1
|
Kim SY, Kim M, Park K, Hong S. A systematic review on analytical methods of the neurotoxin β-N-methylamino-L-alanine (BMAA), and its causative microalgae and distribution in the environment. CHEMOSPHERE 2024; 366:143487. [PMID: 39395475 DOI: 10.1016/j.chemosphere.2024.143487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/20/2024] [Accepted: 10/04/2024] [Indexed: 10/14/2024]
Abstract
β-N-Methylamino-L-alanine (BMAA), a neurotoxin produced by various microalgal groups, is associated with neurodegenerative diseases and is considered a major environmental factor potentially linked to sporadic amyotrophic lateral sclerosis. This study systematically reviews the analytical methods used to study BMAA in publications from 2019 to the present. It also investigates the causative microalgae of BMAA and its geographical distributions in aquatic ecosystems based on studies conducted since 2003. A comprehensive search using the Web of Science database revealed that hydrolysis for extraction (67%), followed by quantification using LC-MS/MS (LC: 84%; MS/MS: 88%), is the most commonly employed method in BMAA analysis. Among analytical methods, RPLC-MS/MS had the highest percentage (88%) of BMAA-positive results and included a high number of quality control (QC) assessments. Various genera of cyanobacteria and diatoms have been reported to produce BMAA. The widespread geographical distribution of BMAA across diverse ecosystems highlights significant environmental and public health concerns. Notably, BMAA accumulation and biomagnification are likely more potent in marine or brackish water ecosystems than in freshwater ecosystems, potentially amplifying its ecological impacts. Future research should prioritize advanced, sensitive methods, particularly LC-MS/MS with as many QC assessments as possible, and should expand investigations to identify novel microalgal producers and previously uncharted geographical areas, with a special focus on marine or brackish water ecosystems. This effort will enhance our understanding of the environmental distribution and impacts of BMAA.
Collapse
Affiliation(s)
- Sea-Yong Kim
- Department of Marine Environmental Sciences & Institute of Marine Environmental Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Mungi Kim
- Department of Earth, Environmental & Space Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Kiho Park
- Department of Earth, Environmental & Space Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Seongjin Hong
- Department of Marine Environmental Sciences & Institute of Marine Environmental Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea; Department of Earth, Environmental & Space Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
2
|
Ma Y, Tang P, Xu J, Li T, Zhang J, Li H, Bai Y, Wang Q, Wang Q. The role of neutrophil extracellular traps in β-methylamino L-alanine-induced liver injury in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 281:116678. [PMID: 38964067 DOI: 10.1016/j.ecoenv.2024.116678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/06/2024]
Abstract
The non-protein amino acid β-N-methylamino-L-alanine (BMAA), produced by cyanobacteria, has been recognized as a neurotoxin. L-serine as an antagonist of BMAA can effectively alleviate BMAA-induced neurotoxicity. Although BMAA has long been emphasized as a neurotoxin, with the emergence of BMAA detected in a variety of algae in freshwater around the world and its clear biological enrichment effect, it is particularly important to study the non-neurotoxic adverse effects of BMAA. However, there is only limited evidence to support the ability of BMAA to cause oxidative damage in the liver. The exact molecular mechanism of BMAA-induced liver injury is still unclear. The formation of neutrophil extracellular traps (NETs) is a 'double-edged sword' for the organism, excessive formation of NETs is associated with inflammatory diseases of the liver. Our results innovatively confirmed that BMAA was able to cause the formation of NETs in the liver during the liver injury. The possible mechanism may associated with the regulation of ERK/p38 and cGAS/STING signaling pathways. The massive formation of NETs was able to exacerbate the BMAA-induced oxidative stress and release of inflammatory factors in the mice liver. And the removal of NETs could alleviate this injury. This article will bring a new laboratory evidence for BMAA-induced non-neurotoxicity and immunotoxicity.
