1
|
Raymond J, Howard IM, Berry J, Larson T, Horton DK, Mehta P. Head Injury and Amyotrophic Lateral Sclerosis: Population-Based Study from the National ALS Registry. Brain Sci 2025; 15:143. [PMID: 40002476 PMCID: PMC11852576 DOI: 10.3390/brainsci15020143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/24/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: To examine if head injury (HI) is associated with age at ALS diagnosis in the United States. Methods: In this cross-sectional populationf-based analysis, we identified patients with ALS who were registered from 2015 to 2023 who completed the Registry's head trauma survey module. The association between HI and age at ALS diagnosis was assessed using multivariate analysis. Results: Of the 3424 respondents, 56.6% had experienced a HI. The adjusted odds ratio (aOR) for an ALS diagnosis before age 60 years for patients with a HI was 1.24 (95% CI, 1.07-1.45). One or two HIs had an aOR of 1.15 (95% CI, 0.97-1.36), and five or more HIs had an aOR of 1.58 (95% CI, 1.19-2.09). HI before age 18 years yielded an aOR of 2.03 (95% CI, 1.53-2.70) as well as HI between the ages of 18 and 30 years (aOR = 1.48, 95% CI: 1.06-2.06)). When narrowing the analysis to patients with HI before age 18 compared with patients with no HI, we found an association with HI that led to an emergency department or hospital visit (aOR = 1.50 (95% CI: 1.21-1.86)). Conclusions: In this cross-sectional analysis of ALS patients, HIs occurring in childhood and early adulthood and the number of HIs increased the odds of being diagnosed before age 60 years. These results suggest that HI continues to be a risk factor for ALS and could be associated with a younger age of diagnosis.
Collapse
Affiliation(s)
- Jaime Raymond
- Agency for Toxic Substances and Disease Registry/Centers for Disease Control and Prevention, Atlanta, GA 30341, USA; (J.B.); (T.L.); (D.K.H.); (P.M.)
| | - Ileana M. Howard
- Rehabilitation Care Services, VA Puget Sound Healthcare System, Seattle, WA 98108, USA;
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98195, USA
| | - Jasmine Berry
- Agency for Toxic Substances and Disease Registry/Centers for Disease Control and Prevention, Atlanta, GA 30341, USA; (J.B.); (T.L.); (D.K.H.); (P.M.)
| | - Theodore Larson
- Agency for Toxic Substances and Disease Registry/Centers for Disease Control and Prevention, Atlanta, GA 30341, USA; (J.B.); (T.L.); (D.K.H.); (P.M.)
| | - D. Kevin Horton
- Agency for Toxic Substances and Disease Registry/Centers for Disease Control and Prevention, Atlanta, GA 30341, USA; (J.B.); (T.L.); (D.K.H.); (P.M.)
| | - Paul Mehta
- Agency for Toxic Substances and Disease Registry/Centers for Disease Control and Prevention, Atlanta, GA 30341, USA; (J.B.); (T.L.); (D.K.H.); (P.M.)
| |
Collapse
|
2
|
Pereira-Santos AR, Candeias E, Magalhães JD, Empadinhas N, Esteves AR, Cardoso SM. Neuronal control of microglia through the mitochondria. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167167. [PMID: 38626829 DOI: 10.1016/j.bbadis.2024.167167] [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: 02/23/2024] [Accepted: 04/08/2024] [Indexed: 04/21/2024]
Abstract
The microbial toxin β-N-methylamino-L-alanine (BMAA), which is derived from cyanobacteria, targets neuronal mitochondria, leading to the activation of neuronal innate immunity and, consequently, neurodegeneration. Although known to modulate brain inflammation, the precise role of aberrant microglial function in the neurodegenerative process remains elusive. To determine if neurons signal microglial cells, we treated primary cortical neurons with BMAA and then co-cultured them with the N9 microglial cell line. Our observations indicate that microglial cell activation requires initial neuronal priming. Contrary to what was observed in cortical neurons, BMAA was not able to activate inflammatory pathways in N9 cells. We observed that microglial activation is dependent on mitochondrial dysfunction signaled by BMAA-treated neurons. In this scenario, the NLRP3 pro-inflammatory pathway is activated due to mitochondrial impairment in N9 cells. These results demonstrate that microglia activation in the presence of BMAA is dependent on neuronal signaling. This study provides evidence that neurons may trigger microglia activation and subsequent neuroinflammation. In addition, we demonstrate that microglial activation may have a protective role in ameliorating neuronal innate immune activation, at least in the initial phase. This work challenges the current understanding of neuroinflammation by assigning the primary role to neurons.
Collapse
Affiliation(s)
- A R Pereira-Santos
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Ph.D. Programme in Biomedicine and Experimental Biology (PDBEB), Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Emanuel Candeias
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - J D Magalhães
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Ph.D. Programme in Biomedicine and Experimental Biology (PDBEB), Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Nuno Empadinhas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - A Raquel Esteves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Sandra M Cardoso
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
3
|
Oliveira NAS, Pinho BR, Pinto J, Guedes de Pinho P, Oliveira JMA. Edaravone counteracts redox and metabolic disruptions in an emerging zebrafish model of sporadic ALS. Free Radic Biol Med 2024; 217:126-140. [PMID: 38531462 DOI: 10.1016/j.freeradbiomed.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/05/2024] [Accepted: 03/20/2024] [Indexed: 03/28/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease in which the death of motor neurons leads to loss of muscle function. Additionally, cognitive and circadian disruptions are common in ALS patients, contributing to disease progression and burden. Most ALS cases are sporadic, and environmental exposures contribute to their aetiology. However, animal models of these sporadic ALS cases are scarce. The small vertebrate zebrafish is a leading organism to model neurodegenerative diseases; previous studies have proposed bisphenol A (BPA) or β-methylamino-l-alanine (BMAA) exposure to model sporadic ALS in zebrafish, damaging motor neurons and altering motor responses. Here we characterise the face and predictive validity of sporadic ALS models, showing their potential for the mechanistic study of ALS drugs. We phenotypically characterise the BPA and BMAA-induced models, going beyond motor activity and motor axon morphology, to include circadian, redox, proteostasis, and metabolomic phenotypes, and assessing their predictive validity for ALS modelling. BPA or BMAA exposure induced concentration-dependent activity impairments. Also, exposure to BPA but not BMAA induced motor axonopathy and circadian alterations in zebrafish larvae. Our further study of the BPA model revealed loss of habituation to repetitive startles, increased oxidative damage, endoplasmic reticulum (ER) stress, and metabolome abnormalities. The BPA-induced model shows predictive validity, since the approved ALS drug edaravone counteracted BPA-induced motor phenotypes, ER stress, and metabolic disruptions. Overall, BPA exposure is a promising model of ALS-related redox and ER imbalances, contributing to fulfil an unmet need for validated sporadic ALS models.
Collapse
Affiliation(s)
- Nuno A S Oliveira
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University of Porto, 4050-313, Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Mitochondria and Neurobiology Lab, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Brígida R Pinho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University of Porto, 4050-313, Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Mitochondria and Neurobiology Lab, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Joana Pinto
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University of Porto, 4050-313, Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Faculty of Pharmacy, Laboratory of Toxicology, University of Porto, 4050-313, Porto, Portugal
| | - Paula Guedes de Pinho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University of Porto, 4050-313, Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Faculty of Pharmacy, Laboratory of Toxicology, University of Porto, 4050-313, Porto, Portugal
| | - Jorge M A Oliveira
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University of Porto, 4050-313, Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Mitochondria and Neurobiology Lab, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
4
|
De Cock L, Bercier V, Van Den Bosch L. New developments in pre-clinical models of ALS to guide translation. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:477-524. [PMID: 38802181 DOI: 10.1016/bs.irn.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder in which selective death of motor neurons leads to muscle weakness and paralysis. Most research has focused on understanding and treating monogenic familial forms, most frequently caused by mutations in SOD1, FUS, TARDBP and C9orf72, although ALS is mostly sporadic and without a clear genetic cause. Rodent models have been developed to study monogenic ALS, but despite numerous pre-clinical studies and clinical trials, few disease-modifying therapies are available. ALS is a heterogeneous disease with complex underlying mechanisms where several genes and molecular pathways appear to play a role. One reason for the high failure rate of clinical translation from the current models could be oversimplification in pre-clinical studies. Here, we review advances in pre-clinical models to better capture the heterogeneous nature of ALS and discuss the value of novel model systems to guide translation and aid in the development of precision medicine.
