1
|
Mohamed ZA, Elnour RO, Alamri S, Hashem M, Campos A, Vasconcelos V, Badawye H. Presence of the neurotoxin β-N-methylamino-L-alanine in irrigation water and accumulation in cereal grains with human exposure risk. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:31479-31491. [PMID: 38635096 DOI: 10.1007/s11356-024-33188-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 03/29/2024] [Indexed: 04/19/2024]
Abstract
The present study demonstrates the presence of the neurotoxin β-N-methylamino-L-alanine and its cyanobacterial producers in irrigation water and grains of some cereal plants from farmlands irrigated with Nile River water in Egypt. BMAA detected by LC-MS/MS in phytoplankton samples was found at higher concentrations of free form (0.84-11.4 μg L-1) than of protein-bound form (0.16-1.6 μg L-1), in association with the dominance of cyanobacteria in irrigation water canals. Dominant cyanobacterial species isolated from these irrigation waters including Aphanocapsa planctonica, Chroococcus minutus, Dolichospermum lemmermanni, Nostoc commune, and Oscillatoria tenuis were found to produce different concentrations of free (4.8-71.1 µg g-1 dry weight) and protein-bound (0.1-11.4 µg g-1 dry weight) BMAA. In the meantime, BMAA was also detected in a protein-bound form only in grains of corn (3.87-4.51 µg g-1 fresh weight) and sorghum (5.1-7.1 µg g-1 fresh weight) plants, but not in wheat grains. The amounts of BMAA accumulated in these grains correlated with BMAA concentrations detected in relevant irrigation water canals. The presence of BMAA in cereal grains would constitute a risk to human and animal health upon consumption of contaminated grains. The study, therefore, suggests continuous monitoring of BMAA and other cyanotoxins in irrigation waters and edible plants to protect the public against exposure to such potent toxins.
Collapse
Affiliation(s)
- Zakaria A Mohamed
- Department of Botany and Microbiology, Faculty of Science, Sohag University, Sohag, 82524, Egypt.
| | - Rehab O Elnour
- Biology Department, Faculty of Sciences and Arts, Dahran Al-Janoub, King Khalid University, Abha, Saudi Arabia
| | - Saad Alamri
- Department of Biology, College of Science, King Khalid University, Abha, 61413, Saudi Arabia
| | - Mohamed Hashem
- Department of Biology, College of Science, King Khalid University, Abha, 61413, Saudi Arabia
| | - Alexandre Campos
- CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, Terminal de Cruzeiros Do Porto de Leixões, Av. General Norton de Matos, S/N, 4450-208, Matosinhos, Portugal
| | - Vitor Vasconcelos
- CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, Terminal de Cruzeiros Do Porto de Leixões, Av. General Norton de Matos, S/N, 4450-208, Matosinhos, Portugal
- Departament of Biology, Faculty of Sciences, University of Porto, Rua Do Campo Alegre, 4169-007, Porto, Portugal
| | - Hanan Badawye
- Department of Botany and Microbiology, Faculty of Science, Sohag University, Sohag, 82524, Egypt
| |
Collapse
|
2
|
Manca E, Noli B, Corda G, El-Hassani M, Manai A, Sanna F, Argiolas A, Melis MR, Manconi B, Contini C, Cocco C. VGF modifications related to nigrostriatal dopaminergic neurodegeneration induced by the pesticide fipronil in adult male rats. Ann Anat 2024; 252:152194. [PMID: 38056781 DOI: 10.1016/j.aanat.2023.152194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/16/2023] [Accepted: 11/28/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Dopamine is reduced in the brain of rats treated with fipronil, a broad-spectrum insecticide. VGF (no acronym) is a neurotrophin-inducible protein expressed as the 75 kDa form (precursor or pro-VGF) or its truncated peptides. VGF immunostaining has been revealed using an antibody against the C-terminal nonapeptide of the rat pro-VGF in the nerve terminals of the rat substantia nigra, where it was reduced after 6-hydroxydopamine treatment. It is unknown whether pro-VGF and/or its shortened peptides are present in these neurons. Therefore, the aim of this study was first to determine which types of VGF are expressed in the normal substantia nigra (and striatum) and then to determine VGF modulations and whether they occur in parallel with locomotor changes after fipronil injection. METHODS Rats were divided into two groups that received a unilateral intranigral infusion of either fipronil (25 µg) diluted in dimethyl sulfoxide (DMSO) or DMSO alone, and then were tested for locomotor activity. An untreated group of rats (n=4) was used for identification of the VGF fragments using high performance liquid chromatography-mass spectrometry and western blot, while changes in treated groups (fipronil vs DMSO, each n=6) were investigated by immunohistochemistry using an antibody against the rat pro-VGF C-terminal nonapeptide in parallel with the anti-tyrosine hydroxylase antibody. RESULTS In untreated rats, the VGF C-terminal antibody identified mostly a 75 kDa band in the substantia nigra and striatum, supporting the finding of high-resolution mass spectrometry, which revealed fragments covering the majority of the pro-VGF sequence. Furthermore, several shortened VGF C-terminal forms (varying from 10 to 55 kDa) were also found by western blot, while high-resolution mass spectrometry revealed a C-terminal peptide overlapping the immunogen used to create the VGF antibody in both substantia nigra and striatum. In the substantia nigra of fipronil-treated rats, immunostaining for tyrosine hydroxylase and VGF was reduced compared to DMSO-treated rat group, and this was related with significant changes in locomotor activity. CONCLUSION Fipronil has the ability to modulate the production of pro-VGF and/or its C-terminal truncated peptides in the nigrostriatal system indicating its intimate interaction with the dopaminergic neurotransmission and implying a potential function in modulating locomotor activity.
Collapse
Affiliation(s)
- Elias Manca
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Barbara Noli
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Giulia Corda
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Majda El-Hassani
- Department of Internal Medicine III, University Hospital RWTH Aachen, Germany
| | - Antonio Manai
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Fabrizio Sanna
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Antonio Argiolas
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | | | - Barbara Manconi
- Department of Life Sciences and Environment, University of Cagliari, Italy
| | - Cristina Contini
- Department of Life Sciences and Environment, University of Cagliari, Italy
| | - Cristina Cocco
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
3
|
Fridjonsdottir E, Nilsson A, Fricker LD, Andrén PE. Two Different Strategies for Stabilization of Brain Tissue and Extraction of Neuropeptides. Methods Mol Biol 2024; 2758:49-60. [PMID: 38549007 DOI: 10.1007/978-1-0716-3646-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Neuropeptides are bioactive peptides that are synthesized and secreted by neurons in signaling pathways in the brain. Peptides and proteins are extremely vulnerable to proteolytic cleavage when their biological surrounding changes. This makes neuropeptidomics challenging due to the rapid alterations that occur to the peptidome after harvesting of brain tissue samples. For a successful neuropeptidomic study, the biological tissue sample analyzed should resemble the living state as much as possible. Heat stabilization has been proven to inhibit postmortem degradation by denaturing proteolytic enzymes, hence increasing identification rates of neuropeptides. Here, we describe two different stabilization protocols for rodent brain samples that increase the number of intact mature neuropeptides and minimize interference from degradation products of abundant proteins. Additionally, we present an extraction protocol that aims to extract a wide range of hydrophilic and hydrophobic neuropeptides by sequentially using an aqueous and an organic extraction medium.