Collapse
Affiliation(s)
- Yu Ma
- Department of Health Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, PR China
| | - Peiyan Tang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian 116044, PR China
| | - Jiaqi Xu
- Department of Health Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, PR China
| | - Tao Li
- Department of Health Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, PR China
| | - Jiahang Zhang
- Department of Health Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, PR China
| | - Haidong Li
- First Affiliated Hospital of Dalian Medical University, Dalian 116044, PR China
| | - Yutan Bai
- First Affiliated Hospital of Dalian Medical University, Dalian 116044, PR China
| | - Qinghui Wang
- Department of Anesthesiology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116023, China.
| | - Qingshan Wang
- Department of Health Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, PR China; National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian 116044, PR China.
| |
Collapse
|
3
|
Liu N, Yu W, Sun M, Zhou D, Sun J, Jiang T, Zhang W, Wang M. Research trends and hotspots of ferroptosis in neurodegenerative diseases from 2013 to 2023: A bibliometrics study. Heliyon 2024; 10:e29418. [PMID: 38638970 PMCID: PMC11024616 DOI: 10.1016/j.heliyon.2024.e29418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/30/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024] Open
Abstract
Background With the aging population, the incidence of neurodegenerative diseases increases yearly, seriously impacting human health. Various journals have published studies on the pathogenesis of ferroptosis in neurodegenerative diseases. However, bibliometric analysis in this field is still lacking. The study aims to visually analyze global research trends in this field over the past decade. Methods The articles and reviews regarding ferroptosis in neurodegenerative diseases were retrieved from the Web of Science on September 1, 2023. Citespace [version 6.2. R4 (64-bit)] and VOSviewer (version 1.6.18) were used to conduct the bibliometric and knowledge-map analysis. Results In total, 370 studies were included in the paper and ranked by their citation frequency. Many articles on ferroptosis in neurodegenerative diseases have been published in the past decade. The country, institution, author, and journal with the highest publications were China, Guangzhou Medical University, Maher, Pamela, and Free Radical Biology And Medicine, respectively. The analysis of keyword co-occurrence indicated that research frontiers were molecular mechanisms of ferroptosis in neurodegenerative diseases, especially a few key pathways that triggered ferroptosis in these diseases, including lipid peroxidation signaling, iron metabolism, and GSH/GPX4 signaling. In addition, ferroptosis inhibitors such as liproxstatins and ferrostatins had protective effects in animal models of neurodegenerative diseases. Therefore, future attention should also be focused on therapeutic drugs that target ferroptosis. Conclusion This study comprehensively analyzed the publications on ferroptosis in neurodegenerative diseases from a bibliometric perspective. Research on this topic is currently expanding at a rapid pace, and the China holds a leading position in this field by its scientific achievements and productivity. Moreover, the research frontiers were molecular mechanisms of ferroptosis in neurodegenerative diseases and developing targeted therapeutic drugs. In summary, our results showed an all-sided overview of the knowledge atlas and a valuable reference for the future research in this field.
Collapse
Affiliation(s)
- Ning Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Wuhan Yu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- Department of Anus and Intestine Surgery, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, 523000, China
| | - Mengjiao Sun
- Capital Medical University, Beijing, 100000, China
| | - Dan Zhou
- Department of Neurology, Xi ‘an Ninth Hospital, Xi ‘an, 710000, China
| | - Jing Sun
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Taotao Jiang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Wenjing Zhang
- Department of Neurology, Qinghai Provincial People's Hospital, Qinghai, 810000, China
| | - Manxia Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| |
Collapse
|
4
|
Li M, Qiu J, Yan G, Zheng X, Li A. How does the neurotoxin β-N-methylamino-L-alanine exist in biological matrices and cause toxicity? THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 922:171255. [PMID: 38417517 DOI: 10.1016/j.scitotenv.2024.171255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/01/2024]
Abstract
The neurotoxin β-N-methylamino-L-alanine (BMAA) has been deemed as a risk factor for some neurodegenerative diseases such as amyotrophic lateral sclerosis/parkinsonism dementia complex (ALS/PDC). This possible link has been proved in some primate models and cell cultures with the appearance that BMAA exposure can cause excitotoxicity, formation of protein aggregates, and/or oxidative stress. The neurotoxin BMAA extensively exists in the environment and can be transferred through the food web to human beings. In this review, the occurrence, toxicological mechanisms, and characteristics of BMAA were comprehensively summarized, and proteins and peptides were speculated as its possible binding substances in biological matrices. It is difficult to compare the published data from previous studies due to the inconsistent analytical methods and components of BMAA. The binding characteristics of BMAA should be focused on to improve our understanding of its health risk to human health in the future.
