1
|
Li M, Qiu J, Yan G, Zheng X, Li A. How does the neurotoxin β-N-methylamino-L-alanine exist in biological matrices and cause toxicity? THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 922:171255. [PMID: 38417517 DOI: 10.1016/j.scitotenv.2024.171255] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/01/2024]
Abstract
The neurotoxin β-N-methylamino-L-alanine (BMAA) has been deemed as a risk factor for some neurodegenerative diseases such as amyotrophic lateral sclerosis/parkinsonism dementia complex (ALS/PDC). This possible link has been proved in some primate models and cell cultures with the appearance that BMAA exposure can cause excitotoxicity, formation of protein aggregates, and/or oxidative stress. The neurotoxin BMAA extensively exists in the environment and can be transferred through the food web to human beings. In this review, the occurrence, toxicological mechanisms, and characteristics of BMAA were comprehensively summarized, and proteins and peptides were speculated as its possible binding substances in biological matrices. It is difficult to compare the published data from previous studies due to the inconsistent analytical methods and components of BMAA. The binding characteristics of BMAA should be focused on to improve our understanding of its health risk to human health in the future.
Collapse
Affiliation(s)
- Min Li
- College of Environmental Science and Engineering, Ocean University of China, Qingdao 266100, China
| | - Jiangbing Qiu
- College of Environmental Science and Engineering, Ocean University of China, Qingdao 266100, China; Key Laboratory of Marine Environment and Ecology, Ocean University of China, Ministry of Education, Qingdao 266100, China
| | - Guowang Yan
- College of Environmental Science and Engineering, Ocean University of China, Qingdao 266100, China
| | - Xianyao Zheng
- College of Environmental Science and Engineering, Ocean University of China, Qingdao 266100, China
| | - Aifeng Li
- College of Environmental Science and Engineering, Ocean University of China, Qingdao 266100, China; Key Laboratory of Marine Environment and Ecology, Ocean University of China, Ministry of Education, Qingdao 266100, China.
| |
Collapse
|
2
|
Koksharova OA, Safronova NA. Non-Proteinogenic Amino Acid β-N-Methylamino-L-Alanine (BMAA): Bioactivity and Ecological Significance. Toxins (Basel) 2022; 14:539. [PMID: 36006201 PMCID: PMC9414260 DOI: 10.3390/toxins14080539] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/28/2022] [Accepted: 08/04/2022] [Indexed: 11/21/2022] Open
Abstract
Research interest in a non-protein amino acid β-N-methylamino-L-alanine (BMAA) arose due to the discovery of a connection between exposure to BMAA and the occurrence of neurodegenerative diseases. Previous reviews on this topic either considered BMAA as a risk factor for neurodegenerative diseases or focused on the problems of detecting BMAA in various environmental samples. Our review is devoted to a wide range of fundamental biological problems related to BMAA, including the molecular mechanisms of biological activity of BMAA and the complex relationships between producers of BMAA and the environment in various natural ecosystems. At the beginning, we briefly recall the most important facts about the producers of BMAA (cyanobacteria, microalgae, and bacteria), the pathways of BMAA biosynthesis, and reliable methods of identification of BMAA. The main distinctive feature of our review is a detailed examination of the molecular mechanisms underlying the toxicity of BMAA to living cells. A brand new aspect, not previously discussed in any reviews, is the effect of BMAA on cyanobacterial cells. These recent studies, conducted using transcriptomics and proteomics, revealed potent regulatory effects of BMAA on the basic metabolism and cell development of these ancient photoautotrophic prokaryotes. Exogenous BMAA strongly influences cell differentiation and primary metabolic processes in cyanobacteria, such as nitrogen fixation, photosynthesis, carbon fixation, and various biosynthetic processes involving 2-oxoglutarate and glutamate. Cyanobacteria were found to be more sensitive to exogenous BMAA under nitrogen-limited growth conditions. We suggest a hypothesis that this toxic diaminoacid can be used by phytoplankton organisms as a possible allelopathic tool for controlling the population of cyanobacterial cells during a period of intense competition for nitrogen and other resources in various ecosystems.
Collapse
Affiliation(s)
- Olga A. Koksharova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, Kurchatov Square, 2, 123182 Moscow, Russia
| | - Nina A. Safronova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| |
Collapse
|
3
|
Deininger SO, Bollwein C, Casadonte R, Wandernoth P, Gonçalves JPL, Kriegsmann K, Kriegsmann M, Boskamp T, Kriegsmann J, Weichert W, Schirmacher P, Ly A, Schwamborn K. Multicenter Evaluation of Tissue Classification by Matrix-Assisted Laser Desorption/Ionization Mass Spectrometry Imaging. Anal Chem 2022; 94:8194-8201. [PMID: 35658398 DOI: 10.1021/acs.analchem.2c00097] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Many studies have demonstrated that tissue phenotyping (tissue typing) based on mass spectrometric imaging data is possible; however, comprehensive studies assessing variation and classifier transferability are largely lacking. This study evaluated the generalization of tissue classification based on Matrix Assisted Laser Desorption/Ionization (MALDI) mass spectrometric imaging (MSI) across measurements performed at different sites. Sections of a tissue microarray (TMA) consisting of different formalin-fixed and paraffin-embedded (FFPE) human tissue samples from different tumor entities (leiomyoma, seminoma, mantle cell lymphoma, melanoma, breast cancer, and squamous cell carcinoma of the lung) were prepared and measured by MALDI-MSI at different sites using a standard protocol (SOP). Technical variation was deliberately introduced on two separate measurements via a different sample preparation protocol and a MALDI Time of Flight mass spectrometer that was not tuned to optimal performance. Using standard data preprocessing, a classification accuracy of 91.4% per pixel was achieved for intrasite classifications. When applying a leave-one-site-out cross-validation strategy, accuracy per pixel over sites was 78.6% for the SOP-compliant data sets and as low as 36.1% for the mistuned instrument data set. Data preprocessing designed to remove technical variation while retaining biological information substantially increased classification accuracy for all data sets with SOP-compliant data sets improved to 94.3%. In particular, classification accuracy of the mistuned instrument data set improved to 81.3% and from 67.0% to 87.8% per pixel for the non-SOP-compliant data set. We demonstrate that MALDI-MSI-based tissue classification is possible across sites when applying histological annotation and an optimized data preprocessing pipeline to improve generalization of classifications over technical variation and increasing overall robustness.
Collapse
Affiliation(s)
| | - Christine Bollwein
- Institute of Pathology, School of Medicine, Technical University of Munich, Trogerstrasse 18, 81675 München, Germany
| | - Rita Casadonte
- Proteopath GmbH, Max-Planck-Strasse 17, 54296 Trier, Germany
| | - Petra Wandernoth
- MVZ für Histologie, Zytologie und molekulare Diagnostik Trier GmbH, Max-Planck-Strasse 5, 54296 Trier, Germany
| | | | - Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Tobias Boskamp
- Bruker Daltonics GmbH & Co KG, Fahrenheitstrasse 4, 28359 Bremen, Germany.,Center for Industrial Mathematics, University of Bremen, 28359 Bremen, Germany
| | - Jörg Kriegsmann
- MVZ für Histologie, Zytologie und molekulare Diagnostik Trier GmbH, Max-Planck-Strasse 5, 54296 Trier, Germany.,Danube Private University (DPU) Faculty of Medicine/Dentistry, Steiner Landstrasse 124, 3500 Krems-Stein, Austria
| | - Wilko Weichert
- Institute of Pathology, School of Medicine, Technical University of Munich, Trogerstrasse 18, 81675 München, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Alice Ly
- Bruker Daltonics GmbH & Co KG, Fahrenheitstrasse 4, 28359 Bremen, Germany
| | - Kristina Schwamborn
- Institute of Pathology, School of Medicine, Technical University of Munich, Trogerstrasse 18, 81675 München, Germany
| |
Collapse
|
4
|
Gois AM, Bispo JM, Lins LC, Medeiros KA, Souza MF, Santos ER, Santos JF, Ribeiro AM, Silva RH, Paixão MO, Leopoldino JF, Marchioro M, Santos JR, Mendonça DM. Motor behavioral abnormalities and histopathological findings in middle aged male Wistar rats inoculated with cerebrospinal fluid from patients with Amyotrophic Lateral Sclerosis. CURRENT RESEARCH IN BEHAVIORAL SCIENCES 2022. [DOI: 10.1016/j.crbeha.2022.100069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
5
|
Marine Neurotoxins' Effects on Environmental and Human Health: An OMICS Overview. Mar Drugs 2021; 20:md20010018. [PMID: 35049872 PMCID: PMC8778346 DOI: 10.3390/md20010018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/27/2022] Open
Abstract
Harmful algal blooms (HAB), and the consequent release of toxic metabolites, can be responsible for seafood poisoning outbreaks. Marine wildlife can accumulate these toxins throughout the food chain, which presents a threat to consumers’ health. Some of these toxins, such as saxitoxin (STX), domoic acid (DA), ciguatoxin (CTX), brevetoxin (BTX), tetrodotoxin (TTX), and β-N-methylamino-L-alanine (BMAA), cause severe neurological symptoms in humans. Considerable information is missing, however, notably the consequences of toxin exposures on changes in gene expression, protein profile, and metabolic pathways. This information could lead to understanding the consequence of marine neurotoxin exposure in aquatic organisms and humans. Nevertheless, recent contributions to the knowledge of neurotoxins arise from OMICS-based research, such as genomics, transcriptomics, proteomics, and metabolomics. This review presents a comprehensive overview of the most recent research and of the available solutions to explore OMICS datasets in order to identify new features in terms of ecotoxicology, food safety, and human health. In addition, future perspectives in OMICS studies are discussed.