Collapse
Affiliation(s)
- Elva Fridjonsdottir
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Anna Nilsson
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lloyd D Fricker
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Per E Andrén
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
4
|
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: 0.7] [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
|
5
|
Ra D, Sa B, Sl B, Js M, Sj M, DA D, Ew S, O K, Eb B, Ad C, Vx T, Gg G, Pa C, Dc M, Wg B. Is Exposure to BMAA a Risk Factor for Neurodegenerative Diseases? A Response to a Critical Review of the BMAA Hypothesis. Neurotox Res 2021; 39:81-106. [PMID: 33547590 PMCID: PMC7904546 DOI: 10.1007/s12640-020-00302-0] [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: 09/09/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/15/2022]
Abstract
In a literature survey, Chernoff et al. (2017) dismissed the hypothesis that chronic exposure to β-N-methylamino-L-alanine (BMAA) may be a risk factor for progressive neurodegenerative disease. They question the growing scientific literature that suggests the following: (1) BMAA exposure causes ALS/PDC among the indigenous Chamorro people of Guam; (2) Guamanian ALS/PDC shares clinical and neuropathological features with Alzheimer's disease, Parkinson's disease, and ALS; (3) one possible mechanism for protein misfolds is misincorporation of BMAA into proteins as a substitute for L-serine; and (4) chronic exposure to BMAA through diet or environmental exposures to cyanobacterial blooms can cause neurodegenerative disease. We here identify multiple errors in their critique including the following: (1) their review selectively cites the published literature; (2) the authors reported favorably on HILIC methods of BMAA detection while the literature shows significant matrix effects and peak coelution in HILIC that may prevent detection and quantification of BMAA in cyanobacteria; (3) the authors build alternative arguments to the BMAA hypothesis, rather than explain the published literature which, to date, has been unable to refute the BMAA hypothesis; and (4) the authors erroneously attribute methods to incorrect studies, indicative of a failure to carefully consider all relevant publications. The lack of attention to BMAA research begins with the review's title which incorrectly refers to BMAA as a "non-essential" amino acid. Research regarding chronic exposure to BMAA as a cause of human neurodegenerative diseases is emerging and requires additional resources, validation, and research. Here, we propose strategies for improvement in the execution and reporting of analytical methods and the need for additional and well-executed inter-lab comparisons for BMAA quantitation. We emphasize the need for optimization and validation of analytical methods to ensure that they are fit-for-purpose. Although there remain gaps in the literature, an increasingly large body of data from multiple independent labs using orthogonal methods provides increasing evidence that chronic exposure to BMAA may be a risk factor for neurological illness.
Collapse
Affiliation(s)
- Dunlop Ra
- Brain Chemistry Labs, Institute for Ethnomedicine, Jackson, WY, USA.
| | - Banack Sa
- Brain Chemistry Labs, Institute for Ethnomedicine, Jackson, WY, USA
| | - Bishop Sl
- Lewis Research Group, Faculty of Science, University of Calgary, Alberta, Canada
| | - Metcalf Js
- Brain Chemistry Labs, Institute for Ethnomedicine, Jackson, WY, USA
| | - Murch Sj
- Department of Chemistry, University of British Columbia, Kelowna, BC, Canada
| | - Davis DA
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Stommel Ew
- Department of Neurology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Karlsson O
- Department of Environmental Science, Stockholm University, Stockholm, Sweden
| | - Brittebo Eb
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | - Tan Vx
- Department of Biological Sciences, Macquarie University Centre for Motor Neuron Disease Research, Macquarie University, Ryde, Australia
| | - Guillemin Gg
- Department of Biological Sciences, Macquarie University Centre for Motor Neuron Disease Research, Macquarie University, Ryde, Australia
| | - Cox Pa
- Brain Chemistry Labs, Institute for Ethnomedicine, Jackson, WY, USA
| | - Mash Dc
- Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Bradley Wg
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
6
|
Pierozan P, Cattani D, Karlsson O. Hippocampal neural stem cells are more susceptible to the neurotoxin BMAA than primary neurons: effects on apoptosis, cellular differentiation, neurite outgrowth, and DNA methylation. Cell Death Dis 2020; 11:910. [PMID: 33099583 PMCID: PMC7585576 DOI: 10.1038/s41419-020-03093-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 12/16/2022]
Abstract
Developmental exposure to the environmental neurotoxin β-N-methylamino-L-alanine (BMAA), a proposed risk factor for neurodegenerative disease, can induce long-term cognitive impairments and neurodegeneration in rats. While rodent studies have demonstrated a low transfer of BMAA to the adult brain, this toxin is capable to cross the placental barrier and accumulate in the fetal brain. Here, we investigated the differential susceptibility of primary neuronal cells and neural stem cells from fetal rat hippocampus to BMAA toxicity. Exposure to 250 µM BMAA induced cell death in neural stem cells through caspase-independent apoptosis, while the proliferation of primary neurons was reduced only at 3 mM BMAA. At the lowest concentrations tested (50 and 100 µM), BMAA disrupted neural stem cell differentiation and impaired neurite development in neural stem cell-derived neurons (e.g., reduced neurite length, the number of processes and branches per cell). BMAA induced no alterations of the neurite outgrowth in primary neurons. This demonstrates that neural stem cells are more susceptible to BMAA exposure than primary neurons. Importantly, the changes induced by BMAA in neural stem cells were mitotically inherited to daughter cells. The persistent nature of the BMAA-induced effects may be related to epigenetic alterations that interfere with the neural stem cell programming, as BMAA exposure reduced the global DNA methylation in the cells. These findings provide mechanistic understanding of how early-life exposure to BMAA may lead to adverse long-term consequences, and potentially predispose for neurodevelopmental disorders or neurodegenerative disease later in life.