Collapse
Affiliation(s)
- Min Li
- College of Environmental Science and Engineering, Ocean University of China, Qingdao 266100, China
| | - Jiangbing Qiu
- College of Environmental Science and Engineering, Ocean University of China, Qingdao 266100, China; Key Laboratory of Marine Environment and Ecology, Ocean University of China, Ministry of Education, Qingdao 266100, China
| | - Guowang Yan
- College of Environmental Science and Engineering, Ocean University of China, Qingdao 266100, China
| | - Xianyao Zheng
- College of Environmental Science and Engineering, Ocean University of China, Qingdao 266100, China
| | - Aifeng Li
- College of Environmental Science and Engineering, Ocean University of China, Qingdao 266100, China; Key Laboratory of Marine Environment and Ecology, Ocean University of China, Ministry of Education, Qingdao 266100, China.
| |
Collapse
|
5
|
Bishop SL, Solonenka JT, Giebelhaus RT, Bakker DTR, Li ITS, Murch SJ. Microbial Diversity Impacts Non-Protein Amino Acid Production in Cyanobacterial Bloom Cultures Collected from Lake Winnipeg. Toxins (Basel) 2024; 16:169. [PMID: 38668594 PMCID: PMC11053616 DOI: 10.3390/toxins16040169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/10/2024] [Accepted: 03/20/2024] [Indexed: 04/29/2024] Open
Abstract
Lake Winnipeg in Manitoba, Canada is heavily impacted by harmful algal blooms that contain non-protein amino acids (NPAAs) produced by cyanobacteria: N-(2-aminoethyl)glycine (AEG), β-aminomethyl-L-alanine (BAMA), β-N-methylamino-L-alanine (BMAA), and 2,4-diaminobutyric acid (DAB). Our objective was to investigate the impact of microbial diversity on NPAA production by cyanobacteria using semi-purified crude cyanobacterial cultures established from field samples collected by the Lake Winnipeg Research Consortium between 2016 and 2021. NPAAs were detected and quantified by ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) using validated analytical methods, while Shannon and Simpson alpha diversity scores were determined from 16S rRNA metagenomic sequences. Alpha diversity in isolate cultures was significantly decreased compared to crude cyanobacterial cultures (p < 0.001), indicating successful semi-purification. BMAA and AEG concentrations were higher in crude compared to isolate cultures (p < 0.0001), and AEG concentrations were correlated to the alpha diversity in cultures (r = 0.554; p < 0.0001). BAMA concentrations were increased in isolate cultures (p < 0.05), while DAB concentrations were similar in crude and isolate cultures. These results demonstrate that microbial community complexity impacts NPAA production by cyanobacteria and related organisms.
Collapse
Affiliation(s)
- Stephanie L. Bishop
- Department of Chemistry, University of British Columbia, Syilx Okanagan Nation Territory, Kelowna, BC V1V 1V7, Canada; (J.T.S.); (R.T.G.); (D.T.R.B.); (I.T.S.L.); (S.J.M.)
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Julia T. Solonenka
- Department of Chemistry, University of British Columbia, Syilx Okanagan Nation Territory, Kelowna, BC V1V 1V7, Canada; (J.T.S.); (R.T.G.); (D.T.R.B.); (I.T.S.L.); (S.J.M.)
| | - Ryland T. Giebelhaus
- Department of Chemistry, University of British Columbia, Syilx Okanagan Nation Territory, Kelowna, BC V1V 1V7, Canada; (J.T.S.); (R.T.G.); (D.T.R.B.); (I.T.S.L.); (S.J.M.)
