|
Anderson BG, Popov P, Cicali AR, Nwamba A, Evans CR, Kennedy RT. In-Depth Chemical Analysis of the Brain Extracellular Space Using In Vivo Microdialysis with Liquid Chromatography-Tandem Mass Spectrometry. Anal Chem 2024; 96:16387-16396. [PMID: 39360623 DOI: 10.1021/acs.analchem.4c03806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Metabolomic analysis of samples acquired in vivo from the brain extracellular space by microdialysis sampling can provide insights into chemical underpinnings of a given brain state and how it changes over time. Small sample volumes and low physiological concentrations have limited the identification of compounds from this compartment, so at present, we have scant knowledge of its composition. As a result, most in vivo measurements have limited depth of analysis. Here, we describe an approach to (1) identify hundreds of compounds in brain dialysate and (2) routinely detect many of these compounds in 5 μL microdialysis samples to enable deep monitoring of brain chemistry in time-resolved studies. Dialysate samples collected over 12 h were concentrated 10-fold and then analyzed using liquid chromatography with iterative tandem mass spectrometry (LC-MS/MS). Using this approach on dialysate from the rat striatum with both reversed-phase and hydrophilic interaction liquid chromatography yielded 479 unique compound identifications. 60% of the identified compounds could be detected in 5 μL of dialysate without further concentration using a single 20 min LC-MS analysis, showing that once identified, most compounds can be detected using small sample volumes and shorter analysis times compatible with routine in vivo monitoring. To detect more neurochemicals, LC-MS analysis of dialysate derivatized with light and isotopically labeled benzoyl chloride was employed. 872 nondegenerate benzoylated features were detected with this approach, including most small-molecule neurotransmitters and several metabolites involved in dopamine metabolism. This strategy allows deeper annotation of the brain extracellular space than previously possible and provides a launching point for defining the chemistry underlying brain states.
Collapse
Affiliation(s)
- Brady G Anderson
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Pavlo Popov
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Psychology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Amanda R Cicali
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Adanna Nwamba
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Charles R Evans
- Biomedical Research Core Facilities Metabolomics Core, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Robert T Kennedy
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
2
|
Wang X, Peng R, Zhao L. Multiscale metabolomics techniques: Insights into neuroscience research. Neurobiol Dis 2024; 198:106541. [PMID: 38806132 DOI: 10.1016/j.nbd.2024.106541] [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: 04/10/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024] Open
Abstract
The field of metabolomics examines the overall composition and dynamic patterns of metabolites in living organisms. The primary methods used in metabolomics include liquid chromatography (LC), nuclear magnetic resonance (NMR), and mass spectrometry (MS) analysis. These methods enable the identification and examination of metabolite types and contents within organisms, as well as modifications to metabolic pathways and their connection to the emergence of diseases. Research in metabolomics has extensive value in basic and applied sciences. The field of metabolomics is growing quickly, with the majority of studies concentrating on biomedicine, particularly early disease diagnosis, therapeutic management of human diseases, and mechanistic knowledge of biochemical processes. Multiscale metabolomics is an approach that integrates metabolomics techniques at various scales, including the holistic, tissue, cellular, and organelle scales, to enable more thorough and in-depth studies of metabolic processes in organisms. Multiscale metabolomics can be combined with methods from systems biology and bioinformatics. In recent years, multiscale metabolomics approaches have become increasingly important in neuroscience research due to the nervous system's high metabolic demands. Multiscale metabolomics can offer novel concepts and approaches for the diagnosis, treatment, and development of medication for neurological illnesses in addition to a more thorough understanding of brain metabolism and nervous system function. In this review, we summarize the use of multiscale metabolomics techniques in neuroscience, address the promise and constraints of these techniques, and provide an overview of the metabolome and its applications in neuroscience.
Collapse
Affiliation(s)
- Xiaoya Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ruiyun Peng
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Li Zhao
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
3
|
Wu X, Xu H, Zeng N, Li H, Yao G, Liu K, Yan C, Wu L. Luteolin alleviates depression-like behavior by modulating glycerophospholipid metabolism in the hippocampus and prefrontal cortex of LOD rats. CNS Neurosci Ther 2024; 30:e14455. [PMID: 37715585 PMCID: PMC10916417 DOI: 10.1111/cns.14455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/01/2023] [Accepted: 08/23/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Late-onset depression (LOD) is defined as primary depression that first manifests after the age of 65. Luteolin (LUT) is a natural flavonoid that has shown promising antidepressant effects and improvement in neurological function in previous studies. AIMS In this study, we utilized UPLC-MS/MS non-targeted metabolomics techniques, along with molecular docking technology and experimental validation, to explore the mechanism of LUT in treating LOD from a metabolomics perspective. RESULTS The behavioral results of our study demonstrate that LUT significantly ameliorated anxiety and depression-like behaviors while enhancing cognitive function in LOD rats. Metabolomic analysis revealed that the effects of LUT on LOD rats were primarily mediated through the glycerophospholipid metabolic pathway in the hippocampus and prefrontal cortex. The levels of key lipid metabolites, phosphatidylserine (PS), phosphatidylcholine (PC), and phosphatidylethanolamine (PE), in the glycerophospholipid metabolic pathway were significantly altered by LUT treatment, with PC and PE showing significant correlations with behavioral indices. Molecular docking analysis indicated that LUT had strong binding activity with phosphatidylserine synthase 1 (PTDSS1), phosphatidylserine synthase 2 (PTDSS2), and phosphatidylserine decarboxylase (PISD), which are involved in the transformation and synthesis of PC, PE, and PS. Lastly, our study explored the reasons for the opposing trends of PC, PE, and PS in the hippocampus and prefrontal cortex from the perspective of autophagy, which may be attributable to the bidirectional regulation of autophagy in distinct brain regions. CONCLUSIONS Our results revealed significant alterations in the glycerophospholipid metabolism pathways in both the hippocampus and prefrontal cortex of LOD rats. Moreover, LUT appears to regulate autophagy disorders by specifically modulating glycerophospholipid metabolism in different brain regions of LOD rats, consequently alleviating depression-like behavior in these animals.