Collapse
Affiliation(s)
- Lenja De Cock
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Louvain-University of Leuven, Leuven, Belgium; Center for Brain and Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
| | - Valérie Bercier
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Louvain-University of Leuven, Leuven, Belgium; Center for Brain and Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Louvain-University of Leuven, Leuven, Belgium; Center for Brain and Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium.
| |
Collapse
|
5
|
Moreno-Jiménez L, Benito-Martín MS, Sanclemente-Alamán I, Matías-Guiu JA, Sancho-Bielsa F, Canales-Aguirre A, Mateos-Díaz JC, Matías-Guiu J, Aguilar J, Gómez-Pinedo U. Murine experimental models of amyotrophic lateral sclerosis: an update. Neurologia 2024; 39:282-291. [PMID: 37116688 DOI: 10.1016/j.nrleng.2021.07.004] [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: 06/10/2021] [Accepted: 07/08/2021] [Indexed: 04/30/2023] Open
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease whose aetiology is unknown. It is characterised by upper and lower motor neuron degeneration. Approximately 90% of cases of ALS are sporadic, whereas the other 10% are familial. Regardless of whether the case is familial o sporadic, patients will develop progressive weakness, muscle atrophy with spasticity, and muscle contractures. Life expectancy of these patients is generally 2 to 5 years after diagnosis. DEVELOPMENT In vivo models have helped to clarify the aetiology and pathogenesis of ALS, as well as the mechanisms of the disease. However, as these mechanisms are not yet fully understood, experimental models are essential to the continued study of the pathogenesis of ALS, as well as in the search for possible therapeutic targets. Although 90% of cases are sporadic, most of the models used to study ALS pathogenesis are based on genetic mutations associated with the familial form of the disease; the pathogenesis of sporadic ALS remains unknown. Therefore, it would be critical to establish models based on the sporadic form. CONCLUSIONS This article reviews the main genetic and sporadic experimental models used in the study of this disease, focusing on those that have been developed using rodents.
Collapse
Affiliation(s)
- L Moreno-Jiménez
- Laboratorio de Neurobiología, Instituto de Neurociencias, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - M S Benito-Martín
- Laboratorio de Neurobiología, Instituto de Neurociencias, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - I Sanclemente-Alamán
- Laboratorio de Neurobiología, Instituto de Neurociencias, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - J A Matías-Guiu
- Departamento de Neurología, Instituto de Neurociencias, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - F Sancho-Bielsa
- Departamento de Fisiología, Facultad de Medicina de Ciudad Real, Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | | | - J C Mateos-Díaz
- Departamento de Biotecnología Industrial, CIATEJ-CONACyT, Zapopan, Mexico
| | - J Matías-Guiu
- Laboratorio de Neurobiología, Instituto de Neurociencias, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain; Departamento de Neurología, Instituto de Neurociencias, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - J Aguilar
- Laboratorio de Neurofisiología Experimental y Circuitos Neuronales del Hospital Nacional de Parapléjicos, Toledo, Spain
| | - U Gómez-Pinedo
- Laboratorio de Neurobiología, Instituto de Neurociencias, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Nugumanova G, Ponomarev ED, Askarova S, Fasler-Kan E, Barteneva NS. Freshwater Cyanobacterial Toxins, Cyanopeptides and Neurodegenerative Diseases. Toxins (Basel) 2023; 15:toxins15030233. [PMID: 36977124 PMCID: PMC10057253 DOI: 10.3390/toxins15030233] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/13/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
Cyanobacteria produce a wide range of structurally diverse cyanotoxins and bioactive cyanopeptides in freshwater, marine, and terrestrial ecosystems. The health significance of these metabolites, which include genotoxic- and neurotoxic agents, is confirmed by continued associations between the occurrence of animal and human acute toxic events and, in the long term, by associations between cyanobacteria and neurodegenerative diseases. Major mechanisms related to the neurotoxicity of cyanobacteria compounds include (1) blocking of key proteins and channels; (2) inhibition of essential enzymes in mammalian cells such as protein phosphatases and phosphoprotein phosphatases as well as new molecular targets such as toll-like receptors 4 and 8. One of the widely discussed implicated mechanisms includes a misincorporation of cyanobacterial non-proteogenic amino acids. Recent research provides evidence that non-proteinogenic amino acid BMAA produced by cyanobacteria have multiple effects on translation process and bypasses the proof-reading ability of the aminoacyl-tRNA-synthetase. Aberrant proteins generated by non-canonical translation may be a factor in neuronal death and neurodegeneration. We hypothesize that the production of cyanopeptides and non-canonical amino acids is a more general mechanism, leading to mistranslation, affecting protein homeostasis, and targeting mitochondria in eukaryotic cells. It can be evolutionarily ancient and initially developed to control phytoplankton communities during algal blooms. Outcompeting gut symbiotic microorganisms may lead to dysbiosis, increased gut permeability, a shift in blood-brain-barrier functionality, and eventually, mitochondrial dysfunction in high-energy demanding neurons. A better understanding of the interaction between cyanopeptides metabolism and the nervous system will be crucial to target or to prevent neurodegenerative diseases.
Collapse
Affiliation(s)
- Galina Nugumanova
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan
| | - Eugene D Ponomarev
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan
| | - Sholpan Askarova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern, 3010 Bern, Switzerland
| | - Natasha S Barteneva
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan
- The Environment & Resource Efficiency Cluster (EREC), Nazarbayev University, Astana 010000, Kazakhstan
| |
Collapse
|
9
|
Oliveira NAS, Pinho BR, Oliveira JMA. Swimming against ALS: How to model disease in zebrafish for pathophysiological and behavioral studies. Neurosci Biobehav Rev 2023; 148:105138. [PMID: 36933816 DOI: 10.1016/j.neubiorev.2023.105138] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/02/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease that leads to progressive disability and motor impairment. Existing therapies provide modest improvements in patient survival, raising a need for new treatments for ALS. Zebrafish is a promising model animal for translational and fundamental research in ALS - it is an experimentally tractable vertebrate, with high homology to humans and an ample experimental toolbox. These advantages allow high-throughput study of behavioral and pathophysiological phenotypes. The last decade saw an increased interest in modelling ALS in zebrafish, leading to the current abundance and variety of available methods and models. Additionally, the rise of gene editing techniques and toxin combination studies has created novel opportunities for ALS studies in zebrafish. In this review, we address the relevance of zebrafish as a model animal for ALS studies, the strategies for model induction and key phenotypical evaluation. Furthermore, we discuss established and emerging zebrafish models of ALS, analyzing their validity, including their potential for drug testing, and highlighting research opportunities in this area.
Collapse
Affiliation(s)
- Nuno A S Oliveira
- UCIBIO-REQUIMTE, Applied Molecular Biosciences Unit, Mitochondria and Neurobiology Lab, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, Department of Drug Sciences, Pharmacology Lab, University of Porto, 4050-313 Porto, Portugal
| | - Brígida R Pinho
- UCIBIO-REQUIMTE, Applied Molecular Biosciences Unit, Mitochondria and Neurobiology Lab, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, Department of Drug Sciences, Pharmacology Lab, University of Porto, 4050-313 Porto, Portugal
| | - Jorge M A Oliveira
- UCIBIO-REQUIMTE, Applied Molecular Biosciences Unit, Mitochondria and Neurobiology Lab, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, Department of Drug Sciences, Pharmacology Lab, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
10
|
Kennedy C, van Onselen R, Downing TG. β-N-methylamino-l-alanine is a non-competitive inhibitor of vesicular monoamine transporter 2. Toxicon 2023; 222:106978. [PMID: 36410456 DOI: 10.1016/j.toxicon.2022.106978] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/04/2022] [Accepted: 11/18/2022] [Indexed: 11/20/2022]
Abstract
The neurotoxic, non-proteinogenic amino acid β-N-methylamino-l-alanine (BMAA) has been implicated in the development of neurodegenerative diseases; however, the mechanism(s) and mode(s) of toxicity remain unclear. Similarities in the neuropathology and behavioural deficits of neonatal rats exposed to either BMAA or reserpine, a known vesicular monoamine transporter 2 (VMAT2) inhibitor, suggest a similar mode of action. The aims of this study were therefore to determine if BMAA could prevent the uptake of serotonin into dense granules via inhibition of VMAT2, and, if so, the type of inhibition caused by BMAA. Exposing platelet dense granules to BMAA resulted in a concentration-dependent reduction in serotonin uptake. The inhibition of VMAT2 was non-competitive. The findings from this study support previous reports that BMAA-associated neuropathologies in a neonatal rat model may be due to VMAT2 inhibition during critical periods of neurogenesis.
Collapse
Affiliation(s)
- Chanté Kennedy
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77000, Port Elizabeth, 6021, South Africa.
| | - Rianita van Onselen
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77000, Port Elizabeth, 6021, South Africa; Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa.
| | - Tim G Downing
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77000, Port Elizabeth, 6021, South Africa.
| |
Collapse
|
11
|
Lopicic S, Svirčev Z, Palanački Malešević T, Kopitović A, Ivanovska A, Meriluoto J. Environmental Neurotoxin β- N-Methylamino-L-alanine (BMAA) as a Widely Occurring Putative Pathogenic Factor in Neurodegenerative Diseases. Microorganisms 2022; 10:2418. [PMID: 36557671 PMCID: PMC9781992 DOI: 10.3390/microorganisms10122418] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
In the present review we have discussed the occurrence of β-N-methylamino-L-alanine (BMAA) and its natural isomers, and the organisms and sample types in which the toxin(s) have been detected. Further, the review discusses general pathogenic mechanisms of neurodegenerative diseases, and how modes of action of BMAA fit in those mechanisms. The biogeography of BMAA occurrence presented here contributes to the planning of epidemiological research based on the geographical distribution of BMAA and human exposure. Analysis of BMAA mechanisms in relation to pathogenic processes of neurodegeneration is used to critically assess the potential significance of the amino acid as well as to identify gaps in our understanding. Taken together, these two approaches provide the basis for the discussion on the potential role of BMAA as a secondary factor in neurodegenerative diseases, the rationale for further research and possible directions the research can take, which are outlined in the conclusions.