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2N4, Canada
- The Metabolomics Innovation Centre, Edmonton, AB T6G 2N4, Canada
| | - David T. R. Bakker
- Department of Chemistry, University of British Columbia, Syilx Okanagan Nation Territory, Kelowna, BC V1V 1V7, Canada; (J.T.S.); (R.T.G.); (D.T.R.B.); (I.T.S.L.); (S.J.M.)
| | - Isaac T. S. Li
- Department of Chemistry, University of British Columbia, Syilx Okanagan Nation Territory, Kelowna, BC V1V 1V7, Canada; (J.T.S.); (R.T.G.); (D.T.R.B.); (I.T.S.L.); (S.J.M.)
| | - Susan J. Murch
- Department of Chemistry, University of British Columbia, Syilx Okanagan Nation Territory, Kelowna, BC V1V 1V7, Canada; (J.T.S.); (R.T.G.); (D.T.R.B.); (I.T.S.L.); (S.J.M.)
| |
Collapse
|
6
|
Lee A, Henderson R, Aylward J, McCombe P. Gut Symptoms, Gut Dysbiosis and Gut-Derived Toxins in ALS. Int J Mol Sci 2024; 25:1871. [PMID: 38339149 PMCID: PMC10856138 DOI: 10.3390/ijms25031871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Many pathogenetic mechanisms have been proposed for amyotrophic lateral sclerosis (ALS). Recently, there have been emerging suggestions of a possible role for the gut microbiota. Gut microbiota have a range of functions and could influence ALS by several mechanisms. Here, we review the possible role of gut-derived neurotoxins/excitotoxins. We review the evidence of gut symptoms and gut dysbiosis in ALS. We then examine a possible role for gut-derived toxins by reviewing the evidence that these molecules are toxic to the central nervous system, evidence of their association with ALS, the existence of biochemical pathways by which these molecules could be produced by the gut microbiota and existence of mechanisms of transport from the gut to the blood and brain. We then present evidence that there are increased levels of these toxins in the blood of some ALS patients. We review the effects of therapies that attempt to alter the gut microbiota or ameliorate the biochemical effects of gut toxins. It is possible that gut dysbiosis contributes to elevated levels of toxins and that these could potentially contribute to ALS pathogenesis, but more work is required.
Collapse
Affiliation(s)
- Aven Lee
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia; (R.H.); (P.M.)
| | - Robert Henderson
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia; (R.H.); (P.M.)
- Department of Neurology, Royal Brisbane & Women’s Hospital, Brisbane, QLD 4029, Australia
- Wesley Research Institute, The Wesley Hospital, Auchenflower, QLD 4066, Australia;
| | - James Aylward
- Wesley Research Institute, The Wesley Hospital, Auchenflower, QLD 4066, Australia;
| | - Pamela McCombe
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia; (R.H.); (P.M.)
- Department of Neurology, Royal Brisbane & Women’s Hospital, Brisbane, QLD 4029, Australia
- Wesley Research Institute, The Wesley Hospital, Auchenflower, QLD 4066, Australia;
| |
Collapse
|
7
|
Gutiérrez-García K, Whitaker MRL, Bustos-Díaz ED, Salzman S, Ramos-Aboites HE, Reitz ZL, Pierce NE, Cibrián-Jaramillo A, Barona-Gómez F. Gut microbiomes of cycad-feeding insects tolerant to β-methylamino-L-alanine (BMAA) are rich in siderophore biosynthesis. ISME COMMUNICATIONS 2023; 3:122. [PMID: 37993724 PMCID: PMC10665472 DOI: 10.1038/s43705-023-00323-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 10/17/2023] [Accepted: 10/25/2023] [Indexed: 11/24/2023]
Abstract
Ingestion of the cycad toxins β-methylamino-L-alanine (BMAA) and azoxyglycosides is harmful to diverse organisms. However, some insects are specialized to feed on toxin-rich cycads with apparent immunity. Some cycad-feeding insects possess a common set of gut bacteria, which might play a role in detoxifying cycad toxins. Here, we investigated the composition of gut microbiota from a worldwide sample of cycadivorous insects and characterized the biosynthetic potential of selected bacteria. Cycadivorous insects shared a core gut microbiome consisting of six bacterial taxa, mainly belonging to the Proteobacteria, which we were able to isolate. To further investigate selected taxa from diverging lineages, we performed shotgun metagenomic sequencing of co-cultured bacterial sub-communities. We characterized the biosynthetic potential of four bacteria from Serratia, Pantoea, and two different Stenotrophomonas lineages, and discovered a suite of biosynthetic gene clusters notably rich in siderophores. Siderophore semi-untargeted metabolomics revealed a broad range of chemically related yet diverse iron-chelating metabolites, including desferrioxamine B, suggesting the occurrence of an unprecedented desferrioxamine-like biosynthetic pathway that remains to be identified. These results provide a foundation for future investigations into how cycadivorous insects tolerate diets rich in azoxyglycosides, BMAA, and other cycad toxins, including a possible role for bacterial siderophores.