Collapse
|
6
|
Lim J, Aguilan JT, Sellers RS, Nagajyothi F, Weiss LM, Angeletti RH, Bortnick AE. Lipid mass spectrometry imaging and proteomic analysis of severe aortic stenosis. J Mol Histol 2020; 51:559-571. [PMID: 32794037 DOI: 10.1007/s10735-020-09905-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 08/09/2020] [Indexed: 12/19/2022]
Abstract
Severe aortic stenosis (AS) is prevalent in adults ≥ 65 years, a significant cause of morbidity and mortality, with no medical therapy. Lipid and proteomic alterations of human AS tissue were determined using mass spectrometry imaging (MSI) and liquid chromatography electrospray ionization tandem mass spectrometry (LC-ESI-MS/MS) to understand histopathology, potential biomarkers of disease, and progression from non-calcified to calcified phenotype. A reproducible MSI method was developed using healthy murine aortic valves (n = 3) and subsequently applied to human AS (n = 2). Relative lipid levels were spatially mapped and associated with different microdomains. Proteomics for non-calcified and calcified microdomains were performed to ascertain differences in expression. Increased pro-osteogenic and inflammatory lysophosphatidylcholine (LPC) 16:0 and 18:0 were co-localized with calcified microdomains. Proteomics analysis identified differential patterns in calcified microdomains with high LPC and low cholesterol as compared to non-calcified microdomains with low LPC and high cholesterol. Calcified microdomains had higher levels of: apolipoproteins (Apo) B-100 (p < 0.001) and Apo A-IV (p < 0.001), complement C3 and C4-B (p < 0.001), C5 (p = 0.007), C8 beta chain (p = 0.013) and C9 (p = 0.010), antithrombotic proteins alpha-2-macroglobulin (p < 0.0001) and antithrombin III (p = 0.002), and higher anti-calcific fetuin-A (p = 0.02), while the osteoblast differentiating factor transgelin (p < 0.0001), extracellular matrix proteins versican, prolargin, and lumican ( p < 0.001) and regulator protein complement factor H (p < 0.001) were higher in non-calcified microdomains. A combined lipidomic and proteomic approach provided insight into factors potentially contributing to progression from non-calcified to calcific disease in severe AS. Additional studies of these candidates and protein networks could yield new targets for slowing progression of AS.
Collapse
Affiliation(s)
- Jihyeon Lim
- Janssen Research and Development, Malvern, PA, USA
| | - Jennifer T Aguilan
- Laboratory for Macromolecular Analysis & Proteomics, Bronx, NY, USA.,Department of Pathology, Montefiore Health System and Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Fnu Nagajyothi
- Department of Pathology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Louis M Weiss
- Laboratory for Macromolecular Analysis & Proteomics, Bronx, NY, USA
| | - Ruth Hogue Angeletti
- Laboratory for Macromolecular Analysis & Proteomics, Bronx, NY, USA.,Department of Biochemistry, Montefiore Health System and Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Developmental and Molecular Biology, Montefiore Health System and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anna E Bortnick
- Department of Medicine, Division of Cardiology, Montefiore Health System and Albert Einstein College of Medicine, Bronx, NY, USA. .,Department of Medicine, Division of Geriatrics, Montefiore Health System and Albert Einstein College of Medicine, Bronx, NY, USA. .,Jack D. Weiler Hospital, 1825 Eastchester Road, Suite 2S-46, Bronx, NY, 10461, USA.
| |
Collapse
|
7
|
The cyanobacterial neurotoxin β-N-methylamino-L-alanine (BMAA) targets the olfactory bulb region. Arch Toxicol 2020; 94:2799-2808. [PMID: 32435914 PMCID: PMC7395073 DOI: 10.1007/s00204-020-02775-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022]
Abstract
Olfactory dysfunction is implicated in neurodegenerative disorders and typically manifests years before other symptoms. The cyanobacterial neurotoxin β-N-methylamino-l-alanine (BMAA) is suggested as a risk factor for neurodegenerative disease. Detection of BMAA in air filters has increased the concern that aerosolization may lead to human BMAA exposure through the air. The aim of this study was to determine if BMAA targets the olfactory system. Autoradiographic imaging showed a distinct localization of radioactivity in the right olfactory mucosa and bulb following a unilateral intranasal instillation of 3H-BMAA (0.018 µg) in mice, demonstrating a direct transfer of BMAA via the olfactory pathways to the brain circumventing the blood–brain barrier, which was confirmed by liquid scintillation. Treatment of mouse primary olfactory bulb cells with 100 µM BMAA for 24 h caused a disruption of the neurite network, formation of dendritic varicosities and reduced cell viability. The NMDA receptor antagonist MK-801 and the metabotropic glutamate receptor antagonist MCPG protected against the BMAA-induced alterations, demonstrating the importance of glutamatergic mechanisms. The ionotropic non-NMDA receptor antagonist CNQX prevented the BMAA-induced decrease of cell viability in mixed cultures containing both neuronal and glial cells, but not in cultures with neurons only, suggesting a role of neuron–glial interactions and glial AMPA receptors in the BMAA-induced toxicity. The results show that the olfactory region may be a target for BMAA following inhalation exposure. Further studies on the relations between environmental olfactory toxicants and neurodegenerative disorders are warranted.
Collapse
|
8
|
Pierozan P, Karlsson O. Mitotically heritable effects of BMAA on striatal neural stem cell proliferation and differentiation. Cell Death Dis 2019; 10:478. [PMID: 31209203 PMCID: PMC6579766 DOI: 10.1038/s41419-019-1710-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/30/2019] [Accepted: 05/28/2019] [Indexed: 11/24/2022]
Abstract
The widespread environmental contaminant β-methylamino-L-alanine (BMAA) is a developmental neurotoxicant that can induce long-term learning and memory deficits. Studies have shown high transplacental transfer of 3H-BMAA and a significant uptake in fetal brain. Therefore, more information on how BMAA may influence growth and differentiation of neural stem cells is required for assessment of the risk to the developing brain. The aim of this study was to investigate direct and mitotically inherited effects of BMAA exposure using primary striatal neurons and embryonic neural stem cells. The neural stem cells were shown to be clearly more susceptible to BMAA exposure than primary neurons. Exposure to 250 µM BMAA reduced neural stem cell proliferation through apoptosis and G2/M arrest. At lower concentrations (50–100 µM), not affecting cell proliferation, BMAA reduced the differentiation of neural stem cells into astrocytes, oligodendrocytes, and neurons through glutamatergic mechanisms. Neurons that were derived from the BMAA-treated neuronal stem cells demonstrated morphological alterations including reduced neurite length, and decreased number of processes and branches per cell. Interestingly, the BMAA-induced changes were mitotically heritable to daughter cells. The results suggest that early-life exposure to BMAA impairs neuronal stem cell programming, which is vital for development of the nervous system and may result in long-term consequences predisposing for both neurodevelopmental disorders and neurodegenerative disease later in life. More attention should be given to the potential adverse effects of BMAA exposure on brain development.
