1
|
Pu L, Steele JR, Phillips CR, Violi JP, Rodgers KJ. The cyanobacterial toxins BMAA and 2,4-DAB perturb the l-serine biosynthesis pathway and induce systemic changes in energy metabolism in human neuroblastoma cells: A proteomic study. Toxicol In Vitro 2025; 106:106058. [PMID: 40157434 DOI: 10.1016/j.tiv.2025.106058] [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: 11/18/2024] [Revised: 03/06/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Blue-green algae (cyanobacteria), an ancient phylum of bacteria, produce a wide array of secondary metabolites that are toxic to humans. Rapid growth of cyanobacteria in an aquatic environment can result in algal blooms capable of turning waterways green and increasing toxin levels in the environment. Cyanobacterial toxins were first linked to the high incidence of a complex neurodegenerative disorder reported on the island of Guam in the 1940s but more recently have been linked to clusters of sporadic amyotrophic lateral sclerosis (sALS) worldwide. The non-protein amino acid β-N-methylamino-L-alanine (BMAA) and its isomer L-2,4-diaminobutyric acid (2,4-DAB) are produced concurrently by most cyanobacterial species. We carried out proteomic analysis on human neuroblastoma cells treated with BMAA and 2,4-DAB to determine the underlying mechanisms of toxicity resulting from exposure to these cyanotoxins and identified significant changes in the l-serine biosynthesis pathway as well as pathways associated with energy production in the cell such as fatty acid ß-oxidation and glycolysis. The impact on the serine biosynthetic pathway was supported by demonstrating a significant decrease in both mRNA and protein levels of the enzyme 3-phosphoglycerate dehydrogenase (PHGDH) the first committed step in serine biosynthesis. PHGDH uses 3-phospho-D-glycerate (3PG) an intermediate in the glycolytic pathway as a substrate, and co-incubation of cells with l-serine restored expression levels of PHGDH as did cell pre-treatment with the glycolytic product pyruvate. This is the first study to link exposure to BMAA and 2,4-DAB to impairments in the l-serine biosynthesis pathway and broad disturbances in energy metabolism.
Collapse
Affiliation(s)
- Lisa Pu
- Neurotoxin Research Group, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Joel R Steele
- Monash Proteomics and Metabolomics Facility, Monash University, Melbourne, VIC 3800, Australia
| | - Connor R Phillips
- Neurotoxin Research Group, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Jake P Violi
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Kenneth J Rodgers
- Neurotoxin Research Group, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| |
Collapse
|
2
|
Violi JP, Pu L, Pravadali-Cekic S, Bishop DP, Phillips CR, Rodgers KJ. Effects of the Toxic Non-Protein Amino Acid β-Methylamino-L-Alanine (BMAA) on Intracellular Amino Acid Levels in Neuroblastoma Cells. Toxins (Basel) 2023; 15:647. [PMID: 37999510 PMCID: PMC10674354 DOI: 10.3390/toxins15110647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
The cyanobacterial non-protein amino acid (AA) β-Methylamino-L-alanine (BMAA) is considered to be a neurotoxin. BMAA caused histopathological changes in brains and spinal cords of primates consistent with some of those seen in early motor neuron disease; however, supplementation with L-serine protected against some of those changes. We examined the impact of BMAA on AA concentrations in human neuroblastoma cells in vitro. Cells were treated with 1000 µM BMAA and intracellular free AA concentrations in treated and control cells were compared at six time-points over a 48 h culture period. BMAA had a profound effect on intracellular AA levels at specific time points but in most cases, AA homeostasis was re-established in the cell. The most heavily impacted amino acid was serine which was depleted in BMAA-treated cells from 9 h onwards. Correction of serine depletion could be a factor in the observation that supplementation with L-serine protects against BMAA toxicity in vitro and in vivo. AAs that could potentially be involved in protection against BMAA-induced oxidation such as histidine, tyrosine, and phenylalanine were depleted in cells at later time points.
Collapse
Affiliation(s)
- Jake P. Violi
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW 2007, Australia; (J.P.V.); (L.P.); (C.R.P.)
| | - Lisa Pu
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW 2007, Australia; (J.P.V.); (L.P.); (C.R.P.)
| | - Sercan Pravadali-Cekic
- School of Mathematical and Physical Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW 2007, Australia (D.P.B.)
| | - David P. Bishop
- School of Mathematical and Physical Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW 2007, Australia (D.P.B.)
| | - Connor R. Phillips
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW 2007, Australia; (J.P.V.); (L.P.); (C.R.P.)
| | - Kenneth J. Rodgers
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW 2007, Australia; (J.P.V.); (L.P.); (C.R.P.)
| |
Collapse
|
3
|
Anzilotti S, Valente V, Brancaccio P, Franco C, Casamassa A, Lombardi G, Palazzi A, Conte A, Paladino S, Canzoniero LMT, Annunziato L, Pierantoni GM, Pignataro G. Chronic exposure to l-BMAA cyanotoxin induces cytoplasmic TDP-43 accumulation and glial activation, reproducing an amyotrophic lateral sclerosis-like phenotype in mice. Biomed Pharmacother 2023; 167:115503. [PMID: 37729728 DOI: 10.1016/j.biopha.2023.115503] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/24/2023] [Accepted: 09/12/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a progressive and often fatal neurodegenerative disease characterized by the loss of Motor Neurons (MNs) in spinal cord, motor cortex and brainstem. Despite significant efforts in the field, the exact pathogenetic mechanisms underlying both familial and sporadic forms of ALS have not been fully elucidated, and the therapeutic possibilities are still very limited. Here we investigate the molecular mechanisms of neurodegeneration induced by chronic exposure to the environmental cyanotoxin L-BMAA, which causes a form of ALS/Parkinson's disease (PD) in several populations consuming food and/or water containing high amounts of this compound. METHODS In this effort, mice were chronically exposed to L-BMAA and analyzed at different time points to evaluate cellular and molecular alterations and behavioral deficits, performing MTT assay, immunoblot, immunofluorescence and immunohistochemistry analysis, and behavioral tests. RESULTS We found that cyanotoxin L-BMAA determines apoptotic cell death and a marked astrogliosis in spinal cord and motor cortex, and induces neurotoxicity by favoring TDP-43 cytoplasmic accumulation. CONCLUSIONS Overall, our results characterize a new versatile neurotoxic animal model of ALS that may be useful for the identification of new druggable targets to develop innovative therapeutic strategies for this disease.