Collapse
Affiliation(s)
- Karina Gutiérrez-García
- Evolution of Metabolic Diversity Laboratory, Unidad de Genómica Avanzada (Langebio), Cinvestav-IPN, Km 9.6 Libramiento Irapuato - León, Irapuato, Guanajuato, 36824, México
- Department of Embryology, Carnegie Institution for Science, 3520 San Martin Drive, Baltimore, MD, 21218, USA
| | - Melissa R L Whitaker
- Museum of Comparative Zoology, Department of Organismic and Evolutionary Biology, Harvard University, 26 Oxford Street, Cambridge, MA, 02138, USA.
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37614, USA.
| | - Edder D Bustos-Díaz
- Evolution of Metabolic Diversity Laboratory, Unidad de Genómica Avanzada (Langebio), Cinvestav-IPN, Km 9.6 Libramiento Irapuato - León, Irapuato, Guanajuato, 36824, México
- Institute of Biology, Leiden University, Sylviusweg 72, Leiden, 2333 BE, The Netherlands
| | - Shayla Salzman
- Museum of Comparative Zoology, Department of Organismic and Evolutionary Biology, Harvard University, 26 Oxford Street, Cambridge, MA, 02138, USA
- University of Georgia, Entomology Department, Athens, GA, 30602, USA
| | - Hilda E Ramos-Aboites
- Evolution of Metabolic Diversity Laboratory, Unidad de Genómica Avanzada (Langebio), Cinvestav-IPN, Km 9.6 Libramiento Irapuato - León, Irapuato, Guanajuato, 36824, México
| | - Zachary L Reitz
- Bioinformatics Group, Wageningen University, Droevendaalsesteeg 1, 6708PB, Wageningen, The Netherlands
| | - Naomi E Pierce
- Museum of Comparative Zoology, Department of Organismic and Evolutionary Biology, Harvard University, 26 Oxford Street, Cambridge, MA, 02138, USA
| | - Angélica Cibrián-Jaramillo
- Ecological and Evolutionary Genomics Laboratory, Unidad de Genómica Avanzada (Langebio), Cinvestav-IPN, Km 9.6 Libramiento Irapuato - León, Irapuato, Guanajuato, 36824, México
- Naturalis Biodiversity Center, Darwinweg 2, 2333 CR, Leiden, The Netherlands
| | - Francisco Barona-Gómez
- Evolution of Metabolic Diversity Laboratory, Unidad de Genómica Avanzada (Langebio), Cinvestav-IPN, Km 9.6 Libramiento Irapuato - León, Irapuato, Guanajuato, 36824, México.
- Institute of Biology, Leiden University, Sylviusweg 72, Leiden, 2333 BE, The Netherlands.