Collapse
Affiliation(s)
- Srdjan Lopicic
- Faculty of Medicine, University of Belgrade, Dr Subotića Starijeg 8, 11000 Belgrade, Serbia
| | - Zorica Svirčev
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia
- Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, Tykistökatu 6A, 20520 Turku, Finland
| | - Tamara Palanački Malešević
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia
| | - Aleksandar Kopitović
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
| | - Aleksandra Ivanovska
- Innovation Center of the Faculty of Technology and Metallurgy, University of Belgrade, Karnegijeva 4, 11000 Belgrade, Serbia
| | - Jussi Meriluoto
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia
- Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, Tykistökatu 6A, 20520 Turku, Finland
| |
Collapse
|
12
|
Koksharova OA, Safronova NA. Non-Proteinogenic Amino Acid β-N-Methylamino-L-Alanine (BMAA): Bioactivity and Ecological Significance. Toxins (Basel) 2022; 14:539. [PMID: 36006201 PMCID: PMC9414260 DOI: 10.3390/toxins14080539] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/28/2022] [Accepted: 08/04/2022] [Indexed: 11/21/2022] Open
Abstract
Research interest in a non-protein amino acid β-N-methylamino-L-alanine (BMAA) arose due to the discovery of a connection between exposure to BMAA and the occurrence of neurodegenerative diseases. Previous reviews on this topic either considered BMAA as a risk factor for neurodegenerative diseases or focused on the problems of detecting BMAA in various environmental samples. Our review is devoted to a wide range of fundamental biological problems related to BMAA, including the molecular mechanisms of biological activity of BMAA and the complex relationships between producers of BMAA and the environment in various natural ecosystems. At the beginning, we briefly recall the most important facts about the producers of BMAA (cyanobacteria, microalgae, and bacteria), the pathways of BMAA biosynthesis, and reliable methods of identification of BMAA. The main distinctive feature of our review is a detailed examination of the molecular mechanisms underlying the toxicity of BMAA to living cells. A brand new aspect, not previously discussed in any reviews, is the effect of BMAA on cyanobacterial cells. These recent studies, conducted using transcriptomics and proteomics, revealed potent regulatory effects of BMAA on the basic metabolism and cell development of these ancient photoautotrophic prokaryotes. Exogenous BMAA strongly influences cell differentiation and primary metabolic processes in cyanobacteria, such as nitrogen fixation, photosynthesis, carbon fixation, and various biosynthetic processes involving 2-oxoglutarate and glutamate. Cyanobacteria were found to be more sensitive to exogenous BMAA under nitrogen-limited growth conditions. We suggest a hypothesis that this toxic diaminoacid can be used by phytoplankton organisms as a possible allelopathic tool for controlling the population of cyanobacterial cells during a period of intense competition for nitrogen and other resources in various ecosystems.
Collapse
Affiliation(s)
- Olga A. Koksharova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, Kurchatov Square, 2, 123182 Moscow, Russia
| | - Nina A. Safronova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| |
Collapse
|
13
|
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]
|
14
|
Kazemi Shariat Panahi H, Dehhaghi M, Heng B, Lane DJR, Bush AI, Guillemin GJ, Tan VX. Neuropathological Mechanisms of β-N-Methylamino-L-Alanine (BMAA) with a Focus on Iron Overload and Ferroptosis. Neurotox Res 2022; 40:614-635. [PMID: 35023054 DOI: 10.1007/s12640-021-00455-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 11/22/2021] [Accepted: 11/30/2021] [Indexed: 02/08/2023]
Abstract
The incidence of neurodegenerative diseases and cyanobacterial blooms is concomitantly increasing worldwide. The cyanotoxin β-N-methylamino-L-alanine (BMAA) is produced by most of the Cyanobacteria spp. This cyanotoxin is described as a potential environmental etiology factor for some sporadic neurodegenerative diseases. Climate change and eutrophication significantly increase the frequency and intensity of cyanobacterial bloom in water bodies. This review evaluates different neuropathological mechanisms of BMAA at molecular and cellular levels and compares the related studies to provide some useful recommendations. Additionally, the structure and properties of BMAA as well as its microbial origin, especially by gut bacteria, are also briefly covered. Unlike previous reviews, we hypothesize the possible neurotoxic mechanism of BMAA through iron overload. We also discuss the involvement of BMAA in excitotoxicity, TAR DNA-binding protein 43 (TDP-43) translocation and accumulation, tauopathy, and other protein misincorporation and misfolding.
Collapse
Affiliation(s)
- Hamed Kazemi Shariat Panahi
- Neuroinflammation Group, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Mona Dehhaghi
- Neuroinflammation Group, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- PANDIS.Org, Bendigo, Australia
- Department of Microbial Biotechnology, School of Biology and Centre of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
| | - Benjamin Heng
- Neuroinflammation Group, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Gilles J Guillemin
- Neuroinflammation Group, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.
- PANDIS.Org, Bendigo, Australia.
| | - Vanessa X Tan
- Neuroinflammation Group, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- PANDIS.Org, Bendigo, Australia
| |
Collapse
|
15
|
Todd TW, Petrucelli L. Modelling amyotrophic lateral sclerosis in rodents. Nat Rev Neurosci 2022; 23:231-251. [PMID: 35260846 DOI: 10.1038/s41583-022-00564-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2022] [Indexed: 12/11/2022]
Abstract
The efficient study of human disease requires the proper tools, one of the most crucial of which is an accurate animal model that faithfully recapitulates the human condition. The study of amyotrophic lateral sclerosis (ALS) is no exception. Although the majority of ALS cases are considered sporadic, most animal models of this disease rely on genetic mutations identified in familial cases. Over the past decade, the number of genes associated with ALS has risen dramatically and, with each new genetic variant, there is a drive to develop associated animal models. Rodent models are of particular importance as they allow for the study of ALS in the context of a living mammal with a comparable CNS. Such models not only help to verify the pathogenicity of novel mutations but also provide critical insight into disease mechanisms and are crucial for the testing of new therapeutics. In this Review, we aim to summarize the full spectrum of ALS rodent models developed to date.
Collapse
Affiliation(s)
- Tiffany W Todd
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA.
| |
Collapse
|
16
|
Bonifacino T, Zerbo RA, Balbi M, Torazza C, Frumento G, Fedele E, Bonanno G, Milanese M. Nearly 30 Years of Animal Models to Study Amyotrophic Lateral Sclerosis: A Historical Overview and Future Perspectives. Int J Mol Sci 2021; 22:ijms222212236. [PMID: 34830115 PMCID: PMC8619465 DOI: 10.3390/ijms222212236] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 12/20/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal, multigenic, multifactorial, and non-cell autonomous neurodegenerative disease characterized by upper and lower motor neuron loss. Several genetic mutations lead to ALS development and many emerging gene mutations have been discovered in recent years. Over the decades since 1990, several animal models have been generated to study ALS pathology including both vertebrates and invertebrates such as yeast, worms, flies, zebrafish, mice, rats, guinea pigs, dogs, and non-human primates. Although these models show different peculiarities, they are all useful and complementary to dissect the pathological mechanisms at the basis of motor neuron degeneration and ALS progression, thus contributing to the development of new promising therapeutics. In this review, we describe the up to date and available ALS genetic animal models, classified by the different genetic mutations and divided per species, pointing out their features in modeling, the onset and progression of the pathology, as well as their specific pathological hallmarks. Moreover, we highlight similarities, differences, advantages, and limitations, aimed at helping the researcher to select the most appropriate experimental animal model, when designing a preclinical ALS study.
Collapse
Affiliation(s)
- Tiziana Bonifacino
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Genoa, Italy
| | - Roberta Arianna Zerbo
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
| | - Matilde Balbi
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
| | - Carola Torazza
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
| | - Giulia Frumento
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
| | - Ernesto Fedele
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Correspondence:
| | - Giambattista Bonanno
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Marco Milanese
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Genoa, Italy
| |
Collapse
|
17
|
Moreno-Jiménez L, Benito-Martín M, Sanclemente-Alamán I, Matías-Guiu J, Sancho-Bielsa F, Canales-Aguirre A, Mateos-Díaz J, Matías-Guiu J, Aguilar J, Gómez-Pinedo U. Modelos experimentales murinos en la esclerosis lateral amiotrófica. Puesta al día. Neurologia 2021. [DOI: 10.1016/j.nrl.2021.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
18
|
Martínez-González L, Gonzalo-Consuegra C, Gómez-Almería M, Porras G, de Lago E, Martín-Requero Á, Martínez A. Tideglusib, a Non-ATP Competitive Inhibitor of GSK-3β as a Drug Candidate for the Treatment of Amyotrophic Lateral Sclerosis. Int J Mol Sci 2021; 22:ijms22168975. [PMID: 34445680 PMCID: PMC8396476 DOI: 10.3390/ijms22168975] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 12/25/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is the most common degenerative motor neuron disease in adults. About 97% of ALS patients present TDP-43 aggregates with post-translational modifications, such as hyperphosphorylation, in the cytoplasm of affected cells. GSK-3β is one of the protein kinases involved in TDP-43 phosphorylation. Up-regulation of its expression and activity is reported on spinal cord and cortex tissues of ALS patients. Here, we propose the repurposing of Tideglusib, an in-house non-ATP competitive GSK-3β inhibitor that is currently in clinical trials for autism and myotonic dystrophy, as a promising therapeutic strategy for ALS. With this aim we have evaluated the efficacy of Tideglusib in different experimental ALS models both in vitro and in vivo. Moreover, we observed that GSK-3β activity is increased in lymphoblasts from sporadic ALS patients, with a simultaneous increase in TDP-43 phosphorylation and cytosolic TDP-43 accumulation. Treatment with Tideglusib decreased not only phospho-TDP-43 levels but also recovered its nuclear localization in ALS lymphoblasts and in a human TDP-43 neuroblastoma model. Additionally, we found that chronic oral treatment with Tideglusib is able to reduce the increased TDP-43 phosphorylation in the spinal cord of Prp-hTDP-43A315T mouse model. Therefore, we consider Tideglusib as a promising drug candidate for ALS, being proposed to start a clinical trial phase II by the end of the year.