Collapse
Affiliation(s)
- Paula Pierozan
- Science for Life Laboratory, Department of Environmental Science, Stockholm University, 114 18, Stockholm, Sweden
| | - Daiane Cattani
- Science for Life Laboratory, Department of Environmental Science, Stockholm University, 114 18, Stockholm, Sweden
| | - Oskar Karlsson
- Science for Life Laboratory, Department of Environmental Science, Stockholm University, 114 18, Stockholm, Sweden.
| |
Collapse
|
7
|
Wu X, Wu H, Gu X, Zhang R, Ye J, Sheng Q. Biomagnification characteristics and health risk assessment of the neurotoxin BMAA in freshwater aquaculture products of Taihu Lake Basin, China. CHEMOSPHERE 2019; 229:332-340. [PMID: 31078890 DOI: 10.1016/j.chemosphere.2019.04.210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/15/2019] [Accepted: 04/28/2019] [Indexed: 06/09/2023]
Abstract
In freshwater aquaculture ecosystems with high-frequency occurrences of cyanobacteria blooms, a chronic neurotoxic cyanobacteria toxin, β-N-methylamino-l-alanine (BMAA), is a new pollutant that affects the normal growth, development, and reproduction of aquaculture organisms. BMAA poses a great threat to the food quality and food safety of aquatic products. In this paper, high-performance liquid chromatography-mass spectrometry (HPLC-MS/MS) was used to detect the contents of BMAA in the edible portions of six representative freshwater aquaculture products (Corbicula fluminea, Anodonta arcaeformis, Macrobrachium nipponense, Eriocheir sinensis, Ctenopharyngodon idella, and Mylopharyngodon piceus) from Taihu Lake Basin in China. Noncarcinogenic health risks were assessed with reference to the model recommended by the International Environmental Modelling and Software Society and based on the biomagnification characteristics of BMAA in the various aquaculture products investigated by the stable nitrogen isotope technique. The average BMAA concentrations in the edible portions of the six freshwater culture products were from 2.05 ± 1.40 to 4.21 ± 1.26 μg g-1 dry weight (DW), and the difference was significant (p < 0.05), such a difference increased with the increase in the trophic level in the aquaculture products. Although a biomagnification indication was observed, the trophic magnification factor (TMF) was only 1.20 which exhibited a relatively low biomagnification efficiency. The annual health risk values of BMAA in all the measured aquatic products were within the maximum tolerable range (<1 × 10-6 a-1), and the health risk increased with the increase in the trophic level. The risk values of BMAA in the six freshwater aquaculture products for children was slightly higher than the negligible level (<1 × 10-7 a-1), thus there might have potential health risks for children's long-term consumption. Considering China's national conditions, the guidance values of BMAA based on the quality and safety of freshwater aquaculture products were proposed to be 7.2 μg g-1 DW for adults and 1.8 μg g-1 DW for children.
Collapse
Affiliation(s)
- Xiang Wu
- Key Laboratory of Aquatic Resources Conservation and Development Technology Research, College of Life Sciences, Huzhou University, Huzhou City, Zhejiang Province, 313000, China.
| | - Hao Wu
- Environmental Protection Monitoring Centre Station, Huzhou City, Zhejiang Province, 313000, China
| | - Xiaoxiao Gu
- Key Laboratory of Aquatic Resources Conservation and Development Technology Research, College of Life Sciences, Huzhou University, Huzhou City, Zhejiang Province, 313000, China
| | - Rongfei Zhang
- Key Laboratory of Aquatic Resources Conservation and Development Technology Research, College of Life Sciences, Huzhou University, Huzhou City, Zhejiang Province, 313000, China
| | - Jinyun Ye
- Key Laboratory of Aquatic Resources Conservation and Development Technology Research, College of Life Sciences, Huzhou University, Huzhou City, Zhejiang Province, 313000, China
| | - Qiang Sheng
- Key Laboratory of Aquatic Resources Conservation and Development Technology Research, College of Life Sciences, Huzhou University, Huzhou City, Zhejiang Province, 313000, China
| |
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: 14] [Impact Index Per Article: 2.8] [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
|
Fridjonsdottir E, Nilsson A, Wadensten H, Andrén PE. Brain Tissue Sample Stabilization and Extraction Strategies for Neuropeptidomics. Methods Mol Biol 2019; 1719:41-49. [PMID: 29476502 DOI: 10.1007/978-1-4939-7537-2_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Neuropeptides are bioactive peptides that are synthesized and secreted by neurons in signaling pathways in the brain. Peptides and proteins are extremely vulnerable to proteolytic cleavage when their biological surrounding changes. This makes neuropeptidomics challenging due to the rapid alterations that occur to the peptidome after harvesting of brain tissue samples. For a successful neuropeptidomic study the biological tissue sample analyzed should resemble the premortem state as much as possible. Heat stabilization has been proven to inhibit postmortem degradation by denaturing proteolytic enzymes, hence increasing identification rates of neuropeptides. Here, we describe a stabilization protocol of a frozen tissue specimen that increases the number of intact mature neuropeptides identified and minimizes interference of degradation products from abundant proteins. Additionally, we present an extraction protocol that aims to extract a wide range of hydrophilic and hydrophobic neuropeptides by using both an aqueous and an organic extraction medium.
Collapse
Affiliation(s)
- Elva Fridjonsdottir
- Biomolecular Imaging and Proteomics, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Anna Nilsson
- Biomolecular Imaging and Proteomics, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Henrik Wadensten
- Biomolecular Imaging and Proteomics, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Per E Andrén
- Biomolecular Imaging and Proteomics, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
10
|
Wang H, Ji Y, Yin C, Deng M, Tang T, Deng B, Ren W, Deng J, Yin Y, Tan C. Differential Analysis of Gut Microbiota Correlated With Oxidative Stress in Sows With High or Low Litter Performance During Lactation. Front Microbiol 2018; 9:1665. [PMID: 30154758 PMCID: PMC6103269 DOI: 10.3389/fmicb.2018.01665] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 07/04/2018] [Indexed: 01/01/2023] Open
Abstract
It has been suggested that gut microbiota play a critical role in maternal metabolic oxidative stress responses and offspring growth. However, whether the gut microbiota and oxidative stress status of the sows affect the litter performance during lactation is unclear. A total of 66 Yorkshire sows were identified as high (H) or low (L) litter performance sows based on litter weight at day 21 of lactation. Ten sows per group with similar parity, backfat thickness, and litter weight after cross-foster from the H or L group were collected randomly to analyze the oxidative stress and gut microbiota during lactation. The result showed that the serum total antioxidant capacity was higher in the H group, while 8-hydroxy-deoxyguanosine and thiobarbituric acid reactive substances were lower in the H group at farrowing. Four distinct clusters of bacteria were related to litter performance and reproductive periods of sows. Twelve differentially abundant taxa during gestation and 13 taxa during lactation were identified as potential biomarkers between the H group and the L group. Moreover, the litter performance and the antioxidant capacity of sows were positively correlated with Bacteroides_f__Bacteroidaceae but negatively with Phascolarctobacterium and Streptococcus. In conclusion, this study found that gut microbiota and oxidative stress were significantly correlated with the litter performance of sows during lactation.