| |
Collapse
|
8
|
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: 1.0] [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
|
9
|
Shkodrova M, Mishonova M, Chichova M, Sazdova I, Ilieva B, Doncheva-Stoimenova D, Raikova N, Keremidarska-Markova M, Gagov H. β-N-Methylamino-L-Alanine (BMAA) Modulates the Sympathetic Regulation and Homeostasis of Polyamines. Toxins (Basel) 2023; 15:141. [PMID: 36828455 PMCID: PMC9960692 DOI: 10.3390/toxins15020141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
The neurotoxin β-N-methylamino-L-alanine (BMAA) is a non-proteinogenic amino acid produced by cyanobacteria. Non-neuronal toxicity of BMAA is poorly studied with a reported increase in reactive oxygen species and a decrease in the antioxidant capacity of liver, kidney, and colorectal adenocarcinoma cells. The aim of this research is to study the toxicity of BMAA (0.1-1 mM) on mitochondria and submitochondrial particles with ATPase activity, on the semicarbazide-sensitive amino oxidases (SSAOs) activity of rat liver, and on an in vitro model containing functionally active excitable tissues-regularly contracting heart muscle preparation with a preserved autonomic innervation. For the first time the BMAA-dependent inhibition of SSAO activity, the elimination of the positive inotropic effect of adrenergic innervation, and the direct and reversible inhibition of adrenaline signaling in ventricular myocytes with 1 mM BMAA were observed. Additionally, it is confirmed that 1 mM BMAA can activate mitochondrial ATPase indirectly. It is concluded that a higher dose of BMAA may influence multiple physiological and pathological processes as it slows down the degradation of biogenic amines, downregulates the sympathetic neuromediation, and embarrasses the cell signaling of adrenergic receptors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hristo Gagov
- Department of Animal and Human Physiology, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov blvd., 1164 Sofia, Bulgaria
| |
Collapse
|
10
|
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: 0] [Impact Index Per Article: 0] [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
|
11
|
Yan B, Wang S, Liu Z, Wang D, Shi W, Cui F. Degradation mechanisms of cyanobacteria neurotoxin β-N-methylamino-l-alanine (BMAA) during UV 254/H 2O 2 process: Kinetics and pathways. CHEMOSPHERE 2022; 302:134939. [PMID: 35561764 DOI: 10.1016/j.chemosphere.2022.134939] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 05/03/2022] [Accepted: 05/08/2022] [Indexed: 06/15/2023]
Abstract
In this work, the UV254/H2O2 process was utilized to remove β-N-methylamino-l-alanine (BMAA), a kind of cyanobacteria neurotoxin, and the influence of reaction parameters and environmental factors on the degradation of BMAA has been systematically investigated. The results showed that BMAA could be effectively removed in the UV254/H2O2 system compared to UV or H2O2 alone and OH was confirmed as the main ROS to degrade BMAA. The degradation rate of BMAA increased first and then decreased with the increase of pH and the maximum kobs was 0.1545 min-1 obtained at pH 9. The removal of BMAA in the UV254/H2O2 system was inhibited in actual water, while the degradation rate of BMAA in actual water could still exceed 90% by appropriately extending the reaction time. The decrease in the degradation efficiency of BMAA in actual water was primarily due to the ultraviolet light absorption and competition effects of NOM, and anions (Cl- and HCO3-) would also inhibit the degradation of BMAA. Five by-products ([M - H]- = 118, 103, 88, 87 and 59) were identified in this study and the degradation pathways of BMAA were proposed. The production of by-products was attributed to the fracture of the C-N bond and hydroxylation reaction. This study is worthwhile to deepen the understanding of the degradation mechanism of BMAA in the UV254/H2O2 system.
Collapse
Affiliation(s)
- Boyin Yan
- College of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, 266033, China
| | - Songxue Wang
- College of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, 266033, China
| | - Zhiquan Liu
- Institute of Environmental Research at Greater Bay, Key Laboratory by Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, 510006, China.
| | - Da Wang
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of an Environment, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Wenxin Shi
- School of Environment and Ecology, Chongqing University, Chongqing, 400044, China
| | - Fuyi Cui
- School of Environment and Ecology, Chongqing University, Chongqing, 400044, China
| |
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: 4.5] [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
|
Kim SY, Hedberg P, Winder M, Rydberg S. Evidence of 2,4-diaminobutyric acid (DAB) production as a defense mechanism in diatom Thalassiosira pseudonana. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 249:106210. [PMID: 35665646 DOI: 10.1016/j.aquatox.2022.106210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 06/15/2023]
Abstract
The neurotoxic secondary metabolite β-N-methylamino-L-alanine (BMAA) and its structural isomer 2,4-diaminobutyric acid (DAB) are known to be produced by various phytoplankton groups. Despite the worldwide spread of these toxin producers, no obvious role and function of BMAA and DAB in diatoms have been identified. Here, we investigated the effects of biotic factors, i.e., predators and competitors, as possible causes of BMAA and/or DAB regulation in the two diatom species Phaeodactylum tricornutum and Thalassiosira pseudonana. DAB was specifically regulated in T. pseudonana by the presence of predators and competitors. The effects of DAB on both diatoms as competitors and on the copepod Tigriopus sp. as predator at individual and at population levels were examined. The toxic effects of DAB on the growth of T. pseudonana and the population of Tigriopus sp. were