Collapse
Affiliation(s)
- Xiaofeng Wu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese MedicineGuangzhou University of Chinese MedicineGuangzhouChina
| | - Hanfang Xu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese MedicineGuangzhou University of Chinese MedicineGuangzhouChina
| | - Ningxi Zeng
- Department of Rehabilitation Medicine, The People's Hospital of Longhua DistrictShenzhenChina
| | - Huizhen Li
- Key Laboratory of Depression Animal Model Based on TCM Syndrome, Key Laboratory of TCM for Prevention and Treatment of Brain Diseases with Cognitive DysfunctionJiangxi University of Chinese MedicineNanchangChina
| | - Gaolei Yao
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese MedicineGuangzhou University of Chinese MedicineGuangzhouChina
| | - Kaige Liu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese MedicineGuangzhou University of Chinese MedicineGuangzhouChina
| | - Can Yan
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese MedicineGuangzhou University of Chinese MedicineGuangzhouChina
| | - Lili Wu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese MedicineGuangzhou University of Chinese MedicineGuangzhouChina
| |
Collapse
|
4
|
Shao Y, Fu Z, Wang Y, Yang Z, Lin Y, Li S, Cheng C, Wei M, Liu Z, Xu G, Le W. A metabolome atlas of mouse brain on the global metabolic signature dynamics following short-term fasting. Signal Transduct Target Ther 2023; 8:334. [PMID: 37679319 PMCID: PMC10484938 DOI: 10.1038/s41392-023-01552-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 06/07/2023] [Accepted: 06/21/2023] [Indexed: 09/09/2023] Open
Abstract
Calorie restriction (CR) or a fasting regimen is considered one of the most potent non-pharmacological interventions to prevent chronic metabolic disorders, ameliorate autoimmune diseases, and attenuate aging. Despite efforts, the mechanisms by which CR improves health, particularly brain health, are still not fully understood. Metabolic homeostasis is vital for brain function, and a detailed metabolome atlas of the brain is essential for understanding the networks connecting different brain regions. Herein, we applied gas chromatography-mass spectrometry-based metabolomics and lipidomics, covering 797 structurally annotated metabolites, to investigate the metabolome of seven brain regions in fasted (3, 6, 12, and 24 h) and ad libitum fed mice. Using multivariate and univariate statistical techniques, we generated a metabolome atlas of mouse brain on the global metabolic signature dynamics across multiple brain regions following short-term fasting (STF). Significant metabolic differences across brain regions along with STF-triggered region-dependent metabolic remodeling were identified. We found that STF elicited triacylglycerol degradation and lipolysis to compensate for energy demand under fasting conditions. Besides, changes in amino acid profiles were observed, which may play crucial roles in the regulation of energy metabolism, neurotransmitter signaling, and anti-inflammatory and antioxidant in response to STF. Additionally, this study reported, for the first time, that STF triggers a significant elevation of N-acylethanolamines, a class of neuroprotective lipids, in the brain and liver. These findings provide novel insights into the molecular basis and mechanisms of CR and offer a comprehensive resource for further investigation.
Collapse
Affiliation(s)
- Yaping Shao
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, 193 Lianhe Road, 116021, Dalian, China.
| | - Zhenfa Fu
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, 193 Lianhe Road, 116021, Dalian, China
| | - Yanfeng Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, 116023, Dalian, China
| | - Zhaofei Yang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, 193 Lianhe Road, 116021, Dalian, China
| | - Yushan Lin
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, 193 Lianhe Road, 116021, Dalian, China
| | - Song Li
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, 193 Lianhe Road, 116021, Dalian, China
| | - Cheng Cheng
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, 193 Lianhe Road, 116021, Dalian, China
| | - Min Wei
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, 193 Lianhe Road, 116021, Dalian, China
| | - Zheyi Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, 116023, Dalian, China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, 116023, Dalian, China.
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, 193 Lianhe Road, 116021, Dalian, China.
- Institute of Neurology, Sichuan Academy of Medical Science-Sichuan Provincial Hospital, Medical School of UESTC, 611731, Chengdu, China.
| |
Collapse
|
5
|
Cappuccio G, Khalil SM, Osenberg S, Li F, Maletic-Savatic M. Mass spectrometry imaging as an emerging tool for studying metabolism in human brain organoids. Front Mol Biosci 2023; 10:1181965. [PMID: 37304070 PMCID: PMC10251497 DOI: 10.3389/fmolb.2023.1181965] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/02/2023] [Indexed: 06/13/2023] Open
Abstract
Human brain organoids are emerging models to study human brain development and pathology as they recapitulate the development and characteristics of major neural cell types, and enable manipulation through an in vitro system. Over the past decade, with the advent of spatial technologies, mass spectrometry imaging (MSI) has become a prominent tool for metabolic microscopy, providing label-free, non-targeted molecular and spatial distribution information of the metabolites within tissue, including lipids. This technology has never been used for studies of brain organoids and here, we set out to develop a standardized protocol for preparation and mass spectrometry imaging of human brain organoids. We present an optimized and validated sample preparation protocol, including sample fixation, optimal embedding solution, homogenous deposition of matrices, data acquisition and processing to maximize the molecular information derived from mass spectrometry imaging. We focus on lipids in organoids, as they play critical roles during cellular and brain development. Using high spatial and mass resolution in positive- and negative-ion modes, we detected 260 lipids in the organoids. Seven of them were uniquely localized within the neurogenic niches or rosettes as confirmed by histology, suggesting their importance for neuroprogenitor proliferation. We observed a particularly striking distribution of ceramide-phosphoethanolamine CerPE 36:1; O2 which was restricted within rosettes and of phosphatidyl-ethanolamine PE 38:3, which was distributed throughout the organoid tissue but not in rosettes. This suggests that ceramide in this particular lipid species might be important for neuroprogenitor biology, while its removal may be important for terminal differentiation of their progeny. Overall, our study establishes the first optimized experimental pipeline and data processing strategy for mass spectrometry imaging of human brain organoids, allowing direct comparison of lipid signal intensities and distributions in these tissues. Further, our data shed new light on the complex processes that govern brain development by identifying specific lipid signatures that may play a role in cell fate trajectories. Mass spectrometry imaging thus has great potential in advancing our understanding of early brain development as well as disease modeling and drug discovery.
Collapse
Affiliation(s)
- Gerarda Cappuccio
- Department of Pediatrics–Neurology, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Saleh M. Khalil
- Department of Pediatrics–Neurology, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Sivan Osenberg
- Department of Pediatrics–Neurology, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Feng Li
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States
| | - Mirjana Maletic-Savatic
- Department of Pediatrics–Neurology, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
6
|
Ramírez-Acosta S, Huertas-Abril PV, Selma-Royo M, Prieto-Álamo MJ, Collado MC, Abril N, García-Barrera T. The role of selenium in shaping mice brain metabolome and selenoproteome through the gut-brain axis by combining metabolomics, metallomics, gene expression and amplicon sequencing. J Nutr Biochem 2023; 117:109323. [PMID: 36958417 DOI: 10.1016/j.jnutbio.2023.109323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 02/17/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023]
Abstract
Selenium (Se) is a trace element crucial for human health. Recently, the impact of Se supplementation on gut microbiota has been pointed out as well as its influence on the expression of certain selenoproteins and gut metabolites. This study aims to elucidate the link between Se supplementation, brain selenoproteins and brain metabolome as well as the possible connection with the gut-brain axis. To this end, an in vivo study with 40 BALB/c mice was carried out. The study included conventional (n=20) and mice model with microbiota depleted by antibiotics (n=20) under a regular or Se supplemented diet. Brain selenoproteome was determined by a transcriptomic/gene expression profile, while brain metabolome and gut microbiota profiles were accomplished by untargeted metabolomics and amplicon sequencing, respectively. The total content of Se in brain was also determined. The selenoproteins genes Dio and Gpx isoenzymes, SelenoH, SelenoI, SelenoT, SelenoV and SelenoW and 31 metabolites were significantly altered in the brain after Se supplementation in conventional mice, while 11 selenoproteins and 26 metabolites were altered in microbiota depleted mice. The main altered brain metabolites were related to glyoxylate and dicarboxylate metabolism, amino acid metabolism, and gut microbiota that have been previously related with the gut-brain axis (e.g., members of Lachnospiraceae and Ruminococcaceae families). Moreover, specific associations were determined between brain selenoproteome and metabolome, which correlated with the same bacteria, suggesting an intertwined mechanism. Our results demonstrated the effect of Se on brain metabolome through specific selenoproteins gene expression and gut microbiota.