Collapse
Affiliation(s)
- Paula Pierozan
- Science for Life Laboratory, Department of Environmental Sciences and Analytical Chemistry, Stockholm University, 114 18, Stockholm, Sweden.,Department of Pharmaceutical Biosciences, Uppsala University, Box 591, 751 24, Uppsala, Sweden
| | - Oskar Karlsson
- Science for Life Laboratory, Department of Environmental Sciences and Analytical Chemistry, Stockholm University, 114 18, Stockholm, Sweden. .,Department of Pharmaceutical Biosciences, Uppsala University, Box 591, 751 24, Uppsala, Sweden.
| |
Collapse
|
9
|
Optimization of MALDI-TOF mass spectrometry imaging for the visualization and comparison of peptide distributions in dry-cured ham muscle fibers. Food Chem 2019; 283:275-286. [DOI: 10.1016/j.foodchem.2018.12.126] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 12/29/2018] [Accepted: 12/29/2018] [Indexed: 01/02/2023]
|
10
|
Ucal Y, Coskun A, Ozpinar A. Quality will determine the future of mass spectrometry imaging in clinical laboratories: the need for standardization. Expert Rev Proteomics 2019; 16:521-532. [DOI: 10.1080/14789450.2019.1624165] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Yasemin Ucal
- School of Medicine, Department of Medical Biochemistry, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Abdurrahman Coskun
- School of Medicine, Department of Medical Biochemistry, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Aysel Ozpinar
- School of Medicine, Department of Medical Biochemistry, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| |
Collapse
|
11
|
Michno W, Wehrli PM, Blennow K, Zetterberg H, Hanrieder J. Molecular imaging mass spectrometry for probing protein dynamics in neurodegenerative disease pathology. J Neurochem 2018; 151:488-506. [PMID: 30040875 DOI: 10.1111/jnc.14559] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/03/2018] [Accepted: 07/12/2018] [Indexed: 12/14/2022]
Abstract
Recent advances in the understanding of basic pathological mechanisms in various neurological diseases depend directly on the development of novel bioanalytical technologies that allow sensitive and specific chemical imaging at high resolution in cells and tissues. Mass spectrometry-based molecular imaging (IMS) has gained increasing popularity in biomedical research for mapping the spatial distribution of molecular species in situ. The technology allows for comprehensive, untargeted delineation of in situ distribution profiles of metabolites, lipids, peptides and proteins. A major advantage of IMS over conventional histochemical techniques is its superior molecular specificity. Imaging mass spectrometry has therefore great potential for probing molecular regulations in CNS-derived tissues and cells for understanding neurodegenerative disease mechanism. The goal of this review is to familiarize the reader with the experimental workflow, instrumental developments and methodological challenges as well as to give a concise overview of the major advances and recent developments and applications of IMS-based protein and peptide profiling with particular focus on neurodegenerative diseases. This article is part of the Special Issue "Proteomics".
Collapse
Affiliation(s)
- Wojciech Michno
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Patrick M Wehrli
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK.,Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
12
|
Pierozan P, Andersson M, Brandt I, Karlsson O. The environmental neurotoxin β-N-methylamino-L-alanine inhibits melatonin synthesis in primary pinealocytes and a rat model. J Pineal Res 2018. [PMID: 29528516 DOI: 10.1111/jpi.12488] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The environmental neurotoxin β-N-methylamino-L-alanine (BMAA) is a glutamate receptor agonist that can induce oxidative stress and has been implicated as a possible risk factor for neurodegenerative disease. Detection of BMAA in mussels, crustaceans, and fish illustrates that the sources of human exposure to this toxin are more abundant than previously anticipated. The aim of this study was to determine uptake of BMAA in the pineal gland and subsequent effects on melatonin production in primary pinealocyte cultures and a rat model. Autoradiographic imaging of 10-day-old male rats revealed a high and selective uptake in the pineal gland at 30 minutes to 24 hours after 14 C-L-BMAA administration (0.68 mg/kg). Primary pinealocyte cultures exposed to 0.05-3 mmol/L BMAA showed a 57%-93% decrease in melatonin synthesis in vitro. Both the metabotropic glutamate receptor 3 (mGluR3) antagonist Ly341495 and the protein kinase C (PKC) activator phorbol-12-myristate-13-acetate prevented the decrease in melatonin secretion, suggesting that BMAA inhibits melatonin synthesis by mGluR3 activation and PKC inhibition. Serum analysis revealed a 45% decrease in melatonin concentration in neonatal rats assessed 2 weeks after BMAA administration (460 mg/kg) and confirmed an inhibition of melatonin synthesis in vivo. Given that melatonin is a most important neuroprotective molecule in the brain, the etiology of BMAA-induced neurodegeneration may include mechanisms beyond direct excitotoxicity and oxidative stress.
Collapse
Affiliation(s)
- Paula Pierozan
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Marie Andersson
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden
| | - Ingvar Brandt
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden
| | - Oskar Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
13
|
Andersson M, Karlsson O, Brandt I. The environmental neurotoxin β-N-methylamino-l-alanine (l-BMAA) is deposited into birds' eggs. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 147:720-724. [PMID: 28942274 DOI: 10.1016/j.ecoenv.2017.09.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/08/2017] [Accepted: 09/12/2017] [Indexed: 06/07/2023]
Abstract
The neurotoxic amino acid β-N-methylamino-L-alanine (BMAA) has been implicated in the etiology of neurodegenerative disorders. BMAA is also a known developmental neurotoxin and research indicates that the sources of human and wildlife exposure may be more diverse than previously anticipated. The aim of the present study was therefore to examine whether BMAA can be transferred into birds' eggs. Egg laying quail were dosed with 14C-labeled BMAA. The distribution of radioactivity in the birds and their laid eggs was then examined at different time points by autoradiography and phosphoimaging analysis. To evaluate the metabolic stability of the BMAA molecule, the distribution of 14C-methyl- and 14C-carboxyl-labeled BMAA were compared. The results revealed a pronounced incorporation of radioactivity in the eggs, predominantly in the yolk but also in the albumen. Imaging analysis showed that the concentrations of radioactivity in the liver decreased about seven times between the 24h and the 72h time points, while the concentrations in egg yolk remained largely unchanged. At 72h the egg yolk contained about five times the concentration of radioactivity in the liver. Both BMAA preparations gave rise to similar distribution pattern in the bird tissues and in the eggs, indicating metabolic stability of the labeled groups. The demonstrated deposition into eggs warrants studies of BMAAs effects on bird development. Moreover, birds' eggs may be a source of human BMAA exposure, provided that the laying birds are exposed to BMAA via their diet.
Collapse
Affiliation(s)
- Marie Andersson
- Department of Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Oskar Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, SE-751 24 Uppsala, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, SE-17176 Stockholm, Sweden
| | - Ingvar Brandt
- Department of Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden.
| |
Collapse
|
14
|
Laugeray A, Oummadi A, Jourdain C, Feat J, Meyer-Dilhet G, Menuet A, Plé K, Gay M, Routier S, Mortaud S, Guillemin GJ. Perinatal Exposure to the Cyanotoxin β-N-Méthylamino-L-Alanine (BMAA) Results in Long-Lasting Behavioral Changes in Offspring-Potential Involvement of DNA Damage and Oxidative Stress. Neurotox Res 2017; 33:87-112. [PMID: 28879461 DOI: 10.1007/s12640-017-9802-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/25/2017] [Accepted: 08/18/2017] [Indexed: 12/13/2022]
Abstract
We recently demonstrated that perinatal exposure to the glutamate-related herbicide, glufosinate ammonium, has deleterious effects on neural stem cell (NSC) homeostasis within the sub-ventricular zone (SVZ), probably leading to ASD-like symptoms in offspring later in life. In the present study, we aimed to investigate whether perinatal exposure to another glutamate-related toxicant, the cyanobacterial amino acid β-N-methylamino-L-alanine (BMAA), might also trigger neurodevelopmental disturbances. With this aim, female mice were intranasally exposed to low doses of BMAA, 50 mg kg-1 three times a week from embryonic days 7-10 to postnatal day 21. Behavioral analyses were performed during the offspring's early life and during adulthood. Developmental analyses revealed that perinatal exposure to BMAA hastened the appearance of some reflexes and communicative skills. BMAA-exposed offspring displayed sex-dependent changes in emotional cognition shortly after exposure. Later in life, the female offspring continued to express emotional defects and to display abnormal sociability, while males were less affected. To assess whether early exposure to BMAA had deleterious effects on NSC homeostasis, we exposed mice NSCs to 1 and 3 mM BMAA during 24 h. We found that BMAA-exposed NSCs produced high levels of ROS, highlighting the ability of BMAA to induce oxidative stress. We also showed that BMAA exposure increased the number of γH2AX/53BP1 foci per nucleus, suggesting that BMAA-induced DNA damage in NSCs. Collectively, this data strongly suggests that perinatal exposure to the cyanobacteria BMAA, even at low doses, results in neurobehavioral disturbances during both the postnatal period and adulthood. This is considered to be underpinned at the cellular level through dysregulation of NSC homeostasis in the developing brain.