Collapse
Affiliation(s)
- Loreto Martínez-González
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Calle Ramiro Maétzu 9, 28040 Madrid, Spain; (L.M.-G.); (G.P.)
| | - Claudia Gonzalo-Consuegra
- Instituto de Investigación en Neuroquίmica, Departamento de Bioquίmica y Biologίa Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (C.G.-C.); (M.G.-A.); (E.d.L.)
| | - Marta Gómez-Almería
- Instituto de Investigación en Neuroquίmica, Departamento de Bioquίmica y Biologίa Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (C.G.-C.); (M.G.-A.); (E.d.L.)
| | - Gracia Porras
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Calle Ramiro Maétzu 9, 28040 Madrid, Spain; (L.M.-G.); (G.P.)
| | - Eva de Lago
- Instituto de Investigación en Neuroquίmica, Departamento de Bioquίmica y Biologίa Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (C.G.-C.); (M.G.-A.); (E.d.L.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Ángeles Martín-Requero
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Calle Ramiro Maétzu 9, 28040 Madrid, Spain; (L.M.-G.); (G.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
- Correspondence: (Á.M.-R.); (A.M.); Tel.: +34-918-37-12 (ext. 4222) (Á.M.-R.); +34-918-37-31-12 (ext. 4437) (A.M.)
| | - Ana Martínez
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Calle Ramiro Maétzu 9, 28040 Madrid, Spain; (L.M.-G.); (G.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
- Correspondence: (Á.M.-R.); (A.M.); Tel.: +34-918-37-12 (ext. 4222) (Á.M.-R.); +34-918-37-31-12 (ext. 4437) (A.M.)
| |
Collapse
|
19
|
Cyanobacteria, Cyanotoxins, and Neurodegenerative Diseases: Dangerous Liaisons. Int J Mol Sci 2021; 22:ijms22168726. [PMID: 34445429 PMCID: PMC8395864 DOI: 10.3390/ijms22168726] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 02/06/2023] Open
Abstract
The prevalence of neurodegenerative disease (ND) is increasing, partly owing to extensions in lifespan, with a larger percentage of members living to an older age, but the ND aetiology and pathogenesis are not fully understood, and effective treatments are still lacking. Neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis are generally thought to progress as a consequence of genetic susceptibility and environmental influences. Up to now, several environmental triggers have been associated with NDs, and recent studies suggest that some cyanotoxins, produced by cyanobacteria and acting through a variety of molecular mechanisms, are highly neurotoxic, although their roles in neuropathy and particularly in NDs are still controversial. In this review, we summarize the most relevant and recent evidence that points at cyanotoxins as environmental triggers in NDs development.
Collapse
|
20
|
Interaction of the neutral amino acid transporter ASCT2 with basic amino acids. Biochem J 2020; 477:1443-1457. [PMID: 32242892 DOI: 10.1042/bcj20190859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/16/2020] [Accepted: 04/03/2020] [Indexed: 12/20/2022]
Abstract
Glutamine transport across cell membranes is performed by a variety of transporters, including the alanine serine cysteine transporter 2 (ASCT2). The substrate-binding site of ASCT2 was proposed to be specific for small amino acids with neutral side chains, excluding basic substrates such as lysine. A series of competitive inhibitors of ASCT2 with low µM affinity were developed previously, on the basis of the 2,4-diaminobutyric acid (DAB) scaffold with a potential positive charge in the side chain. Therefore, we tested whether basic amino acids with side chains shorter than lysine can interact with the ASCT2 binding site. Molecular docking of L-1,3-diaminopropionic acid (L-DAP) and L-DAB suggested that these compounds bind to ASCT2. Consistent with this prediction, L-DAP and L-DAB, but not ornithine, lysine or D-DAP, elicited currents when applied to ASCT2-expressing cells. The currents were carried by anions and showed the hallmark properties of ASCT2 currents induced by transported substrates. The L-DAP response could be eliminated by a competitive ASCT2 inhibitor, suggesting that binding occurs at the substrate binding site. The KM for L-DAP was weakly voltage dependent. Furthermore, the pH dependence of the L-DAP response showed that the compound can bind in several protonation states. Together, these results suggest that the ASCT2 binding site is able to recognize L-amino acids with short, basic side chains, such as the L-DAP derivative β-N-methylamino-l-Alanine (BMAA), a well-studied neurotoxin. Our results expand the substrate specificity of ASCT2 to include amino acid substrates with positively charged side chains.
Collapse
|
21
|
Soto T, Buzzi ED, Rotstein NP, German OL, Politi LE. Damaging effects of BMAA on retina neurons and Müller glial cells. Exp Eye Res 2020; 202:108342. [PMID: 33144094 DOI: 10.1016/j.exer.2020.108342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 10/23/2022]
Abstract
B-N-methylamino-L-alanine (BMAA), a cyanotoxin produced by most cyanobacteria, has been proposed to cause long term damages leading to neurodegenerative diseases, including Amyotrophic Lateral Sclerosis/Parkinsonism Dementia complex (ALS/PDC) and retinal pathologies. Previous work has shown diverse mechanisms leading to BMAA-induced degeneration; however, the underlying mechanisms of toxicity affecting retina cells are not fully elucidated. We here show that BMAA treatment of rat retina neurons in vitro induced nuclear fragmentation and cell death in both photoreceptors (PHRs) and amacrine neurons, provoking mitochondrial membrane depolarization. Pretreatment with the N-Methyl-D-aspartate (NMDA) receptor antagonist MK-801 prevented BMAA-induced death of amacrine neurons, but not that of PHRs, implying activation of NMDA receptors participated only in amacrine cell death. Noteworthy, BMAA stimulated a selective axonal outgrowth in amacrine neurons, simultaneously promoting growth cone destabilization. BMAA partially decreased the viability of Müller glial cells (MGC), the main glial cell type in the retina, induced marked alterations in their actin cytoskeleton and impaired their capacity to protect retinal neurons. BMAA also induced cell death and promoted axonal outgrowth in differentiated rat pheochromocytoma (PC12) cells, implying these effects were not limited to amacrine neurons. These results suggest that BMAA is toxic for retina neurons and MGC and point to the involvement of NMDA receptors in amacrine cell death, providing new insight into the mechanisms involved in BMAA neurotoxic effects in the retina.
Collapse
Affiliation(s)
- Tamara Soto
- Instituto de Investigaciones Bioquímicas, Depto. de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)-CONICET, 8000, Bahía Blanca, Buenos Aires, Argentina
| | - Edgardo D Buzzi
- Instituto de Investigaciones Bioquímicas, Depto. de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)-CONICET, 8000, Bahía Blanca, Buenos Aires, Argentina; Department of Biology, Biochemistry and Pharmacy, Universidad Nacional Del Sur (UNS)-CONICET, Argentina
| | - Nora P Rotstein
- Instituto de Investigaciones Bioquímicas, Depto. de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)-CONICET, 8000, Bahía Blanca, Buenos Aires, Argentina; Department of Biology, Biochemistry and Pharmacy, Universidad Nacional Del Sur (UNS)-CONICET, Argentina
| | - O Lorena German
- Instituto de Investigaciones Bioquímicas, Depto. de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)-CONICET, 8000, Bahía Blanca, Buenos Aires, Argentina; Department of Biology, Biochemistry and Pharmacy, Universidad Nacional Del Sur (UNS)-CONICET, Argentina
| | - Luis E Politi
- Instituto de Investigaciones Bioquímicas, Depto. de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)-CONICET, 8000, Bahía Blanca, Buenos Aires, Argentina.