Collapse
Affiliation(s)
- Sara Ramírez-Acosta
- Research Center of Natural Resources, Health and the Environment (RENSMA). Department of Chemistry, Faculty of Experimental Sciences, University of Huelva, Fuerzas Armadas Ave., 21007, Huelva, Spain
| | - Paula V Huertas-Abril
- Department of Biochemistry and Molecular Biology, University of Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain
| | - Marta Selma-Royo
- Institute of Agrochemistry and Food Technology (IATA-CSIC), Department of Biotechnology, Agustin Escardino 7. 46980 Paterna, Valencia, Spain
| | - Maria J Prieto-Álamo
- Department of Biochemistry and Molecular Biology, University of Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain
| | - M Carmen Collado
- Institute of Agrochemistry and Food Technology (IATA-CSIC), Department of Biotechnology, Agustin Escardino 7. 46980 Paterna, Valencia, Spain
| | - Nieves Abril
- Department of Biochemistry and Molecular Biology, University of Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain
| | - Tamara García-Barrera
- Research Center of Natural Resources, Health and the Environment (RENSMA). Department of Chemistry, Faculty of Experimental Sciences, University of Huelva, Fuerzas Armadas Ave., 21007, Huelva, Spain.
| |
Collapse
|
7
|
Zhang F, Rakhimbekova A, Lashley T, Madl T. Brain regions show different metabolic and protein arginine methylation phenotypes in frontotemporal dementias and Alzheimer's disease. Prog Neurobiol 2023; 221:102400. [PMID: 36581185 DOI: 10.1016/j.pneurobio.2022.102400] [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: 08/06/2022] [Revised: 11/05/2022] [Accepted: 12/19/2022] [Indexed: 12/27/2022]
Abstract
Frontotemporal dementia (FTD) is a heterogeneous neurodegenerative disease with multiple histopathological subtypes. FTD patients share similar symptoms with Alzheimer's disease (AD). Hence, FTD patients are commonly misdiagnosed as AD, despite the consensus clinical diagnostic criteria. It is therefore of great clinical need to identify a biomarker that can distinguish FTD from AD and control individuals, and potentially further differentiate between FTD pathological subtypes. We conducted a metabolomic analysis on post-mortem human brain tissue from three regions: cerebellum, frontal cortex and occipital cortex from control, FTLD-TDP type A, type A-C9, type C and AD. Our results indicate that the brain subdivisions responsible for different functions show different metabolic patterns. We further explored the region-specific metabolic characteristics of different FTD subtypes and AD patients. Different FTD subtypes and AD share similar metabolic phenotypes in the cerebellum, but AD exhibited distinct metabolic patterns in the frontal and occipital regions compared to FTD. The identified brain region-specific metabolite biomarkers could provide a tool for distinguishing different FTD subtypes and AD and provide the first insights into the metabolic changes of FTLD-TDP type A, type A-C9, type C and AD in different regions of the brain. The importance of protein arginine methylation in neurodegenerative disease has come to light, so we investigated whether the arginine methylation level contributes to disease pathogenesis. Our findings provide new insights into the relationship between arginine methylation and metabolic changes in FTD subtypes and AD that could be further explored, to study the molecular mechanism of pathogenesis.
Collapse
Affiliation(s)
- Fangrong Zhang
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China; Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Research Unit Integrative Structural Biology, Medical University of Graz, 8010 Graz, Austria.
| | - Anastasia Rakhimbekova
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Research Unit Integrative Structural Biology, Medical University of Graz, 8010 Graz, Austria.
| | - Tammaryn Lashley
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; Queen Square Brain Bank for Neurological Diseases, UCL Queen Square Institute of Neurology, London, UK.
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Research Unit Integrative Structural Biology, Medical University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria.
| |
Collapse
|
8
|
Shih BB, Brown SM, Barrington J, Lefevre L, Mabbott NA, Priller J, Thompson G, Lawrence AB, McColl BW. Defining the pig microglial transcriptome reveals its core signature, regional heterogeneity, and similarity with human and rodent microglia. Glia 2023; 71:334-349. [PMID: 36120803 PMCID: PMC10087207 DOI: 10.1002/glia.24274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/06/2022]
Abstract
Microglia play key roles in brain homeostasis as well as responses to neurodegeneration and neuroinflammatory processes caused by physical disease and psychosocial stress. The pig is a physiologically relevant model species for studying human neurological disorders, many of which are associated with microglial dysfunction. Furthermore, pigs are an important agricultural species, and there is a need to understand how microglial function affects their welfare. As a basis for improved understanding to enhance biomedical and agricultural research, we sought to characterize pig microglial identity at genome-wide scale and conduct inter-species comparisons. We isolated pig hippocampal tissue and microglia from frontal cortex, hippocampus, and cerebellum, as well as alveolar macrophages from the lungs and conducted RNA-sequencing (RNAseq). By comparing the transcriptomic profiles between microglia, macrophages, and hippocampal tissue, we derived a set of 239 highly enriched genes defining the porcine core microglial signature. We found brain regional heterogeneity based on 150 genes showing significant (adjusted p < 0.01) regional variations and that cerebellar microglia were most distinct. We compared normalized gene expression for microglia from human, mice and pigs using microglia signature gene lists derived from each species and demonstrated that a core microglial marker gene signature is conserved across species, but that species-specific expression subsets also exist. Our data provide a valuable resource defining the pig microglial transcriptome signature that validates and highlights pigs as a useful large animal species bridging between rodents and humans in which to study the role of microglia during homeostasis and disease.