Collapse
Affiliation(s)
- Hao Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yongcheng Ji
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Cong Yin
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ming Deng
- The Herbivore Research Laboratory, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Tianyue Tang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Baichuan Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Wenkai Ren
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jinping Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yulong Yin
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China.,National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Chengquan Tan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
11
|
Cellular and Molecular Aspects of the β-N-Methylamino-l-alanine (BMAA) Mode of Action within the Neurodegenerative Pathway: Facts and Controversy. Toxins (Basel) 2017; 10:toxins10010006. [PMID: 29271898 PMCID: PMC5793093 DOI: 10.3390/toxins10010006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/19/2017] [Accepted: 12/20/2017] [Indexed: 12/12/2022] Open
Abstract
The implication of the cyanotoxin β-N-methylamino-l-alanine (BMAA) in long-lasting neurodegenerative disorders is still a matter of controversy. It has been alleged that chronic ingestion of BMAA through the food chain could be a causative agent of amyotrophic lateral sclerosis (ALS) and several related pathologies including Parkinson syndrome. Both in vitro and in vivo studies of the BMAA mode of action have focused on different molecular targets, demonstrating its toxicity to neuronal cells, especially motoneurons, and linking it to human neurodegenerative diseases. Historically, the hypothesis of BMAA-induced excitotoxicity following the stimulation of glutamate receptors has been established. However, in this paradigm, most studies have shown acute, rather than chronic effects of BMAA. More recently, the interaction of this toxin with neuromelanin, a pigment present in the nervous system, has opened a new research perspective. The issues raised by this toxin are related to its kinetics of action, and its possible incorporation into cellular proteins. It appears that BMAA neurotoxic activity involves different targets through several mechanisms known to favour the development of neurodegenerative processes.
Collapse
|
12
|
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: 2.3] [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
|
13
|
Tan VX, Lassus B, Lim CK, Tixador P, Courte J, Bessede A, Guillemin GJ, Peyrin JM. Neurotoxicity of the Cyanotoxin BMAA Through Axonal Degeneration and Intercellular Spreading. Neurotox Res 2017; 33:62-75. [PMID: 28842862 DOI: 10.1007/s12640-017-9790-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 07/20/2017] [Accepted: 07/21/2017] [Indexed: 12/12/2022]
Abstract
β-Methylamino-L-alanine (BMAA) is implicated in neurodegeneration and neurotoxicity, particularly in ALS-Parkinson Dementia Complex. Neurotoxic properties of BMAA have been partly elucidated, while its transcellular spreading capacity has not been examined. Using reconstructed neuronal networks in microfluidic chips, separating neuronal cells into two subcompartments-(1) the proximal, containing first-order neuronal soma and dendrites, and (2) a distal compartment, containing either only axons originating from first-order neurons or second-order striatal neurons-creates a cortico-striatal network. Using this system, we investigated the toxicity and spreading of BMAA in murine primary neurons. We used a newly developed antibody to detect BMAA in cells. After treatment with 10 μM BMAA, the cyanotoxin was incorporated in first-degree neurons. We also observed a rapid trans-neuronal spread of BMAA to unexposed second-degree neurons in 48 h, followed by axonal degeneration, with limited somatic death. This in vitro study demonstrates BMAA axonal toxicity at sublethal concentrations and, for the first time, the transcellular spreading abilities of BMAA. This neuronal dying forward spread that could possibly be associated with progression of some neurodegenerative diseases especially amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Vanessa X Tan
- Macquarie University Centre for MND Research, Department of Biological Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine, Adaptation Biologique et vieillissement, F-75005, Paris, France
| | - Benjamin Lassus
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine, Adaptation Biologique et vieillissement, F-75005, Paris, France
| | - Chai K Lim
- Macquarie University Centre for MND Research, Department of Biological Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Philippe Tixador
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine, Adaptation Biologique et vieillissement, F-75005, Paris, France
| | - Josquin Courte
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine, Adaptation Biologique et vieillissement, F-75005, Paris, France
| | | | - Gilles J Guillemin
- Macquarie University Centre for MND Research, Department of Biological Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.
| | - Jean-Michel Peyrin
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine, Adaptation Biologique et vieillissement, F-75005, Paris, France.
| |
Collapse
|
14
|
Caller T, Henegan P, Stommel E. The Potential Role of BMAA in Neurodegeneration. Neurotox Res 2017; 33:222-226. [PMID: 28612294 DOI: 10.1007/s12640-017-9752-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/09/2017] [Accepted: 05/12/2017] [Indexed: 01/09/2023]
Abstract
Neurodegenerative diseases are a major public health issue throughout the world with devastating effects on patients and families. Sporadic forms of neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis are generally thought to develop as a consequence of genetic susceptibility and environmental influences. A number of environmental triggers have been identified in association with amyotrophic lateral sclerosis and Parkinson's disease. We discuss the role of β-methylamino-L-alanine in the development of neurodegeneration and the potential importance of this neurotoxin as a risk for neurodegeneration.
Collapse
Affiliation(s)
- Tracie Caller
- Department of Neurology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA. .,Cheyenne Regional Medical Group, Cheyenne, WY, 82001, USA. .,Institute for Ethnomedicine, PO Box 3464, Jackson, WY, 83001, USA.
| | - Patricia Henegan
- Department of Neurology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | - Elijah Stommel
- Department of Neurology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| |
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: 8.4] [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
|
Noli B, Sanna F, Brancia C, D'Amato F, Manconi B, Vincenzoni F, Messana I, Melis MR, Argiolas A, Ferri GL, Cocco C. Profiles of VGF Peptides in the Rat Brain and Their Modulations after Phencyclidine Treatment. Front Cell Neurosci 2017. [PMID: 28626390 PMCID: PMC5454051 DOI: 10.3389/fncel.2017.00158] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
From the VGF precursor protein originate several low molecular weight peptides, whose distribution in the brain and blood circulation is not entirely known. Among the VGF peptides, those containing the N-terminus portion were altered in the cerebro-spinal fluid (CSF) and hypothalamus of schizophrenia patients. "Hence, we aimed to better investigate the involvement of the VGF peptides in schizophrenia by studying their localization in the brain regions relevant for the disease, and revealing their possible modulations in response to certain neuronal alterations occurring in schizophrenia". We produced antibodies against different VGF peptides encompassing the N-terminus, but also C-terminus-, TLQP-, GGGE- peptide sequences, and the so named NERP-3 and -4. These antibodies were used to carry out specific ELISA and immunolocalization studies while mass spectrometry (MS) analysis was also performed to recognize the intact brain VGF fragments. We used a schizophrenia rat model, in which alterations in the prepulse inhibition (PPI) of the acoustic startle response occurred after PCP treatment. In normal rats, all the VGF peptides studied were distributed in the brain areas examined including hypothalamus, prefrontal cortex, hippocampus, accumbens and amygdaloid nuclei and also in the plasma. By liquid chromatography-high resolution mass, we identified different intact VGF peptide fragments, including those encompassing the N-terminus and the NERPs. PCP treatment caused behavioral changes that closely mimic schizophrenia, estimated by us as a disruption of PPI of the acoustic startle response. The PCP treatment also induced selective changes in the VGF peptide levels within certain brain areas. Indeed, an increase in VGF C-terminus and TLQP peptides was revealed in the prefrontal cortex (p < 0.01) where they were localized within parvoalbumin and tyrosine hydroxylase (TH) containing neurons, respectively. Conversely, in the nucleus accumbens, PCP treatment produced a down-regulation in the levels of VGF C-terminus-, N-terminus- and GGGE- peptides (p < 0.01), expressed in GABAergic- (C-terminus/GGGE) and somatostatin- (N-terminus) neurons. These results confirm that VGF peptides are widely distributed in the brain and modulated in specific areas involved in schizophrenia.