Collapse
Affiliation(s)
- Anthony Laugeray
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355, Centre National de la Recherche Scientifique, 3b, rue de la Férollerie, 45071, Orléans, France. .,University of Orléans, Orléans, France.
| | | | | | - Justyne Feat
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355, Centre National de la Recherche Scientifique, 3b, rue de la Férollerie, 45071, Orléans, France
| | - Géraldine Meyer-Dilhet
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355, Centre National de la Recherche Scientifique, 3b, rue de la Férollerie, 45071, Orléans, France
| | - Arnaud Menuet
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355, Centre National de la Recherche Scientifique, 3b, rue de la Férollerie, 45071, Orléans, France.,University of Orléans, Orléans, France
| | - Karen Plé
- University of Orléans, Orléans, France.,Institute de Chimie Organique et Analytique, UMR 7311, Center National de la Recherche Scientifique, Orléans, France
| | - Marion Gay
- University of Orléans, Orléans, France.,Institute de Chimie Organique et Analytique, UMR 7311, Center National de la Recherche Scientifique, Orléans, France
| | - Sylvain Routier
- University of Orléans, Orléans, France.,Institute de Chimie Organique et Analytique, UMR 7311, Center National de la Recherche Scientifique, Orléans, France
| | - Stéphane Mortaud
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355, Centre National de la Recherche Scientifique, 3b, rue de la Férollerie, 45071, Orléans, France. .,University of Orléans, Orléans, France.
| | - Gilles J Guillemin
- Neuroinflammation Group, MND and Neurodegenerative Diseases Research Center, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
15
|
Chernoff N, Hill DJ, Diggs DL, Faison BD, Francis BM, Lang JR, Larue MM, Le TT, Loftin KA, Lugo JN, Schmid JE, Winnik WM. A critical review of the postulated role of the non-essential amino acid, β-N-methylamino-L-alanine, in neurodegenerative disease in humans. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2017; 20:1-47. [PMID: 28598725 PMCID: PMC6503681 DOI: 10.1080/10937404.2017.1297592] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
The compound BMAA (β-N-methylamino-L-alanine) has been postulated to play a significant role in four serious neurological human diseases: Amyotrophic Lateral Sclerosis/Parkinsonism Dementia Complex (ALS/PDC) found on Guam, and ALS, Parkinsonism, and dementia that occur globally. ALS/PDC with symptoms of all three diseases first came to the attention of the scientific community during and after World War II. It was initially associated with cycad flour used for food because BMAA is a product of symbiotic cycad root-dwelling cyanobacteria. Human consumption of flying foxes that fed on cycad seeds was later suggested as a source of BMAA on Guam and a cause of ALS/PDC. Subsequently, the hypothesis was expanded to include a causative role for BMAA in other neurodegenerative diseases including Alzheimer's disease (AD) through exposures attributed to proximity to freshwaters and/or consumption of seafood due to its purported production by most species of cyanobacteria. The hypothesis that BMAA is the critical factor in the genesis of these neurodegenerative diseases received considerable attention in the medical, scientific, and public arenas. This review examines the history of ALS/PDC and the BMAA-human disease hypotheses; similarities and differences between ALS/PDC and the other diseases with similar symptomologies; the relationship of ALS/PDC to other similar diseases, studies of BMAA-mediated effects in lab animals, inconsistencies and data gaps in the hypothesis; and other compounds and agents that were suggested as the cause of ALS/PDC on Guam. The review concludes that the hypothesis of a causal BMAA neurodegenerative disease relationship is not supported by existing data.
Collapse
Affiliation(s)
- N. Chernoff
- U.S. Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, NC, USA
| | - D. J. Hill
- U.S. Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, NC, USA
| | - D. L. Diggs
- Oak Ridge Institute for Science and Education Internship/Research Participation Program at the U.S. Environmental Protection Agency, NHEERL, Research Triangle Park, NC, USA
| | - B. D. Faison
- U.S. Environmental Protection Agency, Office of Water, Office of Science and Technology, Washington, DC, USA
| | - B. M. Francis
- Department of Entomology, University of Illinois, Champaign-Urbana, IL, USA
| | - J. R Lang
- Oak Ridge Institute for Science and Education Internship/Research Participation Program at the U.S. Environmental Protection Agency, NHEERL, Research Triangle Park, NC, USA
| | - M. M. Larue
- Oak Ridge Institute for Science and Education Internship/Research Participation Program at the U.S. Environmental Protection Agency, NHEERL, Research Triangle Park, NC, USA
| | - T.-T. Le
- Oak Ridge Institute for Science and Education Internship/Research Participation Program at the U.S. Environmental Protection Agency, NHEERL, Research Triangle Park, NC, USA
| | | | - J. N. Lugo
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, USA
| | - J. E. Schmid
- U.S. Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, NC, USA
| | - W. M. Winnik
- U.S. Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, NC, USA
| |
Collapse
|
16
|
Sui P, Watanabe H, Artemenko K, Sun W, Bakalkin G, Andersson M, Bergquist J. Neuropeptide imaging in rat spinal cord with MALDI-TOF MS: Method development for the application in pain-related disease studies. EUROPEAN JOURNAL OF MASS SPECTROMETRY (CHICHESTER, ENGLAND) 2017; 23:105-115. [PMID: 28657437 DOI: 10.1177/1469066717703272] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Spinal cord as a connection between brain and peripheral nervous system is an essential material for studying neural transmission, especially in pain-related research. This study was the first to investigate pain-related neuropeptide distribution in rat spinal cord using a matrix-assisted laser desorption ionization-time of flight imaging mass spectrometry (MALDI TOF MS) approach. The imaging workflow was evaluated and showed that MALDI TOF MS provides efficient resolution and robustness for neuropeptide imaging in rat spinal cord tissue. The imaging result showed that in naive rat spinal cord the molecular distribution of haeme, phosphatidylcholine, substance P and thymosin beta 4 were well in line with histological features. Three groups of pain-related neuropeptides, which are cleaved from prodynorphin, proenkephalin and protachykinin-1 proteins were detected. All these neuropeptides were found predominantly localized in the dorsal spinal cord and each group had unique distribution pattern. This study set the stage for future MALDI TOF MS application to elucidate signalling mechanism of pain-related diseases in small animal models.
Collapse
Affiliation(s)
- Ping Sui
- 1 Analytical Chemistry, Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden
| | - Hiroyuki Watanabe
- 2 Molecular Neuropsychopharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Konstantin Artemenko
- 1 Analytical Chemistry, Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden
| | - Wei Sun
- 2 Molecular Neuropsychopharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Georgy Bakalkin
- 2 Molecular Neuropsychopharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Malin Andersson
- 3 Drug Safety and Toxicology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Jonas Bergquist
- 1 Analytical Chemistry, Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden
| |
Collapse
|
17
|
Bláhová L, Kohoutek J, Kadlecová E, Kozáková L, Bláha L. Assessment of non-derivatized β-N-methylamino-l-alanine (BMAA) neurotoxin in free form in urine of patients with nonspecific neurological symptoms. Toxicon 2017; 133:48-57. [PMID: 28428069 DOI: 10.1016/j.toxicon.2017.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 04/13/2017] [Accepted: 04/16/2017] [Indexed: 12/12/2022]
Abstract
The beta-N-methylamino-l-alanine (BMAA) is a non-proteinogenic amino acid discussed to be produced by cyanobacteria forming harmful blooms. Since BMAA is suspected etiological agent in neurodegenerative diseases, there is a need to study and validate whether and in what concentrations can BMAA be present in human tissues. The aim of the present study was to validate analytical and extraction procedures for quantification of non-derivatized BMAA in the urine using liquid chromatography and commercial ELISA Kit. The study was focused on BMAA in different forms - dissolved, protein associated and total. The validated protocol included SPE followed by HILIC MS/MS for analyses of non-derivatized free form of BMAA with a limit of quantification 20 ng/mL. The methods for other BMAA forms (i.e. protein-associated and total) were also assessed but high matrix interferences did not allow their implementation. The method was used for analyses of free BMAA in 23 urine samples from healthy volunteers and psychiatric patients suffering from nonspecific neurological symptoms. Traces of BMAA were suspectedly detected in a single urine sample but they were not unequivocally proved according to all conservative analytical criteria. BMAA was also not confirmed in a repeatedly collected sample from the same person. The evaluated commercial BMAA ELISA Kit (Abraxis) was not suitable for determination of BMAA in extracted urine samples because of systematically highly false positive results. In agreement with recent findings, analyses of BMAA appear to methodologically challenging, and further research on BMAA in human tissues (or its precursors with potency to form BMAA under natural conditions or - eventually - during sample processing) is needed to clarify its potential ethiological role in neurodegenerative diseases.