Collapse
Affiliation(s)
- Barbara B Shih
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Sarah M Brown
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Jack Barrington
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh Medical School, The Chancellor's Building, Edinburgh, UK.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Lucas Lefevre
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh Medical School, The Chancellor's Building, Edinburgh, UK.,Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Neil A Mabbott
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Josef Priller
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh Medical School, The Chancellor's Building, Edinburgh, UK.,Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University Munich, Munich, Germany.,DZNE, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Gerard Thompson
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Alistair B Lawrence
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK.,Scotland's Rural College (SRUC), Edinburgh, UK
| | - Barry W McColl
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh Medical School, The Chancellor's Building, Edinburgh, UK.,Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
9
|
Yeo T, Bayuangga H, Augusto-Oliveira M, Sealey M, Claridge TDW, Tanner R, Leppert D, Palace J, Kuhle J, Probert F, Anthony DC. Metabolomics detects clinically silent neuroinflammatory lesions earlier than neurofilament-light chain in a focal multiple sclerosis animal model. J Neuroinflammation 2022; 19:252. [PMID: 36210459 PMCID: PMC9549622 DOI: 10.1186/s12974-022-02614-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 09/26/2022] [Indexed: 11/29/2022] Open
Abstract
Background Despite widespread searches, there are currently no validated biofluid markers for the detection of subclinical neuroinflammation in multiple sclerosis (MS). The dynamic nature of human metabolism in response to changes in homeostasis, as measured by metabolomics, may allow early identification of clinically silent neuroinflammation. Using the delayed-type hypersensitivity (DTH) MS rat model, we investigated the serum and cerebrospinal fluid (CSF) metabolomics profiles and neurofilament-light chain (NfL) levels, as a putative marker of neuroaxonal damage, arising from focal, clinically silent neuroinflammatory brain lesions and their discriminatory abilities to distinguish DTH animals from controls. Methods 1H nuclear magnetic resonance (NMR) spectroscopy metabolomics and NfL measurements were performed on serum and CSF at days 12, 28 and 60 after DTH lesion initiation. Supervised multivariate analyses were used to determine metabolomics differences between DTH animals and controls. Immunohistochemistry was used to assess the extent of neuroinflammation and tissue damage. Results Serum and CSF metabolomics perturbations were detectable in DTH animals (vs. controls) at all time points, with the greatest change occurring at the earliest time point (day 12) when the neuroinflammatory response was most intense (mean predictive accuracy [SD]—serum: 80.6 [10.7]%, p < 0.0001; CSF: 69.3 [13.5]%, p < 0.0001). The top discriminatory metabolites at day 12 (serum: allantoin, cytidine; CSF: glutamine, glucose) were all reduced in DTH animals compared to controls, and correlated with histological markers of neuroinflammation, particularly astrogliosis (Pearson coefficient, r—allantoin: r = − 0.562, p = 0.004; glutamine: r = − 0.528, p = 0.008). Serum and CSF NfL levels did not distinguish DTH animals from controls at day 12, rather, significant differences were observed at day 28 (mean [SEM]—serum: 38.5 [4.8] vs. 17.4 [2.6] pg/mL, p = 0.002; CSF: 1312.0 [379.1] vs. 475.8 [74.7] pg/mL, p = 0.027). Neither serum nor CSF NfL levels correlated with markers of neuroinflammation; serum NfL did, however, correlate strongly with axonal loss (r = 0.641, p = 0.001), but CSF NfL did not (p = 0.137). Conclusions While NfL levels were elevated later in the pathogenesis of the DTH lesion, serum and CSF metabolomics were able to detect early, clinically silent neuroinflammation and are likely to present sensitive biomarkers for the assessment of subclinical disease activity in patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02614-8.
Collapse
|
10
|
Abstract
In this systematic review, we highlight the differences between the male and female zebrafish brains to understand their differentiation and their use in studying sex-specific neurological diseases. Male and female brains display subtle differences at the cellular level which may be important in driving sex-specific signaling. Sex differences in the brain have been observed in humans as well as in non-human species. However, the molecular mechanisms of brain sex differentiation remain unclear. The classical model of brain sex differentiation suggests that the steroid hormones derived from the gonads are the primary determinants in establishing male and female neural networks. Recent studies indicate that the developing brain shows sex-specific differences in gene expression prior to gonadal hormone action. Hence, genetic differences may also be responsible for differentiating the brain into male and female types. Understanding the signaling mechanisms involved in brain sex differentiation could help further elucidate the sex-specific incidences of certain neurological diseases. The zebrafish model could be appropriate for enhancing our understanding of brain sex differentiation and the signaling involved in neurological diseases. Zebrafish brains show sex-specific differences at the hormonal level, and recent advances in RNA sequencing have highlighted critical sex-specific differences at the transcript level. The differences are also evident at the cellular and metabolite levels, which could be important in organizing sex-specific neuronal signaling. Furthermore, in addition to having one ortholog for 70% of the human gene, zebrafish also shares brain structural similarities with other higher eukaryotes, including mammals. Hence, deciphering brain sex differentiation in zebrafish will help further enhance the diagnostic and pharmacological intervention of neurological diseases.
Collapse
|
11
|
Nguyen TVV, Crumpacker RH, Calderon KE, Garcia FG, Zbesko JC, Frye JB, Gonzalez S, Becktel DA, Yang T, Tavera-Garcia MA, Morrison HW, Schnellmann RG, Longo FM, Doyle KP. Post-Stroke Administration of the p75 Neurotrophin Receptor Modulator, LM11A-31, Attenuates Chronic Changes in Brain Metabolism, Increases Neurotransmitter Levels, and Improves Recovery. J Pharmacol Exp Ther 2022; 380:126-141. [PMID: 34893553 PMCID: PMC11048261 DOI: 10.1124/jpet.121.000711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 11/29/2021] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to test whether poststroke oral administration of a small molecule p75 neurotrophin receptor (p75NTR) modulator (LM11A-31) can augment neuronal survival and improve recovery in a mouse model of stroke. Mice were administered LM11A-31 for up to 12 weeks, beginning 1 week after stroke. Metabolomic analysis revealed that after 2 weeks of daily treatment, mice that received LM11A-31 were distinct from vehicle-treated mice by principal component analysis and had higher levels of serotonin, acetylcholine, and dopamine in their ipsilateral hemisphere. LM11A-31 treatment also improved redox homeostasis by restoring reduced glutathione. It also offset a stroke-induced reduction in glycolysis by increasing acetyl-CoA. There was no effect on cytokine levels in the infarct. At 13 weeks after stroke, adaptive immune cell infiltration in the infarct was unchanged in LM11A-31-treated mice, indicating that LM11A-31 does not alter the chronic inflammatory response to stroke at the site of the infarct. However, LM11A-31-treated mice had less brain atrophy, neurodegeneration, tau pathology, and microglial activation in other regions of the ipsilateral hemisphere. These findings correlated with improved recovery of motor function on a ladder test, improved sensorimotor and cognitive abilities on a nest construction test, and less impulsivity in an open field test. These data support small molecule modulation of the p75NTR for preserving neuronal health and function during stroke recovery. SIGNIFICANCE STATEMENT: The findings from this study introduce the p75 neurotrophin receptor as a novel small molecule target for promotion of stroke recovery. Given that LM11A-31 is in clinical trials as a potential therapy for Alzheimer's disease, it could be considered as a candidate for assessment in stroke or vascular dementia studies.
Collapse
Affiliation(s)
- Thuy-Vi V Nguyen
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Rachel H Crumpacker
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Kylie E Calderon
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Frankie G Garcia
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Jacob C Zbesko
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Jennifer B Frye
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Selena Gonzalez
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Danielle A Becktel
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Tao Yang
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Marco A Tavera-Garcia
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Helena W Morrison
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Rick G Schnellmann
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Frank M Longo
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Kristian P Doyle
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| |
Collapse
|
12
|
Jaskiw GE, Xu D, Obrenovich ME, Donskey CJ. Small phenolic and indolic gut-dependent molecules in the primate central nervous system: levels vs. bioactivity. Metabolomics 2022; 18:8. [PMID: 34989922 DOI: 10.1007/s11306-021-01866-4] [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: 04/26/2021] [Accepted: 12/12/2021] [Indexed: 10/19/2022]
Abstract
INTRODUCTION A rapidly growing body of data documents associations between disease of the brain and small molecules generated by gut-microbiota (GMB). While such metabolites can affect brain function through a variety of mechanisms, the most direct action would be on the central nervous system (CNS) itself. OBJECTIVE Identify indolic and phenolic GMB-dependent small molecules that reach bioactive concentrations in primate CNS. METHODS We conducted a PubMed search for metabolomic studies of the primate CNS [brain tissue or cerebrospinal fluid (CSF)] and then selected for phenolic or indolic metabolites that (i) had been quantified, (ii) were GMB-dependent. For each chemical we then conducted a search for studies of bioactivity conducted in vitro in human cells of any kind or in CNS cells from the mouse or rat. RESULTS 36 metabolites of interests were identified in primate CNS through targeted metabolomics. Quantification was available for 31/36 and in vitro bioactivity for 23/36. The reported CNS range for 8 metabolites 2-(3-hydroxyphenyl)acetic acid, 2-(4-hydroxyphenyl)acetic acid, 3-(3-hydroxyphenyl)propanoic acid, (E)-3-(3,4-dihydroxyphenyl)prop-2-enoic acid [caffeic acid], 3-hydroxybenzoic acid, 4-hydroxybenzoic acid, 2-acetamido-3-(1H-indol-3-yl)propanoic acid [N-acetyltryptophan], 1H-indol-3-yl hydrogen sulfate [indoxyl-3-sulfate] overlapped with a bioactive concentration. However, the number and quality of relevant studies of CNS neurochemistry as well as of bioactivity were highly limited. Structural isomers, multiple metabolites and potential confounders were inadequately considered. CONCLUSION The potential direct bioactivity of GMB-derived indolic and phenolic molecules on primate CNS remains largely unknown. The field requires additional strategies to identify and prioritize screening of the most promising small molecules that enter the CNS.