Collapse
Affiliation(s)
- Barbara Noli
- Neuro-Endocrine-Fluorecence (NEF) Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| | - Fabrizio Sanna
- Neuropsychobiology Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| | - Carla Brancia
- Neuro-Endocrine-Fluorecence (NEF) Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| | - Filomena D'Amato
- Neuro-Endocrine-Fluorecence (NEF) Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| | - Barbara Manconi
- Department of Life and Environmental Sciences, University of CagliariMonserrato, Italy
| | - Federica Vincenzoni
- Institute of Biochemistry and Clinical Biochemistry, Catholic UniversityRome, Italy
| | - Irene Messana
- Institute of Chemistry of the Molecular Recognition, National Research Council (CNR)Rome, Italy
| | - Maria R Melis
- Neuropsychobiology Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| | - Antonio Argiolas
- Neuropsychobiology Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| | - Gian-Luca Ferri
- Neuro-Endocrine-Fluorecence (NEF) Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| | - Cristina Cocco
- Neuro-Endocrine-Fluorecence (NEF) Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| |
Collapse
|
17
|
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: 1.3] [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
|
18
|
Andersson M, Ersson L, Brandt I, Bergström U. Potential transfer of neurotoxic amino acid β-N-methylamino-alanine (BMAA) from mother to infant during breast-feeding: Predictions from human cell lines. Toxicol Appl Pharmacol 2017; 320:40-50. [PMID: 28174119 DOI: 10.1016/j.taap.2017.02.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/10/2017] [Accepted: 02/03/2017] [Indexed: 12/13/2022]
Abstract
β-N-methylamino-alanine (BMAA) is a non-protein amino acid produced by cyanobacteria, diatoms and dinoflagellates. BMAA has potential to biomagnify in a terrestrial food chain, and to bioaccumulate in fish and shellfish. We have reported that administration of [14C]l-BMAA to lactating mice and rats results in a mother to off-spring transfer via the milk. A preferential enantiomer-specific uptake of [14C]l-BMAA has also been demonstrated in differentiated murine mammary epithelium HC11 cells. These findings, together with neurotoxic effects of BMAA demonstrated both in vitro and in vivo, highlight the need to determine whether such transfer could also occur in humans. Here, we used four cell lines of human origin to examine and compare the transport of the two BMAA enantiomers in vitro. The uptake patterns of [14C]l- and [14C]d-BMAA in the human mammary MCF7 cell line were in agreement with the results in murine HC11 cells, suggesting a potential secretion of BMAA into human breast milk. The permeability coefficients for both [14C]l- and [14C]d-BMAA over monolayers of human intestinal Caco2 cells supported an efficient absorption from the human intestine. As a final step, transport experiments confirmed that [14C]l-and [14C]d-BMAA can be taken up by human SHSY5Y neuroblastoma cells and even more efficiently by human U343 glioblastoma cells. In competition experiments with various amino acids, the ASCT2 specific inhibitor benzylserine was the most effective inhibitor of [14C]l-BMAA uptake tested here. Altogether, our results suggest that BMAA can be transferred from an exposed mother, via the milk, to the brain of the nursed infant.
Collapse
Affiliation(s)
- Marie Andersson
- Department of Environmental Toxicology, Uppsala University, Norbyvägen 18A, SE-752 36 Uppsala, Sweden
| | - Lisa Ersson
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-751 24 Uppsala, Sweden
| | - Ingvar Brandt
- Department of Environmental Toxicology, Uppsala University, Norbyvägen 18A, SE-752 36 Uppsala, Sweden.
| | - Ulrika Bergström
- Department of Environmental Toxicology, Uppsala University, Norbyvägen 18A, SE-752 36 Uppsala, Sweden; Swedish Defence Research Agency, Division of CBRN Defence and Security, SE-164 90 Stockholm, Sweden
| |
Collapse
|
19
|
Karlsson O, Michno W, Ransome Y, Hanrieder J. MALDI imaging delineates hippocampal glycosphingolipid changes associated with neurotoxin induced proteopathy following neonatal BMAA exposure. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1865:740-746. [PMID: 27956354 DOI: 10.1016/j.bbapap.2016.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 12/05/2016] [Accepted: 12/08/2016] [Indexed: 11/16/2022]
Abstract
The environmental toxin β-N-methylamino-L-alanine (BMAA) has been proposed to contribute to neurodegenerative diseases. We have previously shown that neonatal exposure to BMAA results in dose-dependent cognitive impairments, proteomic alterations and progressive neurodegeneration in the hippocampus of adult rats. A high BMAA dose (460mg/kg) also induced intracellular fibril formation, increased protein ubiquitination and enrichment of proteins important for lipid transport and metabolism. The aim of this study was therefore to elucidate the role of neuronal lipids in BMAA-induced neurodegeneration. By using matrix assisted laser desorption/ionization imaging mass spectrometry (MALDI IMS), we characterized the spatial lipid profile in the hippocampus of six month-old rats that were treated neonatally (postnatal days 9-10) with 460mg/kg BMAA. Multivariate statistical analysis revealed long-term changes in distinct ganglioside species (GM, GD, GT) in the dentate gyrus. These changes could be a consequence of direct effects on ganglioside biosynthesis through the b-series (GM3-GD3-GD2-GD1b-GT1b) and may be linked to astrogliosis. Complementary immunohistochemistry experiments towards GFAP and S100β further verified the role of increased astrocyte activity in BMAA-induced brain damage. This highlights the potential of imaging MS for probing chemical changes associated with neuropathological mechanisms in situ. This article is part of a Special Issue entitled: MALDI Imaging, edited by Dr. Corinna Henkel and Prof. Peter Hoffmann.