| |
Collapse
|
22
|
Nunes-Costa D, Magalhães JD, G-Fernandes M, Cardoso SM, Empadinhas N. Microbial BMAA and the Pathway for Parkinson's Disease Neurodegeneration. Front Aging Neurosci 2020; 12:26. [PMID: 32317956 PMCID: PMC7019015 DOI: 10.3389/fnagi.2020.00026] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/23/2020] [Indexed: 12/12/2022] Open
Abstract
The neurotoxin β-N-methylamino-L-alanine (BMAA) is a natural non-proteinogenic diamino acid produced by several species of both prokaryotic (cyanobacteria) and eukaryotic (diatoms and dinoflagellates) microorganisms. BMAA has been shown to biomagnify through the food chain in some ecosystems, accumulating for example in seafood such as shellfish and fish, common dietary sources of BMAA whose ingestion may have possible neuronal consequences. In addition to its excitotoxic potential, BMAA has been implicated in protein misfolding and aggregation, inhibition of specific enzymes and neuroinflammation, all hallmark features of neurodegenerative diseases. However, the exact molecular mechanisms of neurotoxicity remain to be elucidated in detail. Although BMAA is commonly detected in its free form, complex BMAA-containing molecules have also been identified such as the paenilamicins, produced by an insect gut bacterial pathogen. On the other hand, production of BMAA or BMAA-containing molecules by members of the human gut microbiota, for example by non-photosynthetic cyanobacteria, the Melainabacteria, remains only hypothetical. In any case, should BMAA reach the gut it may interact with cells of the mucosal immune system and neurons of the enteric nervous system (ENS) and possibly target the mitochondria. Here, we review the available evidence and hint on possible mechanisms by which chronic exposure to dietary sources of this microbial neurotoxin may drive protein misfolding and mitochondrial dysfunction with concomitant activation of innate immune responses, chronic low-grade gut inflammation, and ultimately the neurodegenerative features observed across the gut-brain axis in Parkinson's disease (PD).
Collapse
Affiliation(s)
- Daniela Nunes-Costa
- CNC–Center for Neuroscience and Cell
Biology, University of Coimbra, Coimbra,
Portugal
- Ph.D. Programme in Biomedicine and Experimental
Biology (PDBEB), Institute for Interdisciplinary Research, University of
Coimbra, Coimbra,
Portugal
| | - João Duarte Magalhães
- CNC–Center for Neuroscience and Cell
Biology, University of Coimbra, Coimbra,
Portugal
- Ph.D. Programme in Biomedicine and Experimental
Biology (PDBEB), Institute for Interdisciplinary Research, University of
Coimbra, Coimbra,
Portugal
| | - Maria G-Fernandes
- CNC–Center for Neuroscience and Cell
Biology, University of Coimbra, Coimbra,
Portugal
| | - Sandra Morais Cardoso
- CNC–Center for Neuroscience and Cell
Biology, University of Coimbra, Coimbra,
Portugal
- Institute of Cellular and Molecular Biology,
Faculty of Medicine, University of Coimbra,
Coimbra, Portugal
| | - Nuno Empadinhas
- CNC–Center for Neuroscience and Cell
Biology, University of Coimbra, Coimbra,
Portugal
- Institute for Interdisciplinary Research
(IIIUC), University of Coimbra, Coimbra,
Portugal
| |
Collapse
|
23
|
Angiopoietin-1 and ανβ3 integrin peptide promote the therapeutic effects of L-serine in an amyotrophic lateral sclerosis/Parkinsonism dementia complex model. Aging (Albany NY) 2019; 10:3507-3527. [PMID: 30476904 PMCID: PMC6286852 DOI: 10.18632/aging.101661] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/15/2018] [Indexed: 01/24/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult disorder of neurodegeneration that manifests as the destruction of upper and lower motor neurons. Beta-N-methylamino-L-alanine (L-BMAA), an amino acid not present in proteins, was found to cause intraneuronal protein misfolding and to induce ALS/Parkinsonism dementia complex (PDC), which presents symptoms analogous to those of Alzheimer’s-like dementia and Parkinsonism. L-serine suppresses the erroneous incorporation of L-BMAA into proteins in the human nervous system. In this study, angiopoietin-1, an endothelial growth factor crucial for vascular development and angiogenesis, and the integrin αvβ3 binding peptide C16, which inhibits inflammatory cell infiltration, were utilized to improve the local microenvironment within the central nervous system of an ALS/PDC rodent model by minimizing inflammation. Our results revealed that L-serine application yielded better effects than C16+ angiopoietin-1 treatment alone for alleviating apoptotic and autophagic changes and improving cognition and electrophysiological dysfunction, but not for improving the inflammatory micro-environment in the central nerve system, while further advances in attenuating the functional disability and pathological impairment induced by L-BMAA could be achieved by co-treatment with C16 and angiopoietin-1 in addition to L-serine. Therefore, C16+ angiopoietin-1 could be beneficial as a supplement to promote the effects of L-serine treatment.
Collapse
|
24
|
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: 18] [Impact Index Per Article: 3.0] [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
|
25
|
Masala A, Sanna S, Esposito S, Rassu M, Galioto M, Zinellu A, Carru C, Carrì MT, Iaccarino C, Crosio C. Epigenetic Changes Associated with the Expression of Amyotrophic Lateral Sclerosis (ALS) Causing Genes. Neuroscience 2018; 390:1-11. [DOI: 10.1016/j.neuroscience.2018.08.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 07/11/2018] [Accepted: 08/07/2018] [Indexed: 12/13/2022]
|
26
|
Pierozan P, Andersson M, Brandt I, Karlsson O. The environmental neurotoxin β-N-methylamino-L-alanine inhibits melatonin synthesis in primary pinealocytes and a rat model. J Pineal Res 2018. [PMID: 29528516 DOI: 10.1111/jpi.12488] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The environmental neurotoxin β-N-methylamino-L-alanine (BMAA) is a glutamate receptor agonist that can induce oxidative stress and has been implicated as a possible risk factor for neurodegenerative disease. Detection of BMAA in mussels, crustaceans, and fish illustrates that the sources of human exposure to this toxin are more abundant than previously anticipated. The aim of this study was to determine uptake of BMAA in the pineal gland and subsequent effects on melatonin production in primary pinealocyte cultures and a rat model. Autoradiographic imaging of 10-day-old male rats revealed a high and selective uptake in the pineal gland at 30 minutes to 24 hours after 14 C-L-BMAA administration (0.68 mg/kg). Primary pinealocyte cultures exposed to 0.05-3 mmol/L BMAA showed a 57%-93% decrease in melatonin synthesis in vitro. Both the metabotropic glutamate receptor 3 (mGluR3) antagonist Ly341495 and the protein kinase C (PKC) activator phorbol-12-myristate-13-acetate prevented the decrease in melatonin secretion, suggesting that BMAA inhibits melatonin synthesis by mGluR3 activation and PKC inhibition. Serum analysis revealed a 45% decrease in melatonin concentration in neonatal rats assessed 2 weeks after BMAA administration (460 mg/kg) and confirmed an inhibition of melatonin synthesis in vivo. Given that melatonin is a most important neuroprotective molecule in the brain, the etiology of BMAA-induced neurodegeneration may include mechanisms beyond direct excitotoxicity and oxidative stress.
Collapse
Affiliation(s)
- Paula Pierozan
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Marie Andersson
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden
| | - Ingvar Brandt
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden
| | - Oskar Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
27
|
Rodríguez-Cueto C, Santos-García I, García-Toscano L, Espejo-Porras F, Bellido ML, Fernández-Ruiz J, Muñoz E, de Lago E. Neuroprotective effects of the cannabigerol quinone derivative VCE-003.2 in SOD1 G93A transgenic mice, an experimental model of amyotrophic lateral sclerosis. Biochem Pharmacol 2018; 157:217-226. [PMID: 30076846 DOI: 10.1016/j.bcp.2018.07.049] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022]
Abstract
Antioxidant phytocannabinoids, synthetic compounds targeting the CB2 receptor, and inhibitors of the endocannabinoid inactivation afforded neuroprotection in SOD1G93A mutant mice, a model of ALS. These effects may involve the activation of PPAR-γ too. Here, we have investigated the neuroprotective effects in SOD1G93A mutant mice of the cannabigerol derivative VCE-003.2, which works as neuroprotectant by activating PPAR-γ. Mice were treated with VCE-003.2 from 60 days up to an advanced stage in disease progression (18 weeks), when they were euthanized and used for analysis of neuropathological signs. As expected, SOD1G93A transgenic mice experienced a progressive weight loss and neurological deterioration, which was associated with a marked loss of spinal cholinergic motor neurons, glial reactivity, and elevations in several biochemical markers (cytokines, glutamate transporters) that indirectly reflect the glial proliferation and activation in the spinal cord. The treatment with VCE-003.2 improved most of these neuropathological signs. It attenuated the weight loss and the anomalies in neurological parameters, preserved spinal cholinergic motor neurons, and reduced astroglial reactivity. VCE-003.2 also reduced the elevations in IL-1β and glial glutamate transporters. Lastly, VCE-003.2 attenuated the LPS-induced generation of TNF-α and IL-1β in cultured astrocytes obtained from SOD1G93A transgenic newborns, an effect also produced by rosiglitazone, then indicating a probable PPAR-γ activation as responsible of its neuroprotective effects. In summary, our results showed benefits with VCE-003.2 in SOD1G93A transgenic mice supporting PPAR-γ as an additional neuroprotective target available for cannabinoids in ALS. Such benefits would need to be validated in other ALS models prior to be translated to the clinical level.