Collapse
Affiliation(s)
- Serenella Anzilotti
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy
| | - Valeria Valente
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, "Federico II" University of Naples, Italy
| | - Paola Brancaccio
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, Italy
| | - Cristina Franco
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy
| | | | - Giovanna Lombardi
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy
| | - Alessandra Palazzi
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, "Federico II" University of Naples, Italy
| | - Andrea Conte
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, "Federico II" University of Naples, Italy
| | - Simona Paladino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, "Federico II" University of Naples, Italy
| | | | | | - Giovanna Maria Pierantoni
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, "Federico II" University of Naples, Italy.
| | - Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, Italy.
| |
Collapse
|
4
|
Weeks RD, Banack SA, Howell S, Thunga P, Metcalf JS, Green AJ, Cox PA, Planchart A. The Effects of Long-term, Low-dose β-N-methylamino-L-alanine (BMAA) Exposures in Adult SOD G93R Transgenic Zebrafish. Neurotox Res 2023; 41:481-495. [PMID: 37552461 PMCID: PMC11216512 DOI: 10.1007/s12640-023-00658-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 06/07/2023] [Accepted: 07/01/2023] [Indexed: 08/09/2023]
Abstract
β-N-Methylamino-L-alanine (BMAA) is a non-proteinogenic amino acid produced by cyanobacteria, which has been implicated in several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). It is postulated that chronic exposure to BMAA can lead to formation of protein aggregates, oxidative stress, and/or excitotoxicity, which are mechanisms involved in the etiology of ALS. While specific genetic mutations are identified in some instances of ALS, it is likely that a combination of genetic and environmental factors, such as exposure to the neurotoxin BMAA, contributes to disease. We used a transgenic zebrafish with an ALS-associated mutation, compared with wild-type fish to explore the potential neurotoxic effects of BMAA through chronic long-term exposures. While our results revealed low concentrations of BMAA in the brains of exposed fish, we found no evidence of decreased swim performance or behavioral differences that might be reflective of neurodegenerative disease. Further research is needed to determine if chronic BMAA exposure in adult zebrafish is a suitable model to study neurodegenerative disease initiation and/or progression.
Collapse
Affiliation(s)
- Ryan D. Weeks
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
- Program in Toxicology, North Carolina State University, Raleigh, NC 27695, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
| | - Sandra A. Banack
- Brain Chemistry Labs, Institute for Ethnomedicine, Box 3464, Jackson, WY 83001, USA
| | - Shaunacee Howell
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Preethi Thunga
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - James S. Metcalf
- Brain Chemistry Labs, Institute for Ethnomedicine, Box 3464, Jackson, WY 83001, USA
| | - Adrian J. Green
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
- Program in Toxicology, North Carolina State University, Raleigh, NC 27695, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
| | - Paul A. Cox
- Brain Chemistry Labs, Institute for Ethnomedicine, Box 3464, Jackson, WY 83001, USA
| | - Antonio Planchart
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
- Program in Toxicology, North Carolina State University, Raleigh, NC 27695, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
5
|
Courtier A, Potheret D, Giannoni P. Environmental bacteria as triggers to brain disease: Possible mechanisms of toxicity and associated human risk. Life Sci 2022; 304:120689. [DOI: 10.1016/j.lfs.2022.120689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/11/2022] [Accepted: 06/01/2022] [Indexed: 11/24/2022]
|
6
|
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: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/27/2022] Open
Abstract
Harmful algal blooms (HAB), and the consequent release of toxic metabolites, can be responsible for seafood poisoning outbreaks. Marine wildlife can accumulate these toxins throughout the food chain, which presents a threat to consumers’ health. Some of these toxins, such as saxitoxin (STX), domoic acid (DA), ciguatoxin (CTX), brevetoxin (BTX), tetrodotoxin (TTX), and β-N-methylamino-L-alanine (BMAA), cause severe neurological symptoms in humans. Considerable information is missing, however, notably the consequences of toxin exposures on changes in gene expression, protein profile, and metabolic pathways. This information could lead to understanding the consequence of marine neurotoxin exposure in aquatic organisms and humans. Nevertheless, recent contributions to the knowledge of neurotoxins arise from OMICS-based research, such as genomics, transcriptomics, proteomics, and metabolomics. This review presents a comprehensive overview of the most recent research and of the available solutions to explore OMICS datasets in order to identify new features in terms of ecotoxicology, food safety, and human health. In addition, future perspectives in OMICS studies are discussed.