Collapse
Affiliation(s)
- George E Jaskiw
- Psychiatry Service 116(A), Veterans Affairs Northeast Ohio Healthcare System (VANEOHS), 10701 East Blvd., Cleveland, OH, 44106, USA.
- School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| | - Dongyan Xu
- Psychiatry Service 116(A), Veterans Affairs Northeast Ohio Healthcare System (VANEOHS), 10701 East Blvd., Cleveland, OH, 44106, USA
- School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Mark E Obrenovich
- Pathology and Laboratory Medicine Service, VANEOHS, Cleveland, OH, USA
- Research Service, VANEOHS, Cleveland, OH, USA
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, USA
| | - Curtis J Donskey
- School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Geriatric Research, Education and Clinical Center (GRECC), VANEOHS, Cleveland, OH, USA
| |
Collapse
|
13
|
Vallianatou T, Bèchet NB, Correia MSP, Lundgaard I, Globisch D. Regional Brain Analysis of Modified Amino Acids and Dipeptides during the Sleep/Wake Cycle. Metabolites 2021; 12:metabo12010021. [PMID: 35050142 PMCID: PMC8780251 DOI: 10.3390/metabo12010021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/19/2021] [Accepted: 12/22/2021] [Indexed: 12/18/2022] Open
Abstract
Sleep is a state in which important restorative and anabolic processes occur. Understanding changes of these metabolic processes during the circadian rhythm in the brain is crucial to elucidate neurophysiological mechanisms important for sleep function. Investigation of amino acid modifications and dipeptides has recently emerged as a valuable approach in the metabolic profiling of the central nervous system. Nonetheless, very little is known about the effects of sleep on the brain levels of amino acid analogues. In the present study, we examined brain regional sleep-induced alterations selective for modified amino acids and dipeptides using Ultra-high performance liquid chromatography-MS/MS (UHPLC-MS/MS) based metabolomics. Our approach enabled the detection and identification of numerous amino acid-containing metabolites in the cortex, the hippocampus, the midbrain, and the cerebellum. In particular, analogues of the aromatic amino acids phenylalanine, tyrosine and tryptophan were significantly altered during sleep in the investigated brain regions. Cortical levels of medium and long chain N-acyl glycines were higher during sleep. Regional specific changes were also detected, especially related to tyrosine analogues in the hippocampus and the cerebellum. Our findings demonstrate a strong correlation between circadian rhythms and amino acid metabolism specific for different brain regions that provide previously unknown insights in brain metabolism.
Collapse
Affiliation(s)
- Theodosia Vallianatou
- Science for Life Laboratory, Department of Chemistry-BMC, Uppsala University, Box 599, SE-75124 Uppsala, Sweden; (T.V.); (M.S.P.C.)
| | - Nicholas B. Bèchet
- Department of Experimental Medical Science, Lund University, SE-22362 Lund, Sweden; (N.B.B.); (I.L.)
- Wallenberg Centre for Molecular Medicine, Lund University, SE-22362 Lund, Sweden
| | - Mario S. P. Correia
- Science for Life Laboratory, Department of Chemistry-BMC, Uppsala University, Box 599, SE-75124 Uppsala, Sweden; (T.V.); (M.S.P.C.)
| | - Iben Lundgaard
- Department of Experimental Medical Science, Lund University, SE-22362 Lund, Sweden; (N.B.B.); (I.L.)
- Wallenberg Centre for Molecular Medicine, Lund University, SE-22362 Lund, Sweden
| | - Daniel Globisch
- Science for Life Laboratory, Department of Chemistry-BMC, Uppsala University, Box 599, SE-75124 Uppsala, Sweden; (T.V.); (M.S.P.C.)
- Correspondence:
| |
Collapse
|
14
|
Vallianatou T, Lin W, Bèchet NB, Correia MSP, Shanbhag NC, Lundgaard I, Globisch D. Differential regulation of oxidative stress, microbiota-derived, and energy metabolites in the mouse brain during sleep. J Cereb Blood Flow Metab 2021; 41:3324-3338. [PMID: 34293940 PMCID: PMC8669215 DOI: 10.1177/0271678x211033358] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/25/2021] [Accepted: 06/23/2021] [Indexed: 11/17/2022]
Abstract
Sleep has evolved as a universal core function to allow for restorative biological processes. Detailed knowledge of metabolic changes necessary for the sleep state in the brain is missing. Herein, we have performed an in-depth metabolic analysis of four mouse brain regions and uncovered region-specific circadian variations. Metabolites linked to oxidative stress were altered during sleep including acylcarnitines, hydroxylated fatty acids, phenolic compounds, and thiol-containing metabolites. These findings provide molecular evidence of a significant metabolic shift of the brain energy metabolism. Specific alterations were observed for brain metabolites that have previously not been associated with a circadian function including the microbiome-derived metabolite ergothioneine that suggests a regulatory function. The pseudopeptide β-citryl-glutamate has been linked to brain development and we have now discovered a previously unknown regioisomer. These metabolites altered by the circadian rhythm represent the foundation for hypothesis-driven studies of the underlying metabolic processes and their function.