Collapse
Affiliation(s)
- L Bláhová
- Masaryk University, Faculty of Science, RECETOX, Kamenice 753/5, Building A29, CZ62500 Brno, Czech Republic
| | - J Kohoutek
- Masaryk University, Faculty of Science, RECETOX, Kamenice 753/5, Building A29, CZ62500 Brno, Czech Republic
| | - E Kadlecová
- Psychiatric Hospital Písek, Vladislavova 490, CZ39701 Písek, Czech Republic
| | - L Kozáková
- Psychiatric Hospital Písek, Vladislavova 490, CZ39701 Písek, Czech Republic
| | - L Bláha
- Masaryk University, Faculty of Science, RECETOX, Kamenice 753/5, Building A29, CZ62500 Brno, Czech Republic.
| |
Collapse
|
18
|
Bittremieux W, Walzer M, Tenzer S, Zhu W, Salek RM, Eisenacher M, Tabb DL. The Human Proteome Organization-Proteomics Standards Initiative Quality Control Working Group: Making Quality Control More Accessible for Biological Mass Spectrometry. Anal Chem 2017; 89:4474-4479. [PMID: 28318237 DOI: 10.1021/acs.analchem.6b04310] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
To have confidence in results acquired during biological mass spectrometry experiments, a systematic approach to quality control is of vital importance. Nonetheless, until now, only scattered initiatives have been undertaken to this end, and these individual efforts have often not been complementary. To address this issue, the Human Proteome Organization-Proteomics Standards Initiative has established a new working group on quality control at its meeting in the spring of 2016. The goal of this working group is to provide a unifying framework for quality control data. The initial focus will be on providing a community-driven standardized file format for quality control. For this purpose, the previously proposed qcML format will be adapted to support a variety of use cases for both proteomics and metabolomics applications, and it will be established as an official PSI format. An important consideration is to avoid enforcing restrictive requirements on quality control but instead provide the basic technical necessities required to support extensive quality control for any type of mass spectrometry-based workflow. We want to emphasize that this is an open community effort, and we seek participation from all scientists with an interest in this field.
Collapse
Affiliation(s)
- Wout Bittremieux
- Department of Mathematics and Computer Science, University of Antwerp , Middelheimlaan 1, 2020 Antwerp, Belgium.,Biomedical Informatics Research Center Antwerp (biomina), University of Antwerp/Antwerp University Hospital , Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Mathias Walzer
- Department of Computer Science, University of Tübingen , Tübingen 72076, Germany.,Center for Bioinformatics, University of Tübingen , Tübingen 72074, Germany
| | - Stefan Tenzer
- Institute for Immunology, University Medical Center of the Johannes-Gutenberg University Mainz D 55131, Germany
| | - Weimin Zhu
- National Center for Protein Science , No. 38, Science Park Road, Changping District, Beijing 102206, China
| | - Reza M Salek
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI) , Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, United Kingdom
| | - Martin Eisenacher
- Medical Bioinformatics, Medizinisches Proteom-Center, Ruhr-University Bochum , Bochum 44801, Germany
| | - David L Tabb
- Division of Molecular Biology and Human Genetics, Stellenbosch University Faculty of Medicine and Health Sciences , Tygerberg Hospital, Francie Van Zijl Drive, Cape Town 7505, South Africa
| |
Collapse
|
19
|
Popova AA, Koksharova OA. Neurotoxic Non-proteinogenic Amino Acid β-N-Methylamino-L-alanine and Its Role in Biological Systems. BIOCHEMISTRY (MOSCOW) 2017; 81:794-805. [PMID: 27677549 DOI: 10.1134/s0006297916080022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Secondary metabolites of photoautotrophic organisms have attracted considerable interest in recent years. In particular, molecules of non-proteinogenic amino acids participating in various physiological processes and capable of producing adverse ecological effects have been actively investigated. For example, the non-proteinogenic amino acid β-N-methylamino-L-alanine (BMAA) is neurotoxic to animals including humans. It is known that BMAA accumulation via the food chain can lead to development of neurodegenerative diseases in humans such as Alzheimer's and Parkinson's diseases as well as amyotrophic lateral sclerosis. Moreover, BMAA can be mistakenly incorporated into a protein molecule instead of serine. Natural sources of BMAA and methods for its detection are discussed in this review, as well as the role of BMAA in metabolism of its producers and possible mechanisms of toxicity of this amino acid in different living organisms.
Collapse
Affiliation(s)
- A A Popova
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia.
| | | |
Collapse
|
20
|
Karlsson O, Hanrieder J. Imaging mass spectrometry in drug development and toxicology. Arch Toxicol 2016; 91:2283-2294. [PMID: 27933369 PMCID: PMC5429351 DOI: 10.1007/s00204-016-1905-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 11/24/2016] [Indexed: 11/25/2022]
Abstract
During the last decades, imaging mass spectrometry has gained significant relevance in biomedical research. Recent advances in imaging mass spectrometry have paved the way for in situ studies on drug development, metabolism and toxicology. In contrast to whole-body autoradiography that images the localization of radiolabeled compounds, imaging mass spectrometry provides the possibility to simultaneously determine the discrete tissue distribution of the parent compound and its metabolites. In addition, imaging mass spectrometry features high molecular specificity and allows comprehensive, multiplexed detection and localization of hundreds of proteins, peptides and lipids directly in tissues. Toxicologists traditionally screen for adverse findings by histopathological examination. However, studies of the molecular and cellular processes underpinning toxicological and pathologic findings induced by candidate drugs or toxins are important to reach a mechanistic understanding and an effective risk assessment strategy. One of IMS strengths is the ability to directly overlay the molecular information from the mass spectrometric analysis with the tissue section and allow correlative comparisons of molecular and histologic information. Imaging mass spectrometry could therefore be a powerful tool for omics profiling of pharmacological/toxicological effects of drug candidates and toxicants in discrete tissue regions. The aim of the present review is to provide an overview of imaging mass spectrometry, with particular focus on MALDI imaging mass spectrometry, and its use in drug development and toxicology in general.
Collapse
Affiliation(s)
- Oskar Karlsson
- Center for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institute, 171 76, Stockholm, Sweden.
- Department of Pharmaceutical Biosciences, Drug Safety and Toxicology, Uppsala University, 751 24, Uppsala, Sweden.
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, 431 80, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, Queen Square, London, WC1N, UK
| |
Collapse
|
21
|
|
22
|
Schweizer N, Viereckel T, Smith-Anttila CJ, Nordenankar K, Arvidsson E, Mahmoudi S, Zampera A, Wärner Jonsson H, Bergquist J, Lévesque D, Konradsson-Geuken Å, Andersson M, Dumas S, Wallén-Mackenzie Å. Reduced Vglut2/Slc17a6 Gene Expression Levels throughout the Mouse Subthalamic Nucleus Cause Cell Loss and Structural Disorganization Followed by Increased Motor Activity and Decreased Sugar Consumption. eNeuro 2016; 3:ENEURO.0264-16.2016. [PMID: 27699212 PMCID: PMC5041164 DOI: 10.1523/eneuro.0264-16.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 09/08/2016] [Indexed: 12/24/2022] Open
Abstract
The subthalamic nucleus (STN) plays a central role in motor, cognitive, and affective behavior. Deep brain stimulation (DBS) of the STN is the most common surgical intervention for advanced Parkinson's disease (PD), and STN has lately gained attention as target for DBS in neuropsychiatric disorders, including obsessive compulsive disorder, eating disorders, and addiction. Animal studies using STN-DBS, lesioning, or inactivation of STN neurons have been used extensively alongside clinical studies to unravel the structural organization, circuitry, and function of the STN. Recent studies in rodent STN models have exposed different roles for STN neurons in reward-related functions. We have previously shown that the majority of STN neurons express the vesicular glutamate transporter 2 gene (Vglut2/Slc17a6) and that reduction of Vglut2 mRNA levels within the STN of mice [conditional knockout (cKO)] causes reduced postsynaptic activity and behavioral hyperlocomotion. The cKO mice showed less interest in fatty rewards, which motivated analysis of reward-response. The current results demonstrate decreased sugar consumption and strong rearing behavior, whereas biochemical analyses show altered dopaminergic and peptidergic activity in the striatum. The behavioral alterations were in fact correlated with opposite effects in the dorsal versus the ventral striatum. Significant cell loss and disorganization of the STN structure was identified, which likely accounts for the observed alterations. Rare genetic variants of the human VGLUT2 gene exist, and this study shows that reduced Vglut2/Slc17a6 gene expression levels exclusively within the STN of mice is sufficient to cause strong modifications in both the STN and the mesostriatal dopamine system.