Collapse
|
7
|
Zakharova MN, Bakulin IS, Abramova AA. Toxic Damage to Motor Neurons. NEUROCHEM J+ 2021. [DOI: 10.1134/s1819712421040164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Abstract—Amyotrophic lateral sclerosis (ALS) is a multifactor disease in the development of which both genetic and environmental factors play a role. Specifically, the effects of organic and inorganic toxic substances can result in an increased risk of ALS development and the acceleration of disease progression. It was described that some toxins can induce potentially curable ALS-like syndromes. In this case, the specific treatment for the prevention of the effects of the toxic factor may result in positive clinical dynamics. In this article, we review the main types of toxins that can damage motor neurons in the brain and spinal cord leading to the development of the clinical manifestation of ALS, briefly present historical data on studies on the role of toxic substances, and describe the main mechanisms of the pathogenesis of motor neuron disease associated with their action.
Collapse
|
8
|
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: 20] [Impact Index Per Article: 4.0] [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
|
9
|
Metabolism of the neurotoxic amino acid β-N-methylamino-L-alanine in human cell culture models. Toxicon 2019; 168:131-139. [PMID: 31330193 DOI: 10.1016/j.toxicon.2019.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 12/12/2022]
Abstract
Human dietary exposure to the environmental neurotoxin β-N-methylamino-L-alanine (BMAA) has been implicated in an increased risk of developing sporadic neurodegenerative diseases like Alzheimer's and amyotrophic lateral sclerosis. Evidence suggests that humans are exposed to BMAA globally, but very little is known about BMAA metabolism in mammalian systems, let alone in humans. The most plausible, evidence-based mechanisms of BMAA toxicity rely on the metabolic stability of the amino acid and that, following ingestion, it enters the circulatory system unmodified. BMAA crosses from the intestinal lumen into the circulatory system, and the small intestine and liver are the first sites for dietary amino acid metabolism. Both tissues have substantial amino acid metabolic needs, which are largely fulfilled by dietary amino acids. Metabolism of BMAA in these tissues has been largely overlooked, yet is important in gauging the true human exposure risk. Here we investigate the potential for BMAA metabolism by the human liver and small intestine, using in vitro cell systems. Data show that BMAA metabolism via common proteinogenic amino acid metabolic pathways is negligible, and that in the presence of other amino acids cellular uptake of BMAA is substantially reduced. These data suggest that the majority of ingested BMAA remains unmodified following passage through the small intestine and liver. This not only supports oral BMAA exposure as a plausible exposure route to toxic doses of BMAA, but also supports previous notions that protein deficient diets or malnutrition may increase an individual's susceptibility to BMAA absorption and subsequent toxicity.
Collapse
|
10
|
Andersson M, Karlsson O, Brandt I. The environmental neurotoxin β-N-methylamino-l-alanine (l-BMAA) is deposited into birds' eggs. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 147:720-724. [PMID: 28942274 DOI: 10.1016/j.ecoenv.2017.09.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/08/2017] [Accepted: 09/12/2017] [Indexed: 06/07/2023]
Abstract
The neurotoxic amino acid β-N-methylamino-L-alanine (BMAA) has been implicated in the etiology of neurodegenerative disorders. BMAA is also a known developmental neurotoxin and research indicates that the sources of human and wildlife exposure may be more diverse than previously anticipated. The aim of the present study was therefore to examine whether BMAA can be transferred into birds' eggs. Egg laying quail were dosed with 14C-labeled BMAA. The distribution of radioactivity in the birds and their laid eggs was then examined at different time points by autoradiography and phosphoimaging analysis. To evaluate the metabolic stability of the BMAA molecule, the distribution of 14C-methyl- and 14C-carboxyl-labeled BMAA were compared. The results revealed a pronounced incorporation of radioactivity in the eggs, predominantly in the yolk but also in the albumen. Imaging analysis showed that the concentrations of radioactivity in the liver decreased about seven times between the 24h and the 72h time points, while the concentrations in egg yolk remained largely unchanged. At 72h the egg yolk contained about five times the concentration of radioactivity in the liver. Both BMAA preparations gave rise to similar distribution pattern in the bird tissues and in the eggs, indicating metabolic stability of the labeled groups. The demonstrated deposition into eggs warrants studies of BMAAs effects on bird development. Moreover, birds' eggs may be a source of human BMAA exposure, provided that the laying birds are exposed to BMAA via their diet.
Collapse
Affiliation(s)
- Marie Andersson
- Department of Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Oskar Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, SE-751 24 Uppsala, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, SE-17176 Stockholm, Sweden
| | - Ingvar Brandt
- Department of Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden.
| |
Collapse
|
11
|
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: 23] [Impact Index Per Article: 2.9] [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
|
12
|
Engskog MKR, Ersson L, Haglöf J, Arvidsson T, Pettersson C, Brittebo E. β-N-Methylamino-L-alanine (BMAA) perturbs alanine, aspartate and glutamate metabolism pathways in human neuroblastoma cells as determined by metabolic profiling. Amino Acids 2017; 49:905-919. [PMID: 28161796 PMCID: PMC5383692 DOI: 10.1007/s00726-017-2391-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/28/2017] [Indexed: 12/15/2022]
Abstract
β-Methylamino-L-alanine (BMAA) is a non-proteinogenic amino acid that induces long-term cognitive deficits, as well as an increased neurodegeneration and intracellular fibril formation in the hippocampus of adult rodents following short-time neonatal exposure and in vervet monkey brain following long-term exposure. It has also been proposed to be involved in the etiology of neurodegenerative disease in humans. The aim of this study was to identify metabolic effects not related to excitotoxicity or oxidative stress in human neuroblastoma SH-SY5Y cells. The effects of BMAA (50, 250, 1000 µM) for 24 h on cells differentiated with retinoic acid were studied. Samples were analyzed using LC-MS and NMR spectroscopy to detect altered intracellular polar metabolites. The analysis performed, followed by multivariate pattern recognition techniques, revealed significant perturbations in protein biosynthesis, amino acid metabolism pathways and citrate cycle. Of specific interest were the BMAA-induced alterations in alanine, aspartate and glutamate metabolism and as well as alterations in various neurotransmitters/neuromodulators such as GABA and taurine. The results indicate that BMAA can interfere with metabolic pathways involved in neurotransmission in human neuroblastoma cells.