Collapse
Affiliation(s)
- Oskar Karlsson
- Center for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institute, 171 76 Stockholm, Sweden; Department of Pharmaceutical Biosciences, Toxicology and Drug Safety, Uppsala University, Box 591, 751 24 Uppsala, Sweden; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Wojciech Michno
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, 431 80 Mölndal, Sweden
| | - Yusuf Ransome
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, 431 80 Mölndal, Sweden; Department of Chemistry and Chemical Engineering, Analytical Chemistry, Chalmers University of Technology, Kemivägen 10, 412 96 Gothenburg, Sweden; Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, Queen Square, WC1N 3BG London, UK.
| |
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: 72] [Impact Index Per Article: 9.0] [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
|
Andersson M, Karlsson O, Banack SA, Brandt I. Transfer of developmental neurotoxin β-N-methylamino-l-alanine (BMAA) via milk to nursed offspring: Studies by mass spectrometry and image analysis. Toxicol Lett 2016; 258:108-114. [PMID: 27320960 DOI: 10.1016/j.toxlet.2016.06.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/13/2016] [Accepted: 06/15/2016] [Indexed: 12/12/2022]
Abstract
The cyanobacterial non-proteinogenic amino acid β-N-methylamino-l-alanine (BMAA) is proposed to be involved in the etiology of amyotrophic lateral sclerosis/parkinsonism dementia complex. When administered as single doses to neonatal rats, BMAA gives rise to cognitive and neurodegenerative impairments in the adult animal. Here, we employed mass spectrometry (LC-MS/MS) and autoradiographic imaging to examine the mother-to-pup transfer of BMAA in rats. The results show that unchanged BMAA was secreted into the milk and distributed to the suckling pups. The concentration of BMAA in pup stomach milk and the neonatal liver peaked after 8h, while the concentration in the pup brain increased throughout the study period. About 1 and 6% of the BMAA recovered from adult liver and brain were released following hydrolysis, suggesting that this fraction was associated with protein. No association to milk protein was observed. Injection of rat pups with [methyl-(14)C]-l-BMAA or [carboxyl-(14)C]-l-BMAA resulted in highly similar distribution patterns, indicating no or low metabolic elimination of the methylamino- or carboxyl groups. In conclusion, BMAA is transported as a free amino acid to rat milk and suckling pups. The results strengthen the proposal that mothers' milk could be a source of exposure for BMAA in human infants.
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
|
23
|
|
24
|
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: 2.4] [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
|
25
|
Ye H, Wang J, Zhang Z, Jia C, Schmerberg C, Catherman AD, Thomas PM, Kelleher NL, Li L. Defining the Neuropeptidome of the Spiny Lobster Panulirus interruptus Brain Using a Multidimensional Mass Spectrometry-Based Platform. J Proteome Res 2015; 14:4776-91. [PMID: 26390183 DOI: 10.1021/acs.jproteome.5b00627] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Decapod crustaceans are important animal models for neurobiologists due to their relatively simple nervous systems with well-defined neural circuits and extensive neuromodulation by a diverse set of signaling peptides. However, biochemical characterization of these endogenous neuropeptides is often challenging due to limited sequence information about these neuropeptide genes and the encoded preprohormones. By taking advantage of sequence homology in neuropeptides observed in related species using a home-built crustacean neuropeptide database, we developed a semi-automated sequencing strategy to characterize the neuropeptidome of Panulirus interruptus, an important aquaculture species, with few known neuropeptide preprohormone sequences. Our streamlined process searched the high mass accuracy and high-resolution data acquired on a LTQ-Orbitrap with a flexible algorithm in ProSight that allows for sequence discrepancy from reported sequences in our database, resulting in the detection of 32 neuropeptides, including 19 novel ones. We further improved the overall coverage to 51 neuropeptides with our multidimensional platform that employed multiple analytical techniques including dimethylation-assisted fragmentation, de novo sequencing using nanoliquid chromatography-electrospray ionization-quadrupole-time-of-flight (nanoLC-ESI-Q-TOF), direct tissue analysis, and mass spectrometry imaging on matrix-assisted laser desorption/ionization (MALDI)-TOF/TOF. The high discovery rate from this unsequenced model organism demonstrated the utility of our neuropeptide discovery pipeline and highlighted the advantage of utilizing multiple sequencing strategies. Collectively, our study expands the catalog of crustacean neuropeptides and more importantly presents an approach that can be adapted to exploring neuropeptidome from species that possess limited sequence information.
Collapse
Affiliation(s)
- Hui Ye
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University , Nanjing 210009, China.,School of Pharmacy, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | | | - Zichuan Zhang
- School of Pharmacy, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Chenxi Jia
- School of Pharmacy, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Claire Schmerberg
- School of Pharmacy, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Adam D Catherman
- Departments of Chemistry and Molecular Biosciences, Proteomics Center of Excellence and Chemistry of Life Processes Institute, Northwestern University , 2145 North Sheridan Road, Evanston, Illinois 60208, United States
| | - Paul M Thomas
- Departments of Chemistry and Molecular Biosciences, Proteomics Center of Excellence and Chemistry of Life Processes Institute, Northwestern University , 2145 North Sheridan Road, Evanston, Illinois 60208, United States
| | - Neil L Kelleher
- Departments of Chemistry and Molecular Biosciences, Proteomics Center of Excellence and Chemistry of Life Processes Institute, Northwestern University , 2145 North Sheridan Road, Evanston, Illinois 60208, United States
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States.,School of Life Sciences, Tianjin University , No. 92 Weijin Road, Nankai District, Tianjin 300072, China
| |
Collapse
|
26
|
Buchberger A, Yu Q, Li L. Advances in Mass Spectrometric Tools for Probing Neuropeptides. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2015; 8:485-509. [PMID: 26070718 PMCID: PMC6314846 DOI: 10.1146/annurev-anchem-071114-040210] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Neuropeptides are important mediators in the functionality of the brain and other neurological organs. Because neuropeptides exist in a wide range of concentrations, appropriate characterization methods are needed to provide dynamic, chemical, and spatial information. Mass spectrometry and compatible tools have been a popular choice in analyzing neuropeptides. There have been several advances and challenges, both of which are the focus of this review. Discussions range from sample collection to bioinformatic tools, although avenues such as quantitation and imaging are included. Further development of the presented methods for neuropeptidomic mass spectrometric analysis is inevitable, which will lead to a further understanding of the complex interplay of neuropeptides and other signaling molecules in the nervous system.