Collapse
Affiliation(s)
- Nadine Schweizer
- Department of Organismal Biology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Thomas Viereckel
- Department of Organismal Biology, Uppsala University, SE-752 36 Uppsala, Sweden
- Department of Neuroscience, Uppsala University, SE-751 24 Uppsala, Sweden
| | | | - Karin Nordenankar
- Department of Neuroscience, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Emma Arvidsson
- Department of Organismal Biology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Souha Mahmoudi
- Faculty of Pharmacy, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | | | - Hanna Wärner Jonsson
- Department of Pharmaceutical Biosciences, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Jonas Bergquist
- Department of Chemistry, BMC - Analytical Chemistry and Neurochemistry, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Daniel Lévesque
- Faculty of Pharmacy, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | | | - Malin Andersson
- Department of Pharmaceutical Biosciences, Uppsala University, SE-751 24 Uppsala, Sweden
| | | | | |
Collapse
|
23
|
Novak M, Hercog K, Žegura B. Assessment of the mutagenic and genotoxic activity of cyanobacterial toxin beta-N-methyl-amino-L-alanine in Salmonella typhimurium. Toxicon 2016; 118:134-40. [DOI: 10.1016/j.toxicon.2016.04.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 04/27/2016] [Accepted: 04/28/2016] [Indexed: 11/26/2022]
|
24
|
Potential of MALDI imaging for the toxicological evaluation of environmental pollutants. J Proteomics 2016; 144:133-9. [PMID: 27178109 DOI: 10.1016/j.jprot.2016.05.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/13/2016] [Accepted: 05/08/2016] [Indexed: 01/02/2023]
Abstract
Risk assessment related to the exposure of humans to chemicals released into the environment is a major concern of our modern societies. In this context, toxicology plays a crucial role to characterize the effects of this exposure on health and identify the targets of these molecules. MALDI imaging mass spectrometry (IMS) is an enabling technology for biodistribution studies of chemicals. Although the majority of published studies are presented in a pharmacological context, the concepts discussed in this review can be applied to the toxicological evaluation of chemicals released into the environment. The major asset of IMS is the simultaneous localization and identification of a parent molecule and its metabolites without labeling and without any prior knowledge. Quantification methods developed in IMS are presented with application to an environmental pollutant. IMS is effective in the localization of chemicals and endogenous species. This opens unique perspectives for the discovery of molecular alterations in metabolites and protein biomarkers that could help for a better understanding of toxicity mechanisms. Distribution studies of agrochemicals in plants by IMS can contribute to a better understanding of their mode of action and to a more effective use of these chemicals, avoiding the current concern of environmental damage.
Collapse
|
25
|
Tikka S, Monogioudi E, Gotsopoulos A, Soliymani R, Pezzini F, Scifo E, Uusi-Rauva K, Tyynelä J, Baumann M, Jalanko A, Simonati A, Lalowski M. Proteomic Profiling in the Brain of CLN1 Disease Model Reveals Affected Functional Modules. Neuromolecular Med 2015; 18:109-33. [PMID: 26707855 DOI: 10.1007/s12017-015-8382-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/15/2015] [Indexed: 02/06/2023]
Abstract
Neuronal ceroid lipofuscinoses (NCL) are the most commonly inherited progressive encephalopathies of childhood. Pathologically, they are characterized by endolysosomal storage with different ultrastructural features and biochemical compositions. The molecular mechanisms causing progressive neurodegeneration and common molecular pathways linking expression of different NCL genes are largely unknown. We analyzed proteome alterations in the brains of a mouse model of human infantile CLN1 disease-palmitoyl-protein thioesterase 1 (Ppt1) gene knockout and its wild-type age-matched counterpart at different stages: pre-symptomatic, symptomatic and advanced. For this purpose, we utilized a combination of laser capture microdissection-based quantitative liquid chromatography tandem mass spectrometry (MS) and matrix-assisted laser desorption/ionization time-of-flight MS imaging to quantify/visualize the changes in protein expression in disease-affected brain thalamus and cerebral cortex tissue slices, respectively. Proteomic profiling of the pre-symptomatic stage thalamus revealed alterations mostly in metabolic processes and inhibition of various neuronal functions, i.e., neuritogenesis. Down-regulation in dynamics associated with growth of plasma projections and cellular protrusions was further corroborated by findings from RNA sequencing of CLN1 patients' fibroblasts. Changes detected at the symptomatic stage included: mitochondrial functions, synaptic vesicle transport, myelin proteome and signaling cascades, such as RhoA signaling. Considerable dysregulation of processes related to mitochondrial cell death, RhoA/Huntington's disease signaling and myelin sheath breakdown were observed at the advanced stage of the disease. The identified changes in protein levels were further substantiated by bioinformatics and network approaches, immunohistochemistry on brain tissues and literature knowledge, thus identifying various functional modules affected in the CLN1 childhood encephalopathy.
Collapse
Affiliation(s)
- Saara Tikka
- Medicum, Biochemistry/Developmental Biology, Meilahti Clinical Proteomics Core Facility, University of Helsinki, P.O. Box 63 (Haartmaninkatu 8), Room C214a, 00014, Helsinki, Finland.,Folkhälsan Institute of Genetics, 00014, Helsinki, Finland
| | - Evanthia Monogioudi
- Folkhälsan Institute of Genetics, 00014, Helsinki, Finland.,Joint Research Centre, Directorate D-Institute for Reference Materials and Measurements, Standards for Innovation and Sustainable Development, Geel, Belgium
| | - Athanasios Gotsopoulos
- Brain and Mind Laboratory, Department of Biomedical Engineering and Computational Science (BECS), Aalto University School of Science, 02150, Espoo, Finland
| | - Rabah Soliymani
- Medicum, Biochemistry/Developmental Biology, Meilahti Clinical Proteomics Core Facility, University of Helsinki, P.O. Box 63 (Haartmaninkatu 8), Room C214a, 00014, Helsinki, Finland
| | - Francesco Pezzini
- Department of Neurological and Movement Sciences, University of Verona, 37134, Verona, Italy
| | - Enzo Scifo
- Medicum, Biochemistry/Developmental Biology, Meilahti Clinical Proteomics Core Facility, University of Helsinki, P.O. Box 63 (Haartmaninkatu 8), Room C214a, 00014, Helsinki, Finland.,Doctoral Program Brain & Mind, University of Helsinki, Helsinki, Finland.,Campbell Family Mental Health Research Institute, CAMH, University of Toronto, Toronto, Canada
| | - Kristiina Uusi-Rauva
- Folkhälsan Institute of Genetics, 00014, Helsinki, Finland.,Genomics and Biomarkers, National Institute for Health and Welfare (THL), P.O. Box 30, 00271, Helsinki, Finland
| | - Jaana Tyynelä
- Medicum, Biochemistry/Developmental Biology, Meilahti Clinical Proteomics Core Facility, University of Helsinki, P.O. Box 63 (Haartmaninkatu 8), Room C214a, 00014, Helsinki, Finland
| | - Marc Baumann
- Medicum, Biochemistry/Developmental Biology, Meilahti Clinical Proteomics Core Facility, University of Helsinki, P.O. Box 63 (Haartmaninkatu 8), Room C214a, 00014, Helsinki, Finland
| | - Anu Jalanko
- Institute for Molecular Medicine (FIMM), University of Helsinki, 00014, Helsinki, Finland.,Genomics and Biomarkers, National Institute for Health and Welfare (THL), P.O. Box 30, 00271, Helsinki, Finland
| | - Alessandro Simonati
- Department of Neurological and Movement Sciences, University of Verona, 37134, Verona, Italy
| | - Maciej Lalowski
- Medicum, Biochemistry/Developmental Biology, Meilahti Clinical Proteomics Core Facility, University of Helsinki, P.O. Box 63 (Haartmaninkatu 8), Room C214a, 00014, Helsinki, Finland. .,Folkhälsan Institute of Genetics, 00014, Helsinki, Finland.