Collapse
Affiliation(s)
- Mikael K R Engskog
- Division of Analytical Pharmaceutical Chemistry, Uppsala University, Box 574, 751 23, Uppsala, Sweden.
| | - Lisa Ersson
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, 751 23, Uppsala, Sweden
| | - Jakob Haglöf
- Division of Analytical Pharmaceutical Chemistry, Uppsala University, Box 574, 751 23, Uppsala, Sweden
| | - Torbjörn Arvidsson
- Division of Analytical Pharmaceutical Chemistry, Uppsala University, Box 574, 751 23, Uppsala, Sweden.,Medical Product Agency, Box 26, Dag Hammarskjölds väg 42, 751 03, Uppsala, Sweden
| | - Curt Pettersson
- Division of Analytical Pharmaceutical Chemistry, Uppsala University, Box 574, 751 23, Uppsala, Sweden
| | - Eva Brittebo
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, 751 23, Uppsala, Sweden
| |
Collapse
|
13
|
Kruk J, Doskocz M, Jodłowska E, Zacharzewska A, Łakomiec J, Czaja K, Kujawski J. NMR Techniques in Metabolomic Studies: A Quick Overview on Examples of Utilization. APPLIED MAGNETIC RESONANCE 2017; 48:1-21. [PMID: 28111499 PMCID: PMC5222922 DOI: 10.1007/s00723-016-0846-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/10/2016] [Indexed: 05/08/2023]
Abstract
Metabolomics is a rapidly developing branch of science that concentrates on identifying biologically active molecules with potential biomarker properties. To define the best biomarkers for diseases, metabolomics uses both models (in vitro, animals) and human, as well as, various techniques such as mass spectroscopy, gas chromatography, liquid chromatography, infrared and UV-VIS spectroscopy and nuclear magnetic resonance. The last one takes advantage of the magnetic properties of certain nuclei, such as 1H, 13C, 31P, 19F, especially their ability to absorb and emit energy, what is crucial for analyzing samples. Among many spectroscopic NMR techniques not only one-dimensional (1D) techniques are known, but for many years two-dimensional (2D, for example, COSY, DOSY, JRES, HETCORE, HMQS), three-dimensional (3D, DART-MS, HRMAS, HSQC, HMBC) and solid-state NMR have been used. In this paper, authors taking apart fundamental division of nuclear magnetic resonance techniques intend to shown their wide application in metabolomic studies, especially in identifying biomarkers.
Collapse
Affiliation(s)
- Joanna Kruk
- Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Grunwaldzka 6 Str., 60-780 Poznan, Poland
| | - Marek Doskocz
- RootInnovation Sp. z o.o., Jana Matejki 11 Str., 50-333 Wrocław, Poland
| | - Elżbieta Jodłowska
- Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Grunwaldzka 6 Str., 60-780 Poznan, Poland
| | - Anna Zacharzewska
- Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Grunwaldzka 6 Str., 60-780 Poznan, Poland
| | - Joanna Łakomiec
- Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Grunwaldzka 6 Str., 60-780 Poznan, Poland
| | - Kornelia Czaja
- Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Grunwaldzka 6 Str., 60-780 Poznan, Poland
| | - Jacek Kujawski
- Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Grunwaldzka 6 Str., 60-780 Poznan, Poland
- Foundation for Development of Science and Business on Medical and Exact Sciences Area, Legnicka 65 Str., 54-206 Wrocław, Poland
| |
Collapse
|
14
|
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: 1.3] [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
|
15
|
Novak M, Hercog K, Žegura B. Assessment of the mutagenic and genotoxic activity of cyanobacterial toxin beta-N-methyl-amino-L-alanine in Salmonella typhimurium. Toxicon 2016; 118:134-40. [DOI: 10.1016/j.toxicon.2016.04.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 04/27/2016] [Accepted: 04/28/2016] [Indexed: 11/26/2022]
|
16
|
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: 21] [Impact Index Per Article: 2.3] [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
|
17
|
Blasco H, Patin F, Madji Hounoum B, Gordon PH, Vourc'h P, Andres CR, Corcia P. Metabolomics in amyotrophic lateral sclerosis: how far can it take us? Eur J Neurol 2016; 23:447-54. [PMID: 26822316 DOI: 10.1111/ene.12956] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 12/04/2015] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common adult-onset motor neuron disease. Alongside identification of aetiologies, development of biomarkers is a foremost research priority. Metabolomics is one promising approach that is being utilized in the search for diagnosis and prognosis markers. Our aim is to provide an overview of the principal research in metabolomics applied to ALS. References were identified using PubMed with the terms 'metabolomics' or 'metabolomic' and 'ALS' or 'amyotrophic lateral sclerosis' or 'MND' or 'motor neuron disorders'. To date, nine articles have reported metabolomics research in patients and a few additional studies examined disease physiology and drug effects in patients or models. Metabolomics contribute to a better understanding of ALS pathophysiology but, to date, no biomarker has been validated for diagnosis, principally due to the heterogeneity of the disease and the absence of applied standardized methodology for biomarker discovery. A consensus on best metabolomics methodology as well as systematic independent validation will be an important accomplishment on the path to identifying the long-awaited biomarkers for ALS and to improve clinical trial designs.