Collapse
Affiliation(s)
- Theodosia Vallianatou
- Department of Chemistry-BMC, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Weifeng Lin
- Department of Chemistry-BMC, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Nicholas B Bèchet
- Department of Experimental Medical Science, Lund University, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund University, Lund, Sweden
| | - Mario SP Correia
- Department of Chemistry-BMC, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Nagesh C Shanbhag
- Department of Experimental Medical Science, Lund University, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund University, Lund, Sweden
| | - Iben Lundgaard
- Department of Experimental Medical Science, Lund University, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund University, Lund, Sweden
| | - Daniel Globisch
- Department of Chemistry-BMC, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
15
|
Iliou A, Vlaikou AM, Nussbaumer M, Benaki D, Mikros E, Gikas E, Filiou MD. Exploring the metabolomic profile of cerebellum after exposure to acute stress. Stress 2021; 24:952-964. [PMID: 34553679 DOI: 10.1080/10253890.2021.1973997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Psychological stress and stress-related disorders constitute a major health problem in modern societies. Although the brain circuits involved in emotional processing are intensively studied, little is known about the implication of cerebellum in stress responses whereas the molecular changes induced by stress exposure in cerebellum remain largely unexplored. Here, we investigated the effects of acute stress exposure on mouse cerebellum. We used a forced swim test (FST) paradigm as an acute stressor. We then analyzed the cerebellar metabolomic profiles of stressed (n = 11) versus control (n = 11) male CD1 mice by a Nuclear Magnetic Resonance (NMR)-based, untargeted metabolomics approach. Our results showed altered levels of 19 out of the 47 annotated metabolites, which are implicated in neurotransmission and N-acetylaspartic acid (NAA) turnover, as well as in energy and purine/pyrimidine metabolism. We also correlated individual metabolite levels with FST behavioral parameters, and reported associations between FST readouts and levels of 4 metabolites. This work indicates an altered metabolomic signature after acute stress in the cerebellum and highlights a previously unexplored involvement of cerebellum in stress responses.
Collapse
Affiliation(s)
- Aikaterini Iliou
- Department of Pharmacy, Section of Pharmaceutical Chemistry, School of Health Sciences, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Angeliki-Maria Vlaikou
- Department of Biological Applications and Technology, Laboratory of Biochemistry, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Biomedical Research Division, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| | - Markus Nussbaumer
- Department of Biological Applications and Technology, Laboratory of Biochemistry, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Biomedical Research Division, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| | - Dimitra Benaki
- Department of Pharmacy, Section of Pharmaceutical Chemistry, School of Health Sciences, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Emmanuel Mikros
- Department of Pharmacy, Section of Pharmaceutical Chemistry, School of Health Sciences, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Evangelos Gikas
- Department of Pharmacy, Section of Pharmaceutical Chemistry, School of Health Sciences, National and Kapodistrian University of Athens (NKUA), Athens, Greece
- Department of Chemistry, Section of Analytical Chemistry, School of Science, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Michaela D Filiou
- Department of Biological Applications and Technology, Laboratory of Biochemistry, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Biomedical Research Division, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| |
Collapse
|
16
|
Ding J, Ji J, Rabow Z, Shen T, Folz J, Brydges CR, Fan S, Lu X, Mehta S, Showalter MR, Zhang Y, Araiza R, Bower LR, Lloyd KCK, Fiehn O. A metabolome atlas of the aging mouse brain. Nat Commun 2021; 12:6021. [PMID: 34654818 PMCID: PMC8519999 DOI: 10.1038/s41467-021-26310-y] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 09/24/2021] [Indexed: 12/30/2022] Open
Abstract
The mammalian brain relies on neurochemistry to fulfill its functions. Yet, the complexity of the brain metabolome and its changes during diseases or aging remain poorly understood. Here, we generate a metabolome atlas of the aging wildtype mouse brain from 10 anatomical regions spanning from adolescence to old age. We combine data from three assays and structurally annotate 1,547 metabolites. Almost all metabolites significantly differ between brain regions or age groups, but not by sex. A shift in sphingolipid patterns during aging related to myelin remodeling is accompanied by large changes in other metabolic pathways. Functionally related brain regions (brain stem, cerebrum and cerebellum) are also metabolically similar. In cerebrum, metabolic correlations markedly weaken between adolescence and adulthood, whereas at old age, cross-region correlation patterns reflect decreased brain segregation. We show that metabolic changes can be mapped to existing gene and protein brain atlases. The brain metabolome atlas is publicly available ( https://mouse.atlas.metabolomics.us/ ) and serves as a foundation dataset for future metabolomic studies.
Collapse
Affiliation(s)
- Jun Ding
- West Coast Metabolomics Center, UC Davis Genome Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA, 95616, USA
- Department of Chemistry, Wuhan University, 430072, Wuhan, Hubei, P.R. China
| | - Jian Ji
- School of Food Science, State Key Laboratory of Food Science and Technology, National Engineering Research Center for Functional Foods, Synergetic Innovation Center of Food Safety and Nutrition, Jiangnan University, 214122, Wuxi, Jiangsu, P.R. China
| | - Zachary Rabow
- West Coast Metabolomics Center, UC Davis Genome Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA, 95616, USA
| | - Tong Shen
- West Coast Metabolomics Center, UC Davis Genome Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA, 95616, USA
| | - Jacob Folz
- West Coast Metabolomics Center, UC Davis Genome Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA, 95616, USA
| | - Christopher R Brydges
- West Coast Metabolomics Center, UC Davis Genome Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA, 95616, USA
| | - Sili Fan
- West Coast Metabolomics Center, UC Davis Genome Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA, 95616, USA
| | - Xinchen Lu
- West Coast Metabolomics Center, UC Davis Genome Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA, 95616, USA
| | - Sajjan Mehta
- West Coast Metabolomics Center, UC Davis Genome Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA, 95616, USA
| | - Megan R Showalter
- West Coast Metabolomics Center, UC Davis Genome Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA, 95616, USA
| | - Ying Zhang
- West Coast Metabolomics Center, UC Davis Genome Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA, 95616, USA
| | - Renee Araiza
- Mouse Biology Program, and Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, 95618, USA
| | - Lynette R Bower
- Mouse Biology Program, and Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, 95618, USA
| | - K C Kent Lloyd
- Mouse Biology Program, and Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, 95618, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, UC Davis Genome Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA, 95616, USA.
| |
Collapse
|
17
|
Multiblock metabolomics: An approach to elucidate whole-body metabolism with multiblock principal component analysis. Comput Struct Biotechnol J 2021; 19:1956-1965. [PMID: 33995897 PMCID: PMC8086023 DOI: 10.1016/j.csbj.2021.04.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/20/2021] [Accepted: 04/04/2021] [Indexed: 12/16/2022] Open
Abstract
“Multiblock metabolomics” elucidates the global metabolic network in a whole body. “Multiblock metabolomics” combines LC/MS-based metabolomics with multiblock PCA. “Multiblock metabolomics” highlights and elicits organ-specific metabolism. TGs with less unsaturated fatty acids were highly accumulated in the diabetic liver.
Principal component analysis (PCA) is a useful tool for omics analysis to identify underlying factors and visualize relationships between biomarkers. However, this approach is limited in addressing life complexity and further improvement is required. This study aimed to develop a new approach that combines mass spectrometry-based metabolomics with multiblock PCA to elucidate the whole-body global metabolic network, thereby generating comparable metabolite maps to clarify the metabolic relationships among several organs. To evaluate the newly developed method, Zucker diabetic fatty (ZDF) rats (n = 6) were used as type 2 diabetic models and Sprague Dawley (SD) rats (n = 6) as controls. Metabolites in the heart, kidney, and liver were analyzed by capillary electrophoresis and liquid chromatography mass spectrometry, respectively, and the detected metabolites were analyzed by multiblock PCA. More than 300 metabolites were detected in the heart, kidney, and liver. When the metabolites obtained from the three organs were analyzed with multiblock PCA, the score and loading maps obtained were highly synchronized and their metabolism patterns were visually comparable. A significant finding in this study was the different expression patterns in lipid metabolism among the three organs; notably triacylglycerols with polyunsaturated fatty acids or less unsaturated fatty acids showed specific accumulation patterns depending on the organs.