| |
Collapse
|
26
|
Palmer A, Ovchinnikova E, Thuné M, Lavigne R, Guével B, Dyatlov A, Vitek O, Pineau C, Borén M, Alexandrov T. Using collective expert judgements to evaluate quality measures of mass spectrometry images. Bioinformatics 2015; 31:i375-84. [PMID: 26072506 PMCID: PMC4765867 DOI: 10.1093/bioinformatics/btv266] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Motivation: Imaging mass spectrometry (IMS) is a maturating technique of molecular imaging. Confidence in the reproducible quality of IMS data is essential for its integration into routine use. However, the predominant method for assessing quality is visual examination, a time consuming, unstandardized and non-scalable approach. So far, the problem of assessing the quality has only been marginally addressed and existing measures do not account for the spatial information of IMS data. Importantly, no approach exists for unbiased evaluation of potential quality measures. Results: We propose a novel approach for evaluating potential measures by creating a gold-standard set using collective expert judgements upon which we evaluated image-based measures. To produce a gold standard, we engaged 80 IMS experts, each to rate the relative quality between 52 pairs of ion images from MALDI-TOF IMS datasets of rat brain coronal sections. Experts’ optional feedback on their expertise, the task and the survey showed that (i) they had diverse backgrounds and sufficient expertise, (ii) the task was properly understood, and (iii) the survey was comprehensible. A moderate inter-rater agreement was achieved with Krippendorff’s alpha of 0.5. A gold-standard set of 634 pairs of images with accompanying ratings was constructed and showed a high agreement of 0.85. Eight families of potential measures with a range of parameters and statistical descriptors, giving 143 in total, were evaluated. Both signal-to-noise and spatial chaos-based measures performed highly with a correlation of 0.7 to 0.9 with the gold standard ratings. Moreover, we showed that a composite measure with the linear coefficients (trained on the gold standard with regularized least squares optimization and lasso) showed a strong linear correlation of 0.94 and an accuracy of 0.98 in predicting which image in a pair was of higher quality. Availability and implementation: The anonymized data collected from the survey and the Matlab source code for data processing can be found at: https://github.com/alexandrovteam/IMS_quality. Contact:theodore.alexandrov@embl.de
Collapse
Affiliation(s)
- Andrew Palmer
- European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technology, Karlsruhe, Germany, Denator, Uppsala, Sweden, Protim, Inserm U1085 - Irset, University of Rennes 1, Rennes, France, SCiLS GmbH, Bremen, Germany, College of Computer and Information Science, Northeastern University, Boston, MA, USA and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technology, Karlsruhe, Germany, Denator, Uppsala, Sweden, Protim, Inserm U1085 - Irset, University of Rennes 1, Rennes, France, SCiLS GmbH, Bremen, Germany, College of Computer and Information Science, Northeastern University, Boston, MA, USA and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Ekaterina Ovchinnikova
- European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technology, Karlsruhe, Germany, Denator, Uppsala, Sweden, Protim, Inserm U1085 - Irset, University of Rennes 1, Rennes, France, SCiLS GmbH, Bremen, Germany, College of Computer and Information Science, Northeastern University, Boston, MA, USA and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technology, Karlsruhe, Germany, Denator, Uppsala, Sweden, Protim, Inserm U1085 - Irset, University of Rennes 1, Rennes, France, SCiLS GmbH, Bremen, Germany, College of Computer and Information Science, Northeastern University, Boston, MA, USA and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Mikael Thuné
- European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technology, Karlsruhe, Germany, Denator, Uppsala, Sweden, Protim, Inserm U1085 - Irset, University of Rennes 1, Rennes, France, SCiLS GmbH, Bremen, Germany, College of Computer and Information Science, Northeastern University, Boston, MA, USA and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Régis Lavigne
- European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technology, Karlsruhe, Germany, Denator, Uppsala, Sweden, Protim, Inserm U1085 - Irset, University of Rennes 1, Rennes, France, SCiLS GmbH, Bremen, Germany, College of Computer and Information Science, Northeastern University, Boston, MA, USA and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Blandine Guével
- European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technology, Karlsruhe, Germany, Denator, Uppsala, Sweden, Protim, Inserm U1085 - Irset, University of Rennes 1, Rennes, France, SCiLS GmbH, Bremen, Germany, College of Computer and Information Science, Northeastern University, Boston, MA, USA and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Andrey Dyatlov
- European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technology, Karlsruhe, Germany, Denator, Uppsala, Sweden, Protim, Inserm U1085 - Irset, University of Rennes 1, Rennes, France, SCiLS GmbH, Bremen, Germany, College of Computer and Information Science, Northeastern University, Boston, MA, USA and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technology, Karlsruhe, Germany, Denator, Uppsala, Sweden, Protim, Inserm U1085 - Irset, University of Rennes 1, Rennes, France, SCiLS GmbH, Bremen, Germany, College of Computer and Information Science, Northeastern University, Boston, MA, USA and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Olga Vitek
- European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technology, Karlsruhe, Germany, Denator, Uppsala, Sweden, Protim, Inserm U1085 - Irset, University of Rennes 1, Rennes, France, SCiLS GmbH, Bremen, Germany, College of Computer and Information Science, Northeastern University, Boston, MA, USA and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Charles Pineau
- European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technology, Karlsruhe, Germany, Denator, Uppsala, Sweden, Protim, Inserm U1085 - Irset, University of Rennes 1, Rennes, France, SCiLS GmbH, Bremen, Germany, College of Computer and Information Science, Northeastern University, Boston, MA, USA and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Mats Borén
- European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technology, Karlsruhe, Germany, Denator, Uppsala, Sweden, Protim, Inserm U1085 - Irset, University of Rennes 1, Rennes, France, SCiLS GmbH, Bremen, Germany, College of Computer and Information Science, Northeastern University, Boston, MA, USA and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Theodore Alexandrov
- European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technology, Karlsruhe, Germany, Denator, Uppsala, Sweden, Protim, Inserm U1085 - Irset, University of Rennes 1, Rennes, France, SCiLS GmbH, Bremen, Germany, College of Computer and Information Science, Northeastern University, Boston, MA, USA and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technology, Karlsruhe, Germany, Denator, Uppsala, Sweden, Protim, Inserm U1085 - Irset, University of Rennes 1, Rennes, France, SCiLS GmbH, Bremen, Germany, College of Computer and Information Science, Northeastern University, Boston, MA, USA and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technology, Karlsruhe, Germany, Denator, Uppsala, Sweden, Protim, Inserm U1085 - Irset, University of Rennes 1, Rennes, France, SCiLS GmbH, Bremen, Germany, College of Computer and Information Science, Northeastern University, Boston, MA, USA and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA European Molecular Biology Laboratory, Heidelberg, Germany, Center for Industrial Mathematics, University of Bremen, Bremen, Germany, High Performance Humanoid Technologies Lab, Institute for Anthropomatics, Karlsruhe Institute of Technolo
| |
Collapse
|
27
|
Environmental neurotoxin interaction with proteins: Dose-dependent increase of free and protein-associated BMAA (β-N-methylamino-L-alanine) in neonatal rat brain. Sci Rep 2015; 5:15570. [PMID: 26498001 PMCID: PMC4620439 DOI: 10.1038/srep15570] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/25/2015] [Indexed: 12/12/2022] Open
Abstract
β-Methylamino-L-alanine (BMAA) is implicated in the aetiology of neurodegenerative disorders. Neonatal exposure to BMAA induces cognitive impairments and progressive neurodegenerative changes including intracellular fibril formation in the hippocampus of adult rats. It is unclear why the neonatal hippocampus is especially vulnerable and the critical cellular perturbations preceding BMAA-induced toxicity remains to be elucidated. The aim of this study was to compare the level of free and protein-associated BMAA in neonatal rat brain and peripheral tissues after different exposures to BMAA. Ultra-high performance liquid chromatography-tandem mass spectrometry analysis revealed that BMAA passed the neonatal blood-brain barrier and was distributed to all studied brain areas. BMAA was also associated to proteins in the brain, especially in the hippocampus. The level in the brain was, however, considerably lower compared to the liver that is not a target organ for BMAA. In contrast to the liver there was a significantly increased level of protein-association of BMAA in the hippocampus and other brain areas following repeated administration suggesting that the degradation of BMAA-associated proteins may be lower in neonatal brain than in the liver. Additional evidence is needed in support of a role for protein misincorporation in the neonatal hippocampus for long-term effects of BMAA.
Collapse
|
28
|
Hanrieder J, Malmberg P, Ewing AG. Spatial neuroproteomics using imaging mass spectrometry. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:718-31. [PMID: 25582083 DOI: 10.1016/j.bbapap.2014.12.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/11/2014] [Accepted: 12/19/2014] [Indexed: 12/12/2022]
Abstract
The nervous system constitutes arguably the most complicated and least understood cellular network in the human body. This consequently manifests itself in the fact that the molecular bases of neurodegenerative diseases remain unknown. The limited understanding of neurobiological mechanisms relates directly to the lack of appropriate bioanalytical technologies that allow highly resolved, sensitive, specific and comprehensive molecular imaging in complex biological matrices. Imaging mass spectrometry (IMS) is an emerging technique for molecular imaging. The technique is characterized by its high chemical specificity allowing comprehensive, spatial protein and peptide profiling in situ. Imaging MS represents therefore a powerful approach for investigation of spatio-temporal protein and peptide regulations in CNS derived tissue and cells. This review aims to provide a concise overview of major developments and applications concerning imaging mass spectrometry based protein and peptide profiling in neurobiological and biomedical research. This article is part of a Special Issue entitled: Neuroproteomics: Applications in Neuroscience and Neurology.
Collapse
Affiliation(s)
- Jörg Hanrieder
- National Center for Imaging Mass Spectrometry, University of Gothenburg and Chalmers University of Technology, Gothenburg, Sweden; Department of Chemical and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden; Institute of Neuroscience and Physiology, Department Psychiatry and Neurochemistry, University of Gothenburg, Sahlgrenska University Hospital Mölndal, Mölndal, Sweden
| | - Per Malmberg
- National Center for Imaging Mass Spectrometry, University of Gothenburg and Chalmers University of Technology, Gothenburg, Sweden; Department of Chemical and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Andrew G Ewing
- National Center for Imaging Mass Spectrometry, University of Gothenburg and Chalmers University of Technology, Gothenburg, Sweden; Department of Chemical and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden; Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
29
|
Glover WB, Mash DC, Murch SJ. The natural non-protein amino acid N-β-methylamino-L-alanine (BMAA) is incorporated into protein during synthesis. Amino Acids 2014; 46:2553-9. [PMID: 25096519 DOI: 10.1007/s00726-014-1812-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 07/14/2014] [Indexed: 11/25/2022]
Abstract
N-β-methylamino-L-alanine (BMAA) is an amino acid produced by cyanobacteria and accumulated through trophic levels in the environment and natural food webs. Human exposure to BMAA has been linked to progressive neurodegenerative diseases, potentially due to incorporation of BMAA into protein. The insertion of BMAA and other non-protein amino acids into proteins may trigger protein misfunction, misfolding and/or aggregation. However, the specific mechanism by which BMAA is associated with proteins remained unidentified. Such studies are challenging because of the complexity of biological systems and samples. A cell-free in vitro protein synthesis system offers an excellent approach for investigation of changing amino acid composition in protein. In this study, we report that BMAA incorporates into protein as an error in synthesis when a template DNA sequence is used. Bicinchoninic acid assay of total protein synthesis determined that BMAA effectively substituted for alanine and serine in protein product. LC-MS/MS confirmed that BMAA was selectively inserted into proteins in place of other amino acids, but isomers N-(2-aminoethyl)glycine (AEG) and 2,4-diaminobutyric acid (DAB) did not share this characteristic. Incorporation of BMAA into proteins was significantly higher when genomic DNA from post-mortem brain was the template. About half of BMAA in the synthetic proteins was released with denaturation with sodium dodecylsulfonate and dithiothreitol, but the remaining BMAA could only be released by acid hydrolysis. Together these data demonstrate that BMAA is incorporated into the amino acid backbone of proteins during synthesis and also associated with proteins through non-covalent bonding.