Collapse
Affiliation(s)
- Carmen Rodríguez-Cueto
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Spain
| | - Irene Santos-García
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Spain
| | - Laura García-Toscano
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Spain
| | - Francisco Espejo-Porras
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Spain
| | | | - Javier Fernández-Ruiz
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Spain
| | - Eduardo Muñoz
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain; Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Eva de Lago
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Spain.
| |
Collapse
|
28
|
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: 39] [Impact Index Per Article: 4.9] [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
|
29
|
Nunn PB. 50 years of research on α-amino-β-methylaminopropionic acid (β-methylaminoalanine). PHYTOCHEMISTRY 2017; 144:271-281. [PMID: 29102875 DOI: 10.1016/j.phytochem.2017.10.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/10/2017] [Accepted: 10/13/2017] [Indexed: 06/07/2023]
Abstract
The isolation of α-amino-β-methylaminopropionic acid from seeds of Cycas circinalis (now C. micronesica Hill) resulted from a purposeful attempt to establish the cause of the profound neurological disease, amyotrophic lateral sclerosis/parkinsonism/dementia, that existed in high frequency amongst the inhabitants of the western Pacific island of Guam (Guam ALS/PD). In the 50 years since its discovery the amino acid has been a stimulus, and sometimes a subject of mockery, for generations of scientists in a remarkably diverse range of subject areas. The number of citations of the original paper has risen in the five decades from a few to 120 within the decade 2007-2016 and continues at a high rate into the next decade. The reasons for this remarkable outcome are discussed and examples from the literature are used to illustrate the wide range of scientific interest that the original paper generated.
Collapse
Affiliation(s)
- Peter B Nunn
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, St Michael's Building, White Swan Road, Portsmouth, Hampshire PO1 2DT, UK.
| |
Collapse
|
30
|
Wang MD, Little J, Gomes J, Cashman NR, Krewski D. Identification of risk factors associated with onset and progression of amyotrophic lateral sclerosis using systematic review and meta-analysis. Neurotoxicology 2017; 61:101-130. [DOI: 10.1016/j.neuro.2016.06.015] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 06/29/2016] [Indexed: 12/11/2022]
|
31
|
Baker TC, Tymm FJM, Murch SJ. Assessing Environmental Exposure to β-N-Methylamino-L-Alanine (BMAA) in Complex Sample Matrices: a Comparison of the Three Most Popular LC-MS/MS Methods. Neurotox Res 2017. [PMID: 28643233 DOI: 10.1007/s12640-017-9764-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
β-N-Methylamino-L-alanine (BMAA) is a naturally occurring non-protein amino acid produced by cyanobacteria, accumulated through natural food webs, found in mammalian brain tissues. Recent evidence indicates an association between BMAA and neurological disease. The accurate detection and quantification of BMAA in food and environmental samples are critical to understanding BMAA metabolism and limiting human exposure. To date, there have been more than 78 reports on BMAA in cyanobacteria and human samples, but different methods give conflicting data and divergent interpretations in the literature. The current work was designed to determine whether orthogonal chromatography and mass spectrometry methods give consistent data interpretation from a single sample matrix using the three most common analytical methods. The methods were recreated as precisely as possible from the literature with optimization of the mass spectrometry parameters specific to the instrument. Four sample matrices, cyanobacteria, human brain, blue crab, and Spirulina, were analyzed as 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate (AQC) derivatives, propyl chloroformate (PCF) derivatives separated by reverse phase chromatography, or underivatized extracts separated by HILIC chromatography. The three methods agreed on positive detection of BMAA in cyanobacteria and no detected BMAA in the sample of human brain matrix. Interpretation was less clear for a sample of blue crab which was strongly positive for BMAA by AQC and PCF but negative by HILIC and for four spirulina raw materials that were negative by PCF but positive by AQC and HILIC. Together, these data demonstrate that the methods gave different results and that the choices in interpretation of the methods determined whether BMAA was detected. Failure to detect BMAA cannot be considered proof of absence.
Collapse
Affiliation(s)
- Teesha C Baker
- Chemistry, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
| | - Fiona J M Tymm
- Chemistry, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
| | - Susan J Murch
- Chemistry, University of British Columbia, Kelowna, BC, V1V 1V7, Canada.
| |
Collapse
|
32
|
Metcalf JS, Lobner D, Banack SA, Cox GA, Nunn PB, Wyatt PB, Cox PA. Analysis of BMAA enantiomers in cycads, cyanobacteria, and mammals: in vivo formation and toxicity of D-BMAA. Amino Acids 2017. [PMID: 28620737 DOI: 10.1007/s00726-017-2445-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Chronic dietary exposure to the cyanobacterial toxin β-N-methylamino-L-alanine (BMAA) triggers neuropathology in non-human primates, providing support for the theory that BMAA causes a fatal neurodegenerative illness among the indigenous Chamorro people of Guam. However, since there are two stereoisomers of BMAA, it is important to know if both can occur in nature, and if so, what role they might play in disease causation. As a first step, we analysed both BMAA enantiomers in cyanobacteria, cycads, and in mammals orally dosed with L-BMAA, to determine if enantiomeric changes could occur in vivo. BMAA in cyanobacteria and cycads was found only as the L-enantiomer. However, while the L-enantiomer in mammals was little changed after digestion, we detected a small pool of D-BMAA in the liver (12.5%) of mice and in the blood plasma of vervets (3.6%). Chiral analysis of cerebrospinal fluid of vervets and hindbrain of mice showed that the free BMAA in the central nervous system was the D-enantiomer. In vitro toxicity investigations with D-BMAA showed toxicity, mediated through AMPA rather than NMDA receptors. These findings raise important considerations concerning the neurotoxicity of BMAA and its relationship to neurodegenerative disease.
Collapse
Affiliation(s)
- J S Metcalf
- Brain Chemistry Labs, Institute for Ethnomedicine, Box 3464, Jackson, WY, 83001, USA.
| | - Doug Lobner
- Department of Biomedical Sciences, College of Health Sciences, Marquette University, Milwaukee, WI, 53201, USA
| | - Sandra Anne Banack
- Brain Chemistry Labs, Institute for Ethnomedicine, Box 3464, Jackson, WY, 83001, USA
| | | | - Peter B Nunn
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, PD1 2DT, UK
| | - Peter B Wyatt
- School of Biological and Chemical Sciences, Queen Mary University of London, London, E1 4NS, UK
| | - Paul Alan Cox
- Brain Chemistry Labs, Institute for Ethnomedicine, Box 3464, Jackson, WY, 83001, USA
| |
Collapse
|
33
|
Chernoff N, Hill DJ, Diggs DL, Faison BD, Francis BM, Lang JR, Larue MM, Le TT, Loftin KA, Lugo JN, Schmid JE, Winnik WM. A critical review of the postulated role of the non-essential amino acid, β-N-methylamino-L-alanine, in neurodegenerative disease in humans. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2017; 20:1-47. [PMID: 28598725 PMCID: PMC6503681 DOI: 10.1080/10937404.2017.1297592] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
The compound BMAA (β-N-methylamino-L-alanine) has been postulated to play a significant role in four serious neurological human diseases: Amyotrophic Lateral Sclerosis/Parkinsonism Dementia Complex (ALS/PDC) found on Guam, and ALS, Parkinsonism, and dementia that occur globally. ALS/PDC with symptoms of all three diseases first came to the attention of the scientific community during and after World War II. It was initially associated with cycad flour used for food because BMAA is a product of symbiotic cycad root-dwelling cyanobacteria. Human consumption of flying foxes that fed on cycad seeds was later suggested as a source of BMAA on Guam and a cause of ALS/PDC. Subsequently, the hypothesis was expanded to include a causative role for BMAA in other neurodegenerative diseases including Alzheimer's disease (AD) through exposures attributed to proximity to freshwaters and/or consumption of seafood due to its purported production by most species of cyanobacteria. The hypothesis that BMAA is the critical factor in the genesis of these neurodegenerative diseases received considerable attention in the medical, scientific, and public arenas. This review examines the history of ALS/PDC and the BMAA-human disease hypotheses; similarities and differences between ALS/PDC and the other diseases with similar symptomologies; the relationship of ALS/PDC to other similar diseases, studies of BMAA-mediated effects in lab animals, inconsistencies and data gaps in the hypothesis; and other compounds and agents that were suggested as the cause of ALS/PDC on Guam. The review concludes that the hypothesis of a causal BMAA neurodegenerative disease relationship is not supported by existing data.