Collapse
Affiliation(s)
- Amanda Buchberger
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706-1322;
| | - Qing Yu
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705-2222;
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706-1322;
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705-2222;
| |
Collapse
|
27
|
Jernerén F, Söderquist M, Karlsson O. Post-sampling release of free fatty acids - effects of heat stabilization and methods of euthanasia. J Pharmacol Toxicol Methods 2014; 71:13-20. [PMID: 25463283 DOI: 10.1016/j.vascn.2014.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 11/03/2014] [Indexed: 01/26/2023]
Abstract
INTRODUCTION The field of lipid research has made progress and it is now possible to study the lipidome of cells and organelles. A basic requirement of a successful lipid study is adequate pre-analytical sample handling, as some lipids can be unstable and postmortem changes can cause substantial accumulation of free fatty acids (FFAs). METHODS The aim of the present study was to investigate the effects of conductive heat stabilization and euthanasia methods on FFA levels in the rat brain and liver using liquid chromatography tandem mass spectrometry. RESULTS The analysis of brain homogenates clearly demonstrated phospholipase activity and time-dependent post-sampling changes in the lipid pool of snap frozen non-stabilized tissue. There was a significant increase in FFAs already at 2min, which continued over time. Heat stabilization was shown to be an efficient method to reduce phospholipase activity and ex vivo lipolysis. Post-sampling effects due to tissue thawing and sample preparation induced a massive release of FFAs (up to 3700%) from non-stabilized liver and brain tissues compared to heat stabilized tissue. Furthermore, the choice of euthanasia method significantly influenced the levels of FFAs in the brain. The FFAs were decreased by 15-44% in the group of animals euthanized by pentobarbital injection compared with CO2 inhalation or decapitation. DISCUSSION Our results highlight the importance of considering euthanasia methods and pre-analytical treatment in lipid analysis, factors which may otherwise interfere with the outcome of the experiments.
Collapse
Affiliation(s)
- Fredrik Jernerén
- Department of Pharmacology, University of Oxford, OX1 3QT Oxford, United Kingdom
| | | | - Oskar Karlsson
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, 17176 Stockholm, Sweden.
| |
Collapse
|
28
|
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: 2.9] [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
|
29
|
Su J, Sandor K, Sköld K, Hökfelt T, Svensson CI, Kultima K. Identification and quantification of neuropeptides in naïve mouse spinal cord using mass spectrometry reveals [des-Ser1]-cerebellin as a novel modulator of nociception. J Neurochem 2014; 130:199-214. [DOI: 10.1111/jnc.12730] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 03/13/2014] [Accepted: 04/01/2014] [Indexed: 12/31/2022]
Affiliation(s)
- Jie Su
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm Sweden
| | - Katalin Sandor
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm Sweden
| | - Karl Sköld
- Research and Development; Denator AB; Uppsala Sweden
- Department of Medical Sciences; Cancer Pharmacology and Computational Medicine; Uppsala University; Uppsala Sweden
| | - Tomas Hökfelt
- Department of Neuroscience; Karolinska Institutet; Stockholm Sweden
| | - Camilla I. Svensson
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm Sweden
| | - Kim Kultima
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm Sweden
- Department of Medical Sciences; Cancer Pharmacology and Computational Medicine; Uppsala University; Uppsala Sweden
| |
Collapse
|
30
|
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: 35] [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
|
31
|
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: 3.7] [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
|
32
|
Roman E, Karlsson O. Increased anxiety-like behavior but no cognitive impairments in adult rats exposed to constant light conditions during perinatal development. Ups J Med Sci 2013; 118:222-7. [PMID: 23902426 PMCID: PMC4190892 DOI: 10.3109/03009734.2013.821191] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Shift-work is suggested to affect fetal development negatively. In particular, maternal hormonal disturbance arising from sleep deprivation or circadian rhythm changes may disturb fetal growth or lead to complications during pregnancy. Exposure to constant light is an environmental stressor that can affect the circadian system and has been shown to induce neurochemical and behavioral changes when used during the prenatal and/or postnatal period in experimental animals. However, studies investigating long-term effects of constant light in the offspring are sparse. METHODS An accidental power outage resulted in pregnant females being housed under constant light (LL) conditions for seven days of the offspring perinatal development (embryonic day 20 to postnatal day 4). The long-term effects of constant light on the behavior in the adult offspring were assessed by means of open field, object recognition, and water maze tests. RESULTS In adulthood, LL-animals displayed an intact recognition memory and no deficits in spatial learning or memory. In the open field test, LL-animals exhibited higher anxiety-like behavior, observed as significantly more thigmotaxis and less ambulation. These results were confirmed in the other behavioral tests as the LL-animals spent less time exploring the objects in the object recognition test, and showed thigmotactic behavior also in the water maze test. CONCLUSION The results confirm that early life experience can cause changes in brain development that shape brain function and add to the sparse literature on long-term effects of constant light conditions during perinatal development on specific behaviors in adulthood.
Collapse
Affiliation(s)
- Erika Roman
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-751 24 Uppsala, Sweden
| | - Oskar Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-751 24 Uppsala, Sweden
| |
Collapse
|
33
|
Andersson M, Karlsson O, Bergström U, Brittebo EB, Brandt I. Maternal transfer of the cyanobacterial neurotoxin β-N-methylamino-L-alanine (BMAA) via milk to suckling offspring. PLoS One 2013; 8:e78133. [PMID: 24194910 PMCID: PMC3806833 DOI: 10.1371/journal.pone.0078133] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Accepted: 09/17/2013] [Indexed: 12/14/2022] Open
Abstract
The cyanobacterial neurotoxin β-N-methylamino-L-alanine (BMAA) has been implicated in the etiology of neurodegenerative disease and proposed to be biomagnified in terrestrial and aquatic food chains. We have previously shown that the neonatal period in rats, which in humans corresponds to the last trimester of pregnancy and the first few years of age, is a particularly sensitive period for exposure to BMAA. The present study aimed to examine the secretion of 14C-labeled L- and D-BMAA into milk in lactating mice and the subsequent transfer of BMAA into the developing brain. The results suggest that secretion into milk is an important elimination pathway of BMAA in lactating mothers and an efficient exposure route predominantly for L-BMAA but also for D-BMAA in suckling mice. Following secretion of [14C]L-BMAA into milk, the levels of [14C]L-BMAA in the brains of the suckling neonatal mice significantly exceeded the levels in the maternal brains. In vitro studies using the mouse mammary epithelial HC11 cell line confirmed a more efficient influx and efflux of L-BMAA than of D-BMAA in cells, suggesting enantiomer-selective transport. Competition experiments with other amino acids and a low sodium dependency of the influx suggests that the amino acid transporters LAT1 and LAT2 are involved in the transport of L-BMAA into milk. Given the persistent neurodevelopmental toxicity following injection of L-BMAA to neonatal rodent pups, the current results highlight the need to determine whether BMAA is enriched mother's and cow's milk.