Collapse
Affiliation(s)
- H Blasco
- Inserm U930, Tours, France
- Université François-Rabelais, Tours, France
- Laboratoire de Biochimie et Biologie Moléculaire, CHRU de Tours, Tours, France
| | - F Patin
- Inserm U930, Tours, France
- Université François-Rabelais, Tours, France
- Laboratoire de Biochimie et Biologie Moléculaire, CHRU de Tours, Tours, France
| | - B Madji Hounoum
- Inserm U930, Tours, France
- Université François-Rabelais, Tours, France
| | - P H Gordon
- Northern Navajo Medical Center, Shiprock, NM, USA
| | - P Vourc'h
- Inserm U930, Tours, France
- Université François-Rabelais, Tours, France
- Laboratoire de Biochimie et Biologie Moléculaire, CHRU de Tours, Tours, France
| | - C R Andres
- Inserm U930, Tours, France
- Université François-Rabelais, Tours, France
- Laboratoire de Biochimie et Biologie Moléculaire, CHRU de Tours, Tours, France
| | - P Corcia
- Inserm U930, Tours, France
- Université François-Rabelais, Tours, France
- Centre SLA, Service de Neurologie, CHRU Bretonneau, Tours, France
| |
Collapse
|
18
|
Tian KW, Jiang H, Wang BB, Zhang F, Han S. Intravenous injection of l-BMAA induces a rat model with comprehensive characteristics of amyotrophic lateral sclerosis/Parkinson-dementia complex. Toxicol Res (Camb) 2015; 5:79-96. [PMID: 30090328 DOI: 10.1039/c5tx00272a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 11/02/2015] [Indexed: 12/13/2022] Open
Abstract
Non-protein amino acid beta-N-methylamino-l-alanine (l-BMAA) is a neurotoxin that was associated with the high incidence of Amyotrophic Lateral Sclerosis/Parkinson-Dementia Complex (ALS/PDC) in Guam. This neurotoxin has been implicated as a potential environmental factor in amyotrophic lateral sclerosis, Alzheimer's disease and other neurodegenerative diseases, and was found to accumulate in brain tissues of ALS/PDC patients. It is extremely important to establish a reliable animal model that has the comprehensive characteristics of ALS/PDC for studying mechanisms underlying neurodegeneration, and exploring effective therapies. However, very few good animal models that mimic ALS/PDC have been established. In this study, an ideal rat model that mimicked most characteristics of ALS/PDC was established by administering continuous intravenous (i.v.) injections of neurotoxic l-BMAA. Based on the data obtained, it was demonstrated that continuous i.v. injections of l-BMAA induced mitochondrial morphology and structural changes, astrogliosis, motor neuronal death, and other relative functional changes, which led to the overexpression of pro-inflammatory cytokines cyclooxygenase-2 (COX-2), nuclear factor kappa B (NF-κB) and tumor necrosis factor-alpha (TNF-α), and resulted in the upregulation of glycogen synthase kinase-3 (GSK3), downregulation of astrocytic glutamate transporter-1 (GLT-1), accumulation of microtubule-associated protein tau and cytosolic aggregates of TAR DNA-binding protein-43 (TDP-43) in degenerating motor neurons. These results suggest that this model could be used as a useful tool for the mechanistic and therapeutic study of ALS/PDC.
Collapse
Affiliation(s)
- Ke-Wei Tian
- Institute of Anatomy and Cell Biology , Medical College , Zhejiang University , Hangzhou 310058 , China . ; ; Tel: +86-571-88208160
| | - Hong Jiang
- Department of Electrophysiology , Sir Run Run Shaw Hospital , Medical College , Zhejiang University , Hangzhou 310058 , China
| | - Bei-Bei Wang
- Core Facilities , Zhejiang University School of Medicine , Hangzhou 310058 , China
| | - Fan Zhang
- Institute of Anatomy and Cell Biology , Medical College , Zhejiang University , Hangzhou 310058 , China . ; ; Tel: +86-571-88208160
| | - Shu Han
- Institute of Anatomy and Cell Biology , Medical College , Zhejiang University , Hangzhou 310058 , China . ; ; Tel: +86-571-88208160
| |
Collapse
|
19
|
Environmental neurotoxin interaction with proteins: Dose-dependent increase of free and protein-associated BMAA (β-N-methylamino-L-alanine) in neonatal rat brain. Sci Rep 2015; 5:15570. [PMID: 26498001 PMCID: PMC4620439 DOI: 10.1038/srep15570] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/25/2015] [Indexed: 12/12/2022] Open
Abstract
β-Methylamino-L-alanine (BMAA) is implicated in the aetiology of neurodegenerative disorders. Neonatal exposure to BMAA induces cognitive impairments and progressive neurodegenerative changes including intracellular fibril formation in the hippocampus of adult rats. It is unclear why the neonatal hippocampus is especially vulnerable and the critical cellular perturbations preceding BMAA-induced toxicity remains to be elucidated. The aim of this study was to compare the level of free and protein-associated BMAA in neonatal rat brain and peripheral tissues after different exposures to BMAA. Ultra-high performance liquid chromatography-tandem mass spectrometry analysis revealed that BMAA passed the neonatal blood-brain barrier and was distributed to all studied brain areas. BMAA was also associated to proteins in the brain, especially in the hippocampus. The level in the brain was, however, considerably lower compared to the liver that is not a target organ for BMAA. In contrast to the liver there was a significantly increased level of protein-association of BMAA in the hippocampus and other brain areas following repeated administration suggesting that the degradation of BMAA-associated proteins may be lower in neonatal brain than in the liver. Additional evidence is needed in support of a role for protein misincorporation in the neonatal hippocampus for long-term effects of BMAA.