Collapse
Key Words
- AMP, adenosine monophosphate
- Biomarkers
- CE/MS, capillary electrophoresis mass spectrometry
- CV, coefficient of variation
- ESI, electrospray ionization
- FABP, fatty acid-binding protein
- GC/MS, gas chromatography mass spectrometry
- LC/MS, liquid chromatography mass spectrometry
- Mass spectrometry
- Metabolomics
- Multiblock PCA
- PCA, principal component analysis
- PPAR, peroxisome proliferator-activated receptor
- QC, quality control
- SD, Sprague Dawley
- TCA, tricarboxylic acid. CoA, coenzyme A
- TG, triacylglycerol
- Type 2 Diabetes
- UPLC, ultra-performance liquid chromatography
- ZDF, Zucker diabetic fatty
Collapse
|
18
|
Dienel GA. Stop the rot. Enzyme inactivation at brain harvest prevents artifacts: A guide for preservation of the in vivo concentrations of brain constituents. J Neurochem 2021; 158:1007-1031. [PMID: 33636013 DOI: 10.1111/jnc.15293] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/30/2020] [Accepted: 01/05/2021] [Indexed: 12/25/2022]
Abstract
Post-mortem metabolism is widely recognized to cause rapid and prolonged changes in the concentrations of multiple classes of compounds in brain, that is, they are labile. Post-mortem changes from levels in living brain include components of pathways of metabolism of glucose and energy compounds, amino acids, lipids, signaling molecules, neuropeptides, phosphoproteins, and proteins. Methods that stop enzyme activity at brain harvest were developed almost 50 years ago and have been extensively used in studies of brain functions and diseases. Unfortunately, these methods are not commonly used to harvest brain tissue for mass spectrometry-based metabolomic studies or for imaging mass spectrometry studies (IMS, also called mass spectrometry imaging, MSI, or matrix-assisted laser desorption/ionization-MSI, MALDI-MSI). Instead these studies commonly kill animals, decapitate, dissect out brain and regions of interest if needed, then 'snap' freeze the tissue to stop enzymatic activity after harvest, with post-mortem intervals typically ranging from ~0.5 to 3 min. To increase awareness of the importance of stopping metabolism at harvest and preventing the unnecessary complications of not doing so, this commentary provides examples of labile metabolites and the magnitudes of their post-mortem changes in concentrations during brain harvest. Brain harvest methods that stop metabolism at harvest eliminate post-mortem enzymatic activities and can improve characterization of normal and diseased brain. In addition, metabolomic studies would be improved by reporting absolute units of concentration along with normalized peak areas or fold changes. Then reported values can be evaluated and compared with the extensive neurochemical literature to help prevent reporting of artifactual data.
Collapse
Affiliation(s)
- Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
19
|
Willacey CC, Karu N, Harms AC, Hankemeier T. Metabolic profiling of material-limited cell samples by dimethylaminophenacyl bromide derivatization with UPLC-MS/MS analysis. Microchem J 2020. [DOI: 10.1016/j.microc.2020.105445] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
20
|
Czarnecka AM, Milewski K, Albrecht J, Zielińska M. The Status of Bile Acids and Farnesoid X Receptor in Brain and Liver of Rats with Thioacetamide-Induced Acute Liver Failure. Int J Mol Sci 2020; 21:ijms21207750. [PMID: 33092050 PMCID: PMC7589210 DOI: 10.3390/ijms21207750] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 12/14/2022] Open
Abstract
Acute liver failure (ALF) leads to neurological symptoms defined as hepatic encephalopathy (HE). Although accumulation of ammonia and neuroinflammation are generally accepted as main contributors to HE pathomechanism, a buildup of bile acids (BA) in the blood is a frequent component of liver injury in HE patients. Recent studies have identified the nuclear farnesoid X receptor (FXR) acting via small heterodimer partner (SHP) as a mediator of BA-induced effects in the brain of ALF animals. The present study investigated the status of the BA–FXR axis in the brain and the liver, including selective changes in pertinent genes in thioacetamide (TAA)-induced ALF in Sprague–Dawley rats. FXR was found in rat neurons, confirming earlier reports for mouse and human brain. BA accumulated in blood but not in the brain tissue. Expression of mRNAs coding for Fxr and Shp was reduced in the hippocampus and of Fxr mRNA also in the cerebellum. Changes in Fxr mRNA levels were not followed by changes in FXR protein. The results leave open the possibility that mobilization of the BA–FXR axis in the brain may not be necessarily pathognomonic to HE but may depend upon ALF-related confounding factors.
Collapse
Affiliation(s)
- Anna Maria Czarnecka
- Correspondence: (A.M.C.); (M.Z.); Tel.: +48-22-6086416 (A.M.C.); +48-22-6086470 (M.Z.)
| | | | | | - Magdalena Zielińska
- Correspondence: (A.M.C.); (M.Z.); Tel.: +48-22-6086416 (A.M.C.); +48-22-6086470 (M.Z.)
| |
Collapse
|
21
|
Metabolomic and Imaging Mass Spectrometric Assays of Labile Brain Metabolites: Critical Importance of Brain Harvest Procedures. Neurochem Res 2020; 45:2586-2606. [PMID: 32949339 DOI: 10.1007/s11064-020-03124-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/29/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022]
Abstract
Metabolomic technologies including imaging mass spectrometry (IMS; also called mass spectrometry imaging, MSI, or matrix-assisted laser desorption/ionization-mass spectrometry imaging, MALDI MSI) are important methods to evaluate levels of many compounds in brain with high spatial resolution, characterize metabolic phenotypes of brain disorders, and identify disease biomarkers. ATP is central to brain energetics, and reports of its heterogeneous distribution in brain and regional differences in ATP/ADP ratios reported in IMS studies conflict with earlier studies. These discordant data were, therefore, analyzed and compared with biochemical literature that used rigorous methods to preserve labile metabolites. Unequal, very low regional ATP levels and low ATP/ADP ratios are explained by rapid metabolism during postmortem ischemia. A critical aspect of any analysis of brain components is their stability during and after tissue harvest so measured concentrations closely approximate their physiological levels in vivo. Unfortunately, the requirement for inactivation of brain enzymes by freezing or heating is not widely recognized outside the neurochemistry discipline, and procedures that do not prevent postmortem autolysis, including decapitation, brain removal/dissection, and 'snap freezing' are commonly used. Strong emphasis is placed on use of supplementary approaches to calibrate metabolite abundance in units of concentration in IMS studies and comparison of IMS results with biochemical data obtained by different methods to help identify potential artifacts.