Collapse
Affiliation(s)
- N. Chernoff
- U.S. Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, NC, USA
| | - D. J. Hill
- U.S. Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, NC, USA
| | - D. L. Diggs
- Oak Ridge Institute for Science and Education Internship/Research Participation Program at the U.S. Environmental Protection Agency, NHEERL, Research Triangle Park, NC, USA
| | - B. D. Faison
- U.S. Environmental Protection Agency, Office of Water, Office of Science and Technology, Washington, DC, USA
| | - B. M. Francis
- Department of Entomology, University of Illinois, Champaign-Urbana, IL, USA
| | - J. R Lang
- Oak Ridge Institute for Science and Education Internship/Research Participation Program at the U.S. Environmental Protection Agency, NHEERL, Research Triangle Park, NC, USA
| | - M. M. Larue
- Oak Ridge Institute for Science and Education Internship/Research Participation Program at the U.S. Environmental Protection Agency, NHEERL, Research Triangle Park, NC, USA
| | - T.-T. Le
- Oak Ridge Institute for Science and Education Internship/Research Participation Program at the U.S. Environmental Protection Agency, NHEERL, Research Triangle Park, NC, USA
| | | | - J. N. Lugo
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, USA
| | - J. E. Schmid
- U.S. Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, NC, USA
| | - W. M. Winnik
- U.S. Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, NC, USA
| |
Collapse
|
34
|
Rodgers KJ, Main BJ, Samardzic K. Cyanobacterial Neurotoxins: Their Occurrence and Mechanisms of Toxicity. Neurotox Res 2017; 33:168-177. [DOI: 10.1007/s12640-017-9757-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/17/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022]
|
35
|
Cox PA, Kostrzewa RM, Guillemin GJ. BMAA and Neurodegenerative Illness. Neurotox Res 2017; 33:178-183. [PMID: 28540663 DOI: 10.1007/s12640-017-9753-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 05/12/2017] [Indexed: 01/02/2023]
Abstract
The cyanobacterial toxin β-N-methylamino-L-alanine (BMAA) now appears to be a cause of Guamanian amyotrophic lateral sclerosis/parkinsonism dementia complex (ALS/PDC). Its production by cyanobacteria throughout the world combined with multiple mechanisms of BMAA neurotoxicity, particularly to vulnerable subpopulations of motor neurons, has significantly increased interest in investigating exposure to this non-protein amino acid as a possible risk factor for other forms of neurodegenerative illness. We here provide a brief overview of BMAA studies and provide an introduction to this collection of scientific manuscripts in this special issue on BMAA.
Collapse
Affiliation(s)
- Paul Alan Cox
- Brain Chemistry Labs, Institute for Ethnomedicine, PO Box 3464, Jackson Hole, WY, 83001, USA.
| | - Richard M Kostrzewa
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, PO Box 70577, Johnson City, TN, 37614, USA
| | - Gilles J Guillemin
- Macquarie University, MND Research Centre, FMHS, 2 Technology Place, Sydney, NSW, 2109, Australia
| |
Collapse
|
36
|
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: 2.0] [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
|
37
|
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
|
38
|
Potjewyd G, Day PJ, Shangula S, Margison GP, Povey AC. L-β-N-methylamino-l-alanine (BMAA) nitrosation generates a cytotoxic DNA damaging alkylating agent: An unexplored mechanism for neurodegenerative disease. Neurotoxicology 2017; 59:105-109. [PMID: 28163087 DOI: 10.1016/j.neuro.2017.01.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/26/2017] [Accepted: 01/26/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND L-β-N-methylamino-l-alanine (BMAA) is a non-proteinic amino acid, that is neurotoxic in vitro and in animals, and is implicated in the causation of amyotrophic lateral sclerosis and parkinsonism-dementia complex (ALS-PDC) on Guam. Given that natural amino acids can be N-nitrosated to form toxic alkylating agents and the structural similarity of BMAA to other amino acids, our hypothesis was that N-nitrosation of BMAA might result in a toxic alkylating agent, providing a novel mechanistic hypothesis for BMAA action. FINDINGS We have chemically nitrosated BMAA with sodium nitrite to produce nitrosated BMAA (N-BMAA) which was shown to react with the alkyl-trapping agent, 4-(p-nitrobenzyl)pyridine, cause DNA strand breaks in vitro and was toxic to the human neuroblastoma cell line SH-SY5Y under conditions in which BMAA itself was minimally toxic. CONCLUSIONS Our results indicate that N-BMAA is an alkylating agent and toxin suggesting a plausible and previously unrecognised mechanism for the neurotoxic effects of BMAA.
Collapse
Affiliation(s)
- G Potjewyd
- Centre for Epidemiology, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - P J Day
- Centre for Epidemiology, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; Manchester Institute for Biotechnology, University of Manchester, Manchester, UK
| | - S Shangula
- Centre for Epidemiology, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - G P Margison
- Centre for Epidemiology, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - A C Povey
- Centre for Epidemiology, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
39
|
de Munck E, Palomo V, Muñoz-Sáez E, Perez DI, Gómez-Miguel B, Solas MT, Gil C, Martínez A, Arahuetes RM. Small GSK-3 Inhibitor Shows Efficacy in a Motor Neuron Disease Murine Model Modulating Autophagy. PLoS One 2016; 11:e0162723. [PMID: 27631495 PMCID: PMC5025054 DOI: 10.1371/journal.pone.0162723] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/26/2016] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive motor neuron degenerative disease that has no effective treatment up to date. Drug discovery tasks have been hampered due to the lack of knowledge in its molecular etiology together with the limited animal models for research. Recently, a motor neuron disease animal model has been developed using β-N-methylamino-L-alanine (L-BMAA), a neurotoxic amino acid related to the appearing of ALS. In the present work, the neuroprotective role of VP2.51, a small heterocyclic GSK-3 inhibitor, is analysed in this novel murine model together with the analysis of autophagy. VP2.51 daily administration for two weeks, starting the first day after L-BMAA treatment, leads to total recovery of neurological symptoms and prevents the activation of autophagic processes in rats. These results show that the L-BMAA murine model can be used to test the efficacy of new drugs. In addition, the results confirm the therapeutic potential of GSK-3 inhibitors, and specially VP2.51, for the disease-modifying future treatment of motor neuron disorders like ALS.
Collapse
Affiliation(s)
- Estefanía de Munck
- Departamento de Biología Animal II, Universidad Complutense de Madrid, Ciudad Universitaria, Madrid, Spain
| | - Valle Palomo
- Centro de Investigaciones Biológicas-CSIC, Ramiro de Maetzu 9, Madrid, Spain
| | - Emma Muñoz-Sáez
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense de Madrid, Ciudad Universitaria, Madrid, Spain
| | - Daniel I. Perez
- Centro de Investigaciones Biológicas-CSIC, Ramiro de Maetzu 9, Madrid, Spain
| | - Begoña Gómez-Miguel
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense de Madrid, Ciudad Universitaria, Madrid, Spain
| | - M. Teresa Solas
- Departamento de Biología Celular, Universidad Complutense de Madrid, Ciudad Universitaria, Madrid, Spain
| | - Carmen Gil
- Centro de Investigaciones Biológicas-CSIC, Ramiro de Maetzu 9, Madrid, Spain
| | - Ana Martínez
- Centro de Investigaciones Biológicas-CSIC, Ramiro de Maetzu 9, Madrid, Spain
- * E-mail: (AM); (RMA)
| | - Rosa M. Arahuetes
- Departamento de Biología Animal II, Universidad Complutense de Madrid, Ciudad Universitaria, Madrid, Spain
- * E-mail: (AM); (RMA)
| |
Collapse
|
40
|
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]
|
41
|
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
|
42
|
Theme 11 ALS Pathogenesis and Neurotoxicity. Amyotroph Lateral Scler Frontotemporal Degener 2015; 16 Suppl 1:186-205. [DOI: 10.3109/21678421.2015.1098816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
43
|
de Pedro N, Cantizani J, Ortiz-López FJ, González-Menéndez V, Cautain B, Rodríguez L, Bills GF, Reyes F, Genilloud O, Vicente F. Protective effects of isolecanoric acid on neurodegenerative in vitro models. Neuropharmacology 2015; 101:538-48. [PMID: 26455662 DOI: 10.1016/j.neuropharm.2015.09.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 09/23/2015] [Accepted: 09/25/2015] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) and Amyotrophic lateral sclerosis (ALS), are neurodegenerative disorders characterized by loss of dopaminergic or motor neurons, respectively. Although understanding of the PD and ALS pathogenesis remains incomplete, increasing evidence from human and animal studies has suggested that aberrant GSK3β, oxidative stress and mitochondrial damage are involved in their pathogenesis. Using two different molecular models, treatment with L-BMAA for ALS and rotenone for PD the effect of isolecanoric acid, a natural product isolated from a fungal culture, was evaluated. Pre-treatment with this molecule caused inhibition of GSK3β and CK1, and a decrease in oxidative stress, mitochondrial damage, apoptosis and cell death. Taken together, these results indicated that isolecanoric acid might have a protective effect against the development of these neurodegenerative disorders.