Collapse
|
20
|
Muñoz-Sáez E, de Munck García E, Arahuetes Portero RM, Martínez A, Solas Alados MT, Miguel BG. Analysis of β-N-methylamino-L-alanine (L-BMAA) neurotoxicity in rat cerebellum. Neurotoxicology 2015; 48:192-205. [PMID: 25898785 DOI: 10.1016/j.neuro.2015.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 12/13/2022]
Abstract
Due to its structural similarity to glutamate, L-BMAA could be a trigger for neurodegenerative disorders caused by changes in the intracellular medium, such as increased oxidative stress, mitochondrial dysfunction, impaired synthesis and protein degradation and the imbalance of some enzymes. It is also important to note that according to some published studies, L-BMAA will be incorporated into proteins, causing the alteration of protein homeostasis. Neuronal cells are particularly prone to suffer damage in protein folding and protein accumulation because they have not performed cellular division. In this work, we will analyse the cerebellum impairment triggered by L-BMAA in treated rats. The cerebellum is one of the most important subcortical motor centres and ensures that movements are performed with spatial and temporal precision. Cerebellum damage caused by L-BMAA can contribute to motor impairment. To characterize this neurodegenerative pathology, we first carried out ultrastructure analysis in Purkinje cells showing altered mitochondria, endoplasmic reticulum (ER), and Golgi apparatus (GA). We then performed biochemical assays of GSK3 and TDP-43 in cerebellum, obtaining an increase of both biomarkers with L-BMAA treatment and, finally, performed autophagy studies that revealed a higher level of these processes after treatment. This work provides evidence of cerebellar damage in rats after treatment with L-BMAA. Three months after treatment, affected rats cannot restore the normal functions of the cerebellum regarding motor coordination and postural control.
Collapse
Affiliation(s)
- Emma Muñoz-Sáez
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | | | | | - Ana Martínez
- Instituto de Química Médica - Centro Superior de Investigaciones Científicas, 28006 Madrid, Spain
| | - Ma Teresa Solas Alados
- Departamento de Biología Celular, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Begoña Gómez Miguel
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense de Madrid, 28040 Madrid, Spain
| |
Collapse
|
21
|
Delzor A, Couratier P, Boumédiène F, Nicol M, Druet-Cabanac M, Paraf F, Méjean A, Ploux O, Leleu JP, Brient L, Lengronne M, Pichon V, Combès A, El Abdellaoui S, Bonneterre V, Lagrange E, Besson G, Bicout DJ, Boutonnat J, Camu W, Pageot N, Juntas-Morales R, Rigau V, Masseret E, Abadie E, Preux PM, Marin B. Searching for a link between the L-BMAA neurotoxin and amyotrophic lateral sclerosis: a study protocol of the French BMAALS programme. BMJ Open 2014; 4:e005528. [PMID: 25180055 PMCID: PMC4156816 DOI: 10.1136/bmjopen-2014-005528] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is the most common motor neurone disease. It occurs in two forms: (1) familial cases, for which several genes have been identified and (2) sporadic cases, for which various hypotheses have been formulated. Notably, the β-N-methylamino-L-alanine (L-BMAA) toxin has been postulated to be involved in the occurrence of sporadic ALS. The objective of the French BMAALS programme is to study the putative link between L-BMAA and ALS. METHODS AND ANALYSIS The programme covers the period from 1 January 2003 to 31 December 2011. Using multiple sources of ascertainment, all the incident ALS cases diagnosed during this period in the area under study (10 counties spread over three French regions) were collected. First, the standardised incidence ratio will be calculated for each municipality under concern. Then, by applying spatial clustering techniques, overincidence and underincidence zones of ALS will be sought. A case-control study, in the subpopulation living in the identified areas, will gather information about patients' occupations, leisure activities and lifestyle habits in order to assess potential risk factors to which they are or have been exposed. Specimens of drinking water, food and biological material (brain tissue) will be examined to assess the presence of L-BMAA in the environment and tissues of ALS cases and controls. ETHICS AND DISSEMINATION The study has been reviewed and approved by the French ethical committee of the CPP SOOM IV (Comité de Protection des Personnes Sud-Ouest & Outre-Mer IV). The results will be published in peer-reviewed journals and presented at national and international conferences.