Collapse
|
22
|
Kaushik G, Gupta K, Harms V, Torr E, Evans J, Johnson HJ, Soref C, Acevedo-Acevedo S, Antosiewicz-Bourget J, Mamott D, Uhl P, Johnson BP, Palecek SP, Beebe DJ, Thomson JA, Daly WT, Murphy WL. Engineered Perineural Vascular Plexus for Modeling Developmental Toxicity. Adv Healthc Mater 2020; 9:e2000825. [PMID: 32613760 PMCID: PMC8016604 DOI: 10.1002/adhm.202000825] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Indexed: 12/19/2022]
Abstract
There is a vital need to develop in vitro models of the developing human brain to recapitulate the biological effects that toxic compounds have on the brain. To model perineural vascular plexus (PNVP) in vitro, which is a key stage in embryonic development, human embryonic stem cells (hESC)-derived endothelial cells (ECs), neural progenitor cells, and microglia (MG) with primary pericytes (PCs) in synthetic hydrogels in a custom-designed microfluidics device are cocultured. The formation of a vascular plexus that includes networks of ECs (CD31+, VE-cadherin+), MG (IBA1+), and PCs (PDGFRβ+), and an overlying neuronal layer that includes differentiated neuronal cells (βIII Tubulin+, GFAP+) and radial glia (Nestin+, Notch2NL+), are characterized. Increased brain-derived neurotrophic factor secretion and differential metabolite secretion by the vascular plexus and the neuronal cells over time are consistent with PNVP functionality. Multiple concentrations of developmental toxicants (teratogens, microglial disruptor, and vascular network disruptors) significantly reduce the migration of ECs and MG toward the neuronal layer, inhibit formation of the vascular network, and decrease vascular endothelial growth factor A (VEGFA) secretion. By quantifying 3D cell migration, metabolic activity, vascular network disruption, and cytotoxicity, the PNVP model may be a useful tool to make physiologically relevant predictions of developmental toxicity.
Collapse
Affiliation(s)
- Gaurav Kaushik
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Ave., WIMR 5418, Madison, WI 53705, USA
| | - Kartik Gupta
- Department of Surgery, University of Wisconsin-Madison, 1111 Highland Ave., Madison, WI 53705, USA
| | - Victoria Harms
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Ave., WIMR 5418, Madison, WI 53705, USA
| | - Elizabeth Torr
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Ave., WIMR 5418, Madison, WI 53705, USA
| | - Jonathan Evans
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI 53706, USA
| | - Hunter J. Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI 53706, USA
| | - Cheryl Soref
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Ave., WIMR 5418, Madison, WI 53705, USA
| | - Suehelay Acevedo-Acevedo
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI 53706, USA
| | | | - Daniel Mamott
- Morgridge Institute for Research, 330 N Orchard St, Madison, WI 53715, USA
| | - Peyton Uhl
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Ave., WIMR 5418, Madison, WI 53705, USA
| | - Brian P. Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI 53706, USA
| | - Sean P. Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI 53706, USA
| | - David J. Beebe
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 1685 Highland Ave., Madison, WI 53705, USA, University of Wisconsin Carbone Center Research, 600 Highland Ave., Madison, WI 53792, USA, Department of Biomedical Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI 53706, USA
| | - James A. Thomson
- Morgridge Institute for Research, 330 N Orchard St, Madison, WI 53715, USA
| | - William T. Daly
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Ave., WIMR 5418, Madison, WI 53705, USA
| | - William L. Murphy
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Ave., WIMR 5418, Madison, WI 53705, USA, Department of Biomedical Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI 53706, USA
| |
Collapse
|
23
|
Lucassen PJ, Fitzsimons CP, Salta E, Maletic-Savatic M. Adult neurogenesis, human after all (again): Classic, optimized, and future approaches. Behav Brain Res 2020; 381:112458. [DOI: 10.1016/j.bbr.2019.112458] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 11/29/2019] [Accepted: 12/28/2019] [Indexed: 02/08/2023]
|
24
|
Teav T, Gallart-Ayala H, van der Velpen V, Mehl F, Henry H, Ivanisevic J. Merged Targeted Quantification and Untargeted Profiling for Comprehensive Assessment of Acylcarnitine and Amino Acid Metabolism. Anal Chem 2019; 91:11757-11769. [DOI: 10.1021/acs.analchem.9b02373] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Tony Teav
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, 1005 Lausanne, Switzerland
| | - Héctor Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, 1005 Lausanne, Switzerland
| | - Vera van der Velpen
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, 1005 Lausanne, Switzerland
| | - Florence Mehl
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, 1005 Lausanne, Switzerland
- Vital-IT−Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Hugues Henry
- Innovation and Development Laboratory, Clinical Chemistry Service, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, 1005 Lausanne, Switzerland
| |
Collapse
|
25
|
Chen K, Baluya D, Tosun M, Li F, Maletic-Savatic M. Imaging Mass Spectrometry: A New Tool to Assess Molecular Underpinnings of Neurodegeneration. Metabolites 2019; 9:E135. [PMID: 31295847 PMCID: PMC6681116 DOI: 10.3390/metabo9070135] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/19/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases are prevalent and devastating. While extensive research has been done over the past decades, we are still far from comprehensively understanding what causes neurodegeneration and how we can prevent it or reverse it. Recently, systems biology approaches have led to a holistic examination of the interactions between genome, metabolome, and the environment, in order to shed new light on neurodegenerative pathogenesis. One of the new technologies that has emerged to facilitate such studies is imaging mass spectrometry (IMS). With its ability to map a wide range of small molecules with high spatial resolution, coupled with the ability to quantify them at once, without the need for a priori labeling, IMS has taken center stage in current research efforts in elucidating the role of the metabolome in driving neurodegeneration. IMS has already proven to be effective in investigating the lipidome and the proteome of various neurodegenerative diseases, such as Alzheimer's, Parkinson's, Huntington's, multiple sclerosis, and amyotrophic lateral sclerosis. Here, we review the IMS platform for capturing biological snapshots of the metabolic state to shed more light on the molecular mechanisms of the diseased brain.
Collapse
Affiliation(s)
- Kevin Chen
- Department of Biosciences, Rice University, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Dodge Baluya
- Chemical Imaging Research Core at MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Mehmet Tosun
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Feng Li
- Center for Drug Discovery and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mirjana Maletic-Savatic
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA.
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA.
- Department of Neuroscience and Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
26
|
Wang K, Liu X, Guo Y, Wu Z, Zhi D, Ruan J, Zhao Z. The International Conference on Intelligent Biology and Medicine (ICIBM) 2018: systems biology on diverse data types. BMC SYSTEMS BIOLOGY 2018; 12:125. [PMID: 30577731 PMCID: PMC6302362 DOI: 10.1186/s12918-018-0648-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Between June 10–12, 2018, the International Conference on Intelligent Biology and Medicine (ICIBM 2018) was held in Los Angeles, California, USA. The conference included 11 scientific sessions, four tutorials, one poster session, four keynote talks and four eminent scholar talks that covered a wide range of topics in 3D genome structure analysis and visualization, next generation sequencing analysis, computational drug discovery, medical informatics, cancer genomics and systems biology. Systems biology has been a main theme in ICIBM 2018, with exciting advances presented in many areas of systems biology, covering various different data types such as gene regulation, circular RNAs expression, single-cell RNA-Seq, inter-chromosomal interactions, metabolomics, proteomics and phosphoproteomics. Here, we describe ten high quality papers to be published in BMC Systems Biology.
Collapse
Affiliation(s)
- Kai Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA. .,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Xiaoming Liu
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,College of Public Health, University of South Florida, Tampa, FL, 33612, USA
| | - Yan Guo
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Zhijin Wu
- Department of Biostatistics, Brown University, Providence, RI, 02912, USA
| | - Degui Zhi
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jianhua Ruan
- Department of Computer Science, The University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|