Collapse
Affiliation(s)
- Nuria de Pedro
- Fundación MEDINA, Parque Tecnológico Ciencias de la Salud, Avda. Conocimiento 34, 18016 Granada, Spain.
| | - Juan Cantizani
- Fundación MEDINA, Parque Tecnológico Ciencias de la Salud, Avda. Conocimiento 34, 18016 Granada, Spain
| | | | - Victor González-Menéndez
- Fundación MEDINA, Parque Tecnológico Ciencias de la Salud, Avda. Conocimiento 34, 18016 Granada, Spain
| | - Bastien Cautain
- Fundación MEDINA, Parque Tecnológico Ciencias de la Salud, Avda. Conocimiento 34, 18016 Granada, Spain
| | - Lorena Rodríguez
- Fundación MEDINA, Parque Tecnológico Ciencias de la Salud, Avda. Conocimiento 34, 18016 Granada, Spain
| | - Gerald F Bills
- Texas Therapeutics Institute, University of Texas Health Science Center at Houston, United States
| | - Fernando Reyes
- Fundación MEDINA, Parque Tecnológico Ciencias de la Salud, Avda. Conocimiento 34, 18016 Granada, Spain
| | - Olga Genilloud
- Fundación MEDINA, Parque Tecnológico Ciencias de la Salud, Avda. Conocimiento 34, 18016 Granada, Spain
| | - Francisca Vicente
- Fundación MEDINA, Parque Tecnológico Ciencias de la Salud, Avda. Conocimiento 34, 18016 Granada, Spain
| |
Collapse
|
44
|
Alves CJ, Maximino JR, Chadi G. Dysregulated expression of death, stress and mitochondrion related genes in the sciatic nerve of presymptomatic SOD1(G93A) mouse model of Amyotrophic Lateral Sclerosis. Front Cell Neurosci 2015; 9:332. [PMID: 26339226 PMCID: PMC4555015 DOI: 10.3389/fncel.2015.00332] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/10/2015] [Indexed: 12/11/2022] Open
Abstract
Schwann cells are the main source of paracrine support to motor neurons. Oxidative stress and mitochondrial dysfunction have been correlated to motor neuron death in Amyotrophic Lateral Sclerosis (ALS). Despite the involvement of Schwann cells in early neuromuscular disruption in ALS, detailed molecular events of a dying-back triggering are unknown. Sciatic nerves of presymptomatic (60-day-old) SOD1(G93A) mice were submitted to a high-density oligonucleotide microarray analysis. DAVID demonstrated the deregulated genes related to death, stress and mitochondrion, which allowed the identification of Cell cycle, ErbB signaling, Tryptophan metabolism and Rig-I-like receptor signaling as the most representative KEGG pathways. The protein-protein interaction networks based upon deregulated genes have identified the top hubs (TRAF2, H2AFX, E2F1, FOXO3, MSH2, NGFR, TGFBR1) and bottlenecks (TRAF2, E2F1, CDKN1B, TWIST1, FOXO3). Schwann cells were enriched from the sciatic nerve of presymptomatic mice using flow cytometry cell sorting. qPCR showed the up regulated (Ngfr, Cdnkn1b, E2f1, Traf2 and Erbb3, H2afx, Cdkn1a, Hspa1, Prdx, Mapk10) and down-regulated (Foxo3, Mtor) genes in the enriched Schwann cells. In conclusion, molecular analyses in the presymptomatic sciatic nerve demonstrated the involvement of death, oxidative stress, and mitochondrial pathways in the Schwann cell non-autonomous mechanisms in the early stages of ALS.
Collapse
Affiliation(s)
- Chrystian J Alves
- Department of Neurology, Neuroregeneration Center, University of São Paulo School of Medicine São Paulo, Brazil
| | - Jessica R Maximino
- Department of Neurology, Neuroregeneration Center, University of São Paulo School of Medicine São Paulo, Brazil
| | - Gerson Chadi
- Department of Neurology, Neuroregeneration Center, University of São Paulo School of Medicine São Paulo, Brazil
| |
Collapse
|
45
|
Bradley WG. The John Walton Muscular Dystrophy Research Centre in the University of Newcastle and the BMAA Theory of Motor Neuron Disease. J Neuromuscul Dis 2015; 2:S77-S81. [PMID: 27858760 PMCID: PMC5271429 DOI: 10.3233/jnd-150090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Affiliation(s)
- Walter G. Bradley
- Correspondence to: Walter G. Bradley, Professor and Chairman Emeritus, Department of Neurology, Miller School of Medicine, University of Miami, USA. Tel.: +305 215 2144; Fax: +305 964 5336;
| |
Collapse
|
46
|
de Munck E, Muñoz-Sáez E, Miguel BG, Solas MT, Martínez A, Arahuetes RM. Morphometric and neurochemical alterations found in l-BMAA treated rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 39:1232-45. [PMID: 26002186 DOI: 10.1016/j.etap.2015.04.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Accepted: 04/30/2015] [Indexed: 05/03/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive muscle paralysis that reflects the motoneurons' degeneration. Several studies support the relationship between β-N-methylamino-l-alanine (l-BMAA), a neurotoxic amino acid produced by cyanobacteria and diatoms, and the sporadic occurrence of ALS and other neurodegenerative diseases. Therefore, the study of its neurotoxicity mechanisms has assumed great relevance in recent years. Recently, our research team has proposed a sporadic ALS animal model by l-BMAA administration in rats, which displays many pathophysiological features of human ALS. In this paper, we deepen the characterization of this model corroborating the occurrence of alterations present in ALS patients such as decreased muscle volume, thinning of the motor cortex, enlarged brain's lateral ventricles, and alteration of both bulbar nuclei and neurotransmitters' levels. Therefore, we conclude that l-BMAA treated rats could be a good model which mimics degenerative features that ALS causes in humans.
Collapse
Affiliation(s)
- Estefanía de Munck
- Departamento de Biología Animal II, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Emma Muñoz-Sáez
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Begoña G Miguel
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - M Teresa Solas
- Departamento de Biología Celular (Morfología Microscópica), Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Ana Martínez
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, 28040 Madrid, Spain.
| | - Rosa M Arahuetes
- Departamento de Biología Animal II, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| |
Collapse
|
47
|
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
|
48
|
García-Escudero V, Rosales M, Muñoz JL, Scola E, Medina J, Khalique H, Garaulet G, Rodriguez A, Lim F. Patient-derived olfactory mucosa for study of the non-neuronal contribution to amyotrophic lateral sclerosis pathology. J Cell Mol Med 2015; 19:1284-95. [PMID: 25807871 PMCID: PMC4459844 DOI: 10.1111/jcmm.12488] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 10/10/2014] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a degenerative motor neuron disease which currently has no cure. Research using rodent ALS models transgenic for mutant superoxide dismutase 1 (SOD1) has implicated that glial–neuronal interactions play a major role in the destruction of motor neurons, but the generality of this mechanism is not clear as SOD1 mutations only account for less than 2% of all ALS cases. Recently, this hypothesis was backed up by observation of similar effects using astrocytes derived from post-mortem spinal cord tissue of ALS patients which did not carry SOD1 mutations. However, such necropsy samples may not be easy to obtain and may not always yield viable cell cultures. Here, we have analysed olfactory mucosa (OM) cells, which can be easily isolated from living ALS patients. Disease-specific changes observed when ALS OM cells were co-cultured with human spinal cord neurons included decreased neuronal viability, aberrant neuronal morphology and altered glial inflammatory responses. Our results show the potential of OM cells as new cell models for ALS.
Collapse
Affiliation(s)
- Vega García-Escudero
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" (C.S.I.C.- U.A.M.), Universidad Autónoma de Madrid, Madrid, Spain
| | - María Rosales
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - José Luis Muñoz
- Departamento de Neurología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Esteban Scola
- Departamento de Otorrinolaringología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Javier Medina
- Departamento de Otorrinolaringología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Hena Khalique
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Guillermo Garaulet
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio Rodriguez
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Filip Lim
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
49
|
Muñoz-Sáez E, de Munck García E, Arahuetes Portero RM, Vicente F, Ortiz-López FJ, Cantizani J, Gómez Miguel B. Neuroprotective role of sphingosine-1-phosphate in L-BMAA treated neuroblastoma cells (SH-SY5Y). Neurosci Lett 2015; 593:83-9. [PMID: 25769802 DOI: 10.1016/j.neulet.2015.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/14/2015] [Accepted: 03/06/2015] [Indexed: 10/23/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive lipid which regulates proliferation, cell migration, survival and differentiation by specific receptors activation. We studied its effects on L-BMAA treated neuroblastoma cells (SH-SY5Y), an amino acid that can trigger neurodegenerative diseases such as amyotrophic lateral sclerosis/Parkinson dementia complex (ALS/PDC). We found that S1P protects from necrosis and prevents the GSK3 increasing as long as the PI3K/AKT pathway is active. Moreover, GSK3 inhibition protects against neuronal death caused by L-BMAA.
Collapse
Affiliation(s)
- Emma Muñoz-Sáez
- Departamento de Bioquímica y Biología Molecular-I, Universidad Complutense de Madrid, 28040-Madrid, Spain.
| | | | | | - Francisca Vicente
- Fundación MEDINA, Centro Excelencia Investigación Medicamentos Innovadores en Andalucía, Parque Tecnológico Ciencias de la Salud, 18016-Armilla-Granada, Spain
| | - Francisco Javier Ortiz-López
- Fundación MEDINA, Centro Excelencia Investigación Medicamentos Innovadores en Andalucía, Parque Tecnológico Ciencias de la Salud, 18016-Armilla-Granada, Spain
| | - Juan Cantizani
- Fundación MEDINA, Centro Excelencia Investigación Medicamentos Innovadores en Andalucía, Parque Tecnológico Ciencias de la Salud, 18016-Armilla-Granada, Spain
| | - Begoña Gómez Miguel
- Departamento de Bioquímica y Biología Molecular-I, Universidad Complutense de Madrid, 28040-Madrid, Spain
| |
Collapse
|
50
|
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
|