Collapse
Affiliation(s)
- Aurélie Delzor
- Tropical Neuroepidemiology, INSERM UMR 1094, Limoges, France
- University of Limoges, School of Medicine, Institute of Neuroepidemiology and Tropical Neurology, Centre national de la recherche scientifique FR 3503 GEIST, Limoges, France
| | - Philippe Couratier
- Tropical Neuroepidemiology, INSERM UMR 1094, Limoges, France
- University of Limoges, School of Medicine, Institute of Neuroepidemiology and Tropical Neurology, Centre national de la recherche scientifique FR 3503 GEIST, Limoges, France
- Department of Neurology, ALS Center, University Hospital Dupuytren, Limoges, France
| | - Farid Boumédiène
- Tropical Neuroepidemiology, INSERM UMR 1094, Limoges, France
- University of Limoges, School of Medicine, Institute of Neuroepidemiology and Tropical Neurology, Centre national de la recherche scientifique FR 3503 GEIST, Limoges, France
| | - Marie Nicol
- Tropical Neuroepidemiology, INSERM UMR 1094, Limoges, France
- University of Limoges, School of Medicine, Institute of Neuroepidemiology and Tropical Neurology, Centre national de la recherche scientifique FR 3503 GEIST, Limoges, France
- Department of Neurology, ALS Center, University Hospital Dupuytren, Limoges, France
| | - Michel Druet-Cabanac
- Tropical Neuroepidemiology, INSERM UMR 1094, Limoges, France
- University of Limoges, School of Medicine, Institute of Neuroepidemiology and Tropical Neurology, Centre national de la recherche scientifique FR 3503 GEIST, Limoges, France
- Department of Neurology, ALS Center, University Hospital Dupuytren, Limoges, France
| | - François Paraf
- Department of Neurology, ALS Center, University Hospital Dupuytren, Limoges, France
| | - Annick Méjean
- Interdisciplinary Laboratory for Tomorrow's Energy Pack (LIED), CNRS UMR 8236, University Paris Diderot-Paris 7, Paris, France
| | - Olivier Ploux
- Interdisciplinary Laboratory for Tomorrow's Energy Pack (LIED), CNRS UMR 8236, University Paris Diderot-Paris 7, Paris, France
| | - Jean-Philippe Leleu
- Tropical Neuroepidemiology, INSERM UMR 1094, Limoges, France
- University of Limoges, School of Medicine, Institute of Neuroepidemiology and Tropical Neurology, Centre national de la recherche scientifique FR 3503 GEIST, Limoges, France
| | - Luc Brient
- UMR 6553 ECOBIO, Ecosystems—Biodiversity—Evolution, University Rennes I, Rennes, France
| | - Marion Lengronne
- UMR 6553 ECOBIO, Ecosystems—Biodiversity—Evolution, University Rennes I, Rennes, France
| | - Valérie Pichon
- Department of Analytical, Bioanalytical Sciences and Miniaturization (LSABM), UMR ESPCI-ParisTech-CNRS 8231 CBI, Paris, France
- University Sorbonne, University Pierre and Marie Curie (UPMC), Paris, France
| | - Audrey Combès
- Department of Analytical, Bioanalytical Sciences and Miniaturization (LSABM), UMR ESPCI-ParisTech-CNRS 8231 CBI, Paris, France
- University Sorbonne, University Pierre and Marie Curie (UPMC), Paris, France
| | - Saïda El Abdellaoui
- Department of Analytical, Bioanalytical Sciences and Miniaturization (LSABM), UMR ESPCI-ParisTech-CNRS 8231 CBI, Paris, France
- University Sorbonne, University Pierre and Marie Curie (UPMC), Paris, France
| | - Vincent Bonneterre
- Environment and Health Prediction in Populations (EPSP), CNRS-TIMC-IMAG UMR 5525 UJF-Grenoble 1, Grenoble, France
| | - Emmeline Lagrange
- Department of Neurology, University Hospital of Grenoble, Grenoble, France
| | - Gérard Besson
- Department of Neurology, University Hospital of Grenoble, Grenoble, France
| | - Dominique J Bicout
- Environment and Health Prediction in Populations (EPSP), CNRS-TIMC-IMAG UMR 5525 UJF-Grenoble 1, Grenoble, France
- Biomathematics and Epidemiology, Environment and Health Prediction in Populations (EPSP), VetAgro Sup, Marcy-l'Etoile, France
| | - Jean Boutonnat
- Department of Neurology, University Hospital of Grenoble, Grenoble, France
| | - William Camu
- Motoneuron Diseases: Neuroinflammation and Therapy, INSERM UMR 1051, Neurosciences Institute, Montpellier, France
- Department of Neurology, ALS Center, University Hospital Gui de Chauliac, Montpellier, France
| | - Nicolas Pageot
- Motoneuron Diseases: Neuroinflammation and Therapy, INSERM UMR 1051, Neurosciences Institute, Montpellier, France
- Department of Neurology, ALS Center, University Hospital Gui de Chauliac, Montpellier, France
| | - Raul Juntas-Morales
- Motoneuron Diseases: Neuroinflammation and Therapy, INSERM UMR 1051, Neurosciences Institute, Montpellier, France
- Department of Neurology, ALS Center, University Hospital Gui de Chauliac, Montpellier, France
| | - Valérie Rigau
- Motoneuron Diseases: Neuroinflammation and Therapy, INSERM UMR 1051, Neurosciences Institute, Montpellier, France
- Department of Neurology, ALS Center, University Hospital Gui de Chauliac, Montpellier, France
| | - Estelle Masseret
- UMR 5119 ECOSYM, Ecology of Coastal Marine Systems, UM2-CNRS-IRD-Ifremer-UM1, University Montpellier II, Montpellier, France
| | - Eric Abadie
- Environment Resources Laboratory/Languedoc-Roussillon, Ifremer, Sète, France
| | - Pierre-Marie Preux
- Tropical Neuroepidemiology, INSERM UMR 1094, Limoges, France
- University of Limoges, School of Medicine, Institute of Neuroepidemiology and Tropical Neurology, Centre national de la recherche scientifique FR 3503 GEIST, Limoges, France
- Department of Neurology, ALS Center, University Hospital Dupuytren, Limoges, France
| | - Benoît Marin
- Tropical Neuroepidemiology, INSERM UMR 1094, Limoges, France
- University of Limoges, School of Medicine, Institute of Neuroepidemiology and Tropical Neurology, Centre national de la recherche scientifique FR 3503 GEIST, Limoges, France
| |
Collapse
|
22
|
Intracellular fibril formation, calcification, and enrichment of chaperones, cytoskeletal, and intermediate filament proteins in the adult hippocampus CA1 following neonatal exposure to the nonprotein amino acid BMAA. Arch Toxicol 2014; 89:423-36. [PMID: 24798087 PMCID: PMC4335130 DOI: 10.1007/s00204-014-1262-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/15/2014] [Indexed: 12/31/2022]