Collapse
Affiliation(s)
- Marie Andersson
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden
| | - Oskar Karlsson
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Ulrika Bergström
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden
| | - Eva B. Brittebo
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Ingvar Brandt
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
34
|
Karlsson O, Bergquist J, Andersson M. Quality measures of imaging mass spectrometry aids in revealing long-term striatal protein changes induced by neonatal exposure to the cyanobacterial toxin β-N-methylamino-L-alanine (BMAA). Mol Cell Proteomics 2013; 13:93-104. [PMID: 24126143 PMCID: PMC3879633 DOI: 10.1074/mcp.m113.031435] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Many pathological processes are not directly correlated to dramatic alterations in protein levels. The changes in local concentrations of important proteins in a subset of cells or at specific loci are likely to play a significant role in disease etiologies, but the precise location might be unknown, or the concentration might be too small to be adequately sampled for traditional proteomic techniques. Matrix-assisted laser desorption ionization (MALDI) imaging mass spectrometry (IMS) is a unique analytical method that combines analysis of multiple molecular species and of their distribution in a single platform. As reproducibility is essential for successful biomarker discovery, it is important to systematically assess data quality in biologically relevant MALDI IMS experiments. In the present study, we applied four simple tools to study the reproducibility for individual sections, within-group variation, and between-group variation of data acquired from brain sections of 21 animals divided into three treatment groups. We also characterized protein changes in distinct regions of the striatum from six-month-old rats treated neonatally (postnatal days 9–10) with the cyanobacterial toxin β-N-methylamino-l-alanine (BMAA), which has been implicated in neurodegenerative diseases. The results showed that optimized experimental settings can yield high-quality MALDI IMS data with relatively low variation (14% to 15% coefficient of variance) that allow the characterization of subtle changes in protein expression in various subregions of the brain. This was further exemplified by the dose-dependent reduction of myelin basic protein in the caudate putamen and the nucleus accumbens of adult rats neonatally treated with BMAA (150 and 460 mg/kg). The reduction in myelin basic protein was confirmed through immunohistochemistry and indicates that developmental exposure to BMAA may induce structural effects on axonal growth and/or directly on the proliferation of oligodendrocytes and myelination, which might be important for the previously shown BMAA-induced long-term cognitive impairments.
Collapse
Affiliation(s)
- Oskar Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, 751 24 Uppsala, Sweden
| | | | | |
Collapse
|
35
|
Dunlop RA, Cox PA, Banack SA, Rodgers KJ. The non-protein amino acid BMAA is misincorporated into human proteins in place of L-serine causing protein misfolding and aggregation. PLoS One 2013; 8:e75376. [PMID: 24086518 PMCID: PMC3783393 DOI: 10.1371/journal.pone.0075376] [Citation(s) in RCA: 210] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 07/17/2013] [Indexed: 12/25/2022] Open
Abstract
Mechanisms of protein misfolding are of increasing interest in the aetiology of neurodegenerative diseases characterized by protein aggregation and tangles including Amyotrophic Lateral Sclerosis (ALS), Alzheimer’s disease (AD), Parkinson’s disease (PD), Lewy Body Dementia (LBD), and Progressive Supranuclear Palsy (PSP). Some forms of neurodegenerative illness are associated with mutations in genes which control assembly of disease related proteins. For example, the mouse sticky mutation sti, which results in undetected mischarging of tRNAAla with serine resulting in the substitution of serine for alanine in proteins causes cerebellar Purkinje cell loss and ataxia in laboratory animals. Replacement of serine 422 with glutamic acid in tau increases the propensity of tau aggregation associated with neurodegeneration. However, the possibility that environmental factors can trigger abnormal folding in proteins remains relatively unexplored. We here report that a non-protein amino acid, β-N-methylamino-L-alanine (BMAA), can be misincorporated in place of l-serine into human proteins. We also report that this misincorporation can be inhibited by l-serine. Misincorporation of BMAA into human neuroproteins may shed light on putative associations between human exposure to BMAA produced by cyanobacteria and an increased incidence of ALS.
Collapse
Affiliation(s)
- Rachael Anne Dunlop
- Cell Biology Group, School of Medical and Molecular Biosciences, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Paul Alan Cox
- Institute for Ethnomedicine, Jackson Hole, Wyoming, United States of America
| | - Sandra Anne Banack
- Institute for Ethnomedicine, Jackson Hole, Wyoming, United States of America
| | - Kenneth John Rodgers
- Cell Biology Group, School of Medical and Molecular Biosciences, University of Technology Sydney, Ultimo, New South Wales, Australia
- * E-mail:
| |
Collapse
|
36
|
Craft GE, Chen A, Nairn AC. Recent advances in quantitative neuroproteomics. Methods 2013; 61:186-218. [PMID: 23623823 PMCID: PMC3891841 DOI: 10.1016/j.ymeth.2013.04.008] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Revised: 03/29/2013] [Accepted: 04/13/2013] [Indexed: 01/07/2023] Open
Abstract
The field of proteomics is undergoing rapid development in a number of different areas including improvements in mass spectrometric platforms, peptide identification algorithms and bioinformatics. In particular, new and/or improved approaches have established robust methods that not only allow for in-depth and accurate peptide and protein identification and modification, but also allow for sensitive measurement of relative or absolute quantitation. These methods are beginning to be applied to the area of neuroproteomics, but the central nervous system poses many specific challenges in terms of quantitative proteomics, given the large number of different neuronal cell types that are intermixed and that exhibit distinct patterns of gene and protein expression. This review highlights the recent advances that have been made in quantitative neuroproteomics, with a focus on work published over the last five years that applies emerging methods to normal brain function as well as to various neuropsychiatric disorders including schizophrenia and drug addiction as well as of neurodegenerative diseases including Parkinson's disease and Alzheimer's disease. While older methods such as two-dimensional polyacrylamide electrophoresis continued to be used, a variety of more in-depth MS-based approaches including both label (ICAT, iTRAQ, TMT, SILAC, SILAM), label-free (label-free, MRM, SWATH) and absolute quantification methods, are rapidly being applied to neurobiological investigations of normal and diseased brain tissue as well as of cerebrospinal fluid (CSF). While the biological implications of many of these studies remain to be clearly established, that there is a clear need for standardization of experimental design and data analysis, and that the analysis of protein changes in specific neuronal cell types in the central nervous system remains a serious challenge, it appears that the quality and depth of the more recent quantitative proteomics studies is beginning to shed light on a number of aspects of neuroscience that relates to normal brain function as well as of the changes in protein expression and regulation that occurs in neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- George E Craft
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06508
| | - Anshu Chen
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06508
| | - Angus C Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06508
- Yale/NIDA Neuroproteomics Center, Yale University School of Medicine, New Haven, CT, 06508
| |
Collapse
|