Collapse
Affiliation(s)
- W Broc Glover
- Department of Chemistry, University of British Columbia, 3247 University Way, Kelowna, BC, V1V 1V7, Canada
| | | | | |
Collapse
|
30
|
Hanrieder J, Gerber L, Persson Sandelius Å, Brittebo EB, Ewing AG, Karlsson O. High resolution metabolite imaging in the hippocampus following neonatal exposure to the environmental toxin BMAA using ToF-SIMS. ACS Chem Neurosci 2014; 5:568-75. [PMID: 24779349 PMCID: PMC4102959 DOI: 10.1021/cn500039b] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/28/2014] [Indexed: 12/11/2022] Open
Abstract
The environmental neurotoxin β-N-methylamino-L-alanine (BMAA) is suggested to be linked with neurodegenerative disease. In a rat model, neonatal exposure to BMAA induced selective uptake in the hippocampus and caused cell loss, mineralization and astrogliosis as well as learning and memory impairments in adulthood. Moreover, neonatal exposure resulted in increased protein ubiquitination in the cornus ammonis 1 (CA1) region of the adult hippocampus indicating that BMAA may induce protein aggregation. Time-of-flight secondary ion mass spectrometry (ToF-SIMS) based imaging is a powerful technology for spatial profiling of small molecular weight compounds in biological tissues with high chemical specificity and high spatial resolution. The aim of this study was to characterize neurochemical changes in the hippocampus of six month-old rats treated neonatally (postnatal days 9-10) with BMAA. Multivariate data analysis of whole section ToF-SIMS scans was performed to delineate anatomical regions of interest based on their chemical distribution pattern. Further analysis of spectral data obtained from the outlined anatomical regions, including CA1 and dentate gyrus (DG) revealed BMAA-induced long-term changes. Increased levels of phospholipids and protein fragments in the histopathologically altered CA1 region as well as phosphate depletion in the DG were observed. Moreover, high resolution SIMS imaging revealed a specific localization of phosphatidylcholine lipids, protein signals and potassium in the histopathologically altered CA1. These findings demonstrate that ToF-SIMS based imaging is a powerful approach for probing biochemical changes in situ and might serve as promising technique for investigating neurotoxin-induced brain pathology.
Collapse
Affiliation(s)
- Jörg Hanrieder
- National Center
for Imaging Mass Spectrometry, University of Gothenburg and Chalmers
University of Technology, SE-412 96 Gothenburg, Sweden
- Department
of Chemical and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg Sweden
| | - Lorenz Gerber
- Umeå
Plant Science Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, SE-901 83 Umeå, Sweden
| | - Åsa Persson Sandelius
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, SE-412 96 Gothenburg, Sweden
| | - Eva B. Brittebo
- Department
of Pharmaceutical Biosciences, Drug Safety and Toxicology, Uppsala University, SE-751 05 Uppsala, Sweden
| | - Andrew G. Ewing
- National Center
for Imaging Mass Spectrometry, University of Gothenburg and Chalmers
University of Technology, SE-412 96 Gothenburg, Sweden
- Department
of Chemical and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg Sweden
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, SE-412 96 Gothenburg, Sweden
| | - Oskar Karlsson
- Department
of Pharmaceutical Biosciences, Drug Safety and Toxicology, Uppsala University, SE-751 05 Uppsala, Sweden
- Department
of Environmental Toxicology, Uppsala University, SE-751 05 Uppsala, Sweden
| |
Collapse
|
31
|
Intracellular fibril formation, calcification, and enrichment of chaperones, cytoskeletal, and intermediate filament proteins in the adult hippocampus CA1 following neonatal exposure to the nonprotein amino acid BMAA. Arch Toxicol 2014; 89:423-36. [PMID: 24798087 PMCID: PMC4335130 DOI: 10.1007/s00204-014-1262-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/15/2014] [Indexed: 12/31/2022]
Abstract
The environmental neurotoxin β-N-methylamino-l-alanine (BMAA) has been implicated in the etiology of neurodegenerative disease, and recent studies indicate that BMAA can be misincorporated into proteins. BMAA is a developmental neurotoxicant that can induce long-term learning and memory deficits, as well as regionally restricted neuronal degeneration and mineralization in the hippocampal CA1. The aim of the study was to characterize long-term changes (2 weeks to 6 months) further in the brain of adult rats treated neonatally (postnatal days 9–10) with BMAA (460 mg/kg) using immunohistochemistry (IHC), transmission electron microscopy, and laser capture microdissection followed by LC-MS/MS for proteomic analysis. The histological examination demonstrated progressive neurodegenerative changes, astrogliosis, microglial activation, and calcification in the hippocampal CA1 3–6 months after exposure. The IHC showed an increased staining for α-synuclein and ubiquitin in the area. The ultrastructural examination revealed intracellular deposition of abundant bundles of closely packed parallel fibrils in neurons, axons, and astrocytes of the CA1. Proteomic analysis of the affected site demonstrated an enrichment of chaperones (e.g., clusterin, GRP-78), cytoskeletal and intermediate filament proteins, and proteins involved in the antioxidant defense system. Several of the most enriched proteins (plectin, glial fibrillar acidic protein, vimentin, Hsp 27, and ubiquitin) are known to form complex astrocytic inclusions, so-called Rosenthal fibers, in the neurodegenerative disorder Alexander disease. In addition, TDP-43 and the negative regulator of autophagy, GLIPR-2, were exclusively detected. The present study demonstrates that neonatal exposure to BMAA may offer a novel model for the study of hippocampal fibril formation in vivo.
Collapse
|
32
|
Karlsson O, Jiang L, Andersson M, Ilag LL, Brittebo EB. Protein association of the neurotoxin and non-protein amino acid BMAA (β-N-methylamino-L-alanine) in the liver and brain following neonatal administration in rats. Toxicol Lett 2014; 226:1-5. [PMID: 24472610 DOI: 10.1016/j.toxlet.2014.01.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 01/16/2014] [Accepted: 01/17/2014] [Indexed: 10/25/2022]
Abstract
The environmental neurotoxin β-N-methylamino-L-alanine (BMAA) is not an amino acid that is normally found in proteins. Our previous autoradiographic study of (3)H-labeled BMAA in adult mice unexpectedly revealed a tissue distribution similar to that of protein amino acids. The aim of this study was to characterize the distribution of free and protein-bound BMAA in neonatal rat tissues following a short exposure using autoradiographic imaging and ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). The autoradiographic imaging of (14)C-L-BMAA demonstrated a distinct uptake of radioactivity that was retained following acid extraction in tissues with a high rate of cell turnover and/or protein synthesis. The UHPLC-MS/MS analysis conclusively demonstrated a dose-dependent increase of protein-associated BMAA in neonatal rat tissues. The level of protein-associated BMAA in the liver was more than 10 times higher than that in brain regions not fully protected by the blood-brain barrier which may be due to the higher rate of protein synthesis in the liver. In conclusion, this study demonstrated that BMAA was associated with rat proteins suggesting that BMAA may be misincorporated into proteins. However, protein-associated BMAA seemed to be cleared over time, as none of the samples from adult rats had any detectable free or protein-associated BMAA.
Collapse
Affiliation(s)
- Oskar Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, SE-751 24 Uppsala, Sweden; Department of Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden.
| | - Liying Jiang
- Department of Analytical Chemistry, Stockholm University, SE-10691 Stockholm, Sweden
| | - Marie Andersson
- Department of Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Leopold L Ilag
- Department of Analytical Chemistry, Stockholm University, SE-10691 Stockholm, Sweden
| | - Eva B Brittebo
- Department of Pharmaceutical Biosciences, Uppsala University, SE-751 24 Uppsala, Sweden
| |
Collapse
|