Abstract
The environmental neurotoxin β-N-methylamino-l-alanine (BMAA) has been implicated in the etiology of neurodegenerative disease, and recent studies indicate that BMAA can be misincorporated into proteins. BMAA is a developmental neurotoxicant that can induce long-term learning and memory deficits, as well as regionally restricted neuronal degeneration and mineralization in the hippocampal CA1. The aim of the study was to characterize long-term changes (2 weeks to 6 months) further in the brain of adult rats treated neonatally (postnatal days 9–10) with BMAA (460 mg/kg) using immunohistochemistry (IHC), transmission electron microscopy, and laser capture microdissection followed by LC-MS/MS for proteomic analysis. The histological examination demonstrated progressive neurodegenerative changes, astrogliosis, microglial activation, and calcification in the hippocampal CA1 3–6 months after exposure. The IHC showed an increased staining for α-synuclein and ubiquitin in the area. The ultrastructural examination revealed intracellular deposition of abundant bundles of closely packed parallel fibrils in neurons, axons, and astrocytes of the CA1. Proteomic analysis of the affected site demonstrated an enrichment of chaperones (e.g., clusterin, GRP-78), cytoskeletal and intermediate filament proteins, and proteins involved in the antioxidant defense system. Several of the most enriched proteins (plectin, glial fibrillar acidic protein, vimentin, Hsp 27, and ubiquitin) are known to form complex astrocytic inclusions, so-called Rosenthal fibers, in the neurodegenerative disorder Alexander disease. In addition, TDP-43 and the negative regulator of autophagy, GLIPR-2, were exclusively detected. The present study demonstrates that neonatal exposure to BMAA may offer a novel model for the study of hippocampal fibril formation in vivo.
Collapse
|
23
|
Daniels O, Fabbro L, Makiela S. The effects of the toxic cyanobacterium Limnothrix (strain AC0243) on Bufo marinus larvae. Toxins (Basel) 2014; 6:1021-35. [PMID: 24662524 PMCID: PMC3968374 DOI: 10.3390/toxins6031021] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 02/17/2014] [Accepted: 02/20/2014] [Indexed: 02/07/2023] Open
Abstract
Limnothrix (strain AC0243) is a cyanobacterium, which has only recently been identified as toxin producing. Under laboratory conditions, Bufo marinus larvae were exposed to 100,000 cells mL(-1) of Limnothrix (strain AC0243) live cultures for seven days. Histological examinations were conducted post mortem and revealed damage to the notochord, eyes, brain, liver, kidney, pancreas, gastrointestinal tract, and heart. The histopathological results highlight the toxicological impact of this strain, particularly during developmental stages. Toxicological similarities to β-N-Methylamino-L-alanine are discussed.
Collapse
Affiliation(s)
- Olivia Daniels
- School of Medical and Applied Sciences, Central Queensland University, Rockhampton 4701, Australia.
| | - Larelle Fabbro
- School of Medical and Applied Sciences, Central Queensland University, Rockhampton 4701, Australia.
| | - Sandrine Makiela
- School of Medical and Applied Sciences, Central Queensland University, Rockhampton 4701, Australia.
| |
Collapse
|
24
|
Karlsson O, Jiang L, Andersson M, Ilag LL, Brittebo EB. Protein association of the neurotoxin and non-protein amino acid BMAA (β-N-methylamino-L-alanine) in the liver and brain following neonatal administration in rats. Toxicol Lett 2014; 226:1-5. [PMID: 24472610 DOI: 10.1016/j.toxlet.2014.01.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 01/16/2014] [Accepted: 01/17/2014] [Indexed: 10/25/2022]
Abstract
The environmental neurotoxin β-N-methylamino-L-alanine (BMAA) is not an amino acid that is normally found in proteins. Our previous autoradiographic study of (3)H-labeled BMAA in adult mice unexpectedly revealed a tissue distribution similar to that of protein amino acids. The aim of this study was to characterize the distribution of free and protein-bound BMAA in neonatal rat tissues following a short exposure using autoradiographic imaging and ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). The autoradiographic imaging of (14)C-L-BMAA demonstrated a distinct uptake of radioactivity that was retained following acid extraction in tissues with a high rate of cell turnover and/or protein synthesis. The UHPLC-MS/MS analysis conclusively demonstrated a dose-dependent increase of protein-associated BMAA in neonatal rat tissues. The level of protein-associated BMAA in the liver was more than 10 times higher than that in brain regions not fully protected by the blood-brain barrier which may be due to the higher rate of protein synthesis in the liver. In conclusion, this study demonstrated that BMAA was associated with rat proteins suggesting that BMAA may be misincorporated into proteins. However, protein-associated BMAA seemed to be cleared over time, as none of the samples from adult rats had any detectable free or protein-associated BMAA.
Collapse
Affiliation(s)
- Oskar Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, SE-751 24 Uppsala, Sweden; Department of Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden.
| | - Liying Jiang
- Department of Analytical Chemistry, Stockholm University, SE-10691 Stockholm, Sweden
| | - Marie Andersson
- Department of Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Leopold L Ilag
- Department of Analytical Chemistry, Stockholm University, SE-10691 Stockholm, Sweden
| | - Eva B Brittebo
- Department of Pharmaceutical Biosciences, Uppsala University, SE-751 24 Uppsala, Sweden
| |
Collapse
|
25
|
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: 2.7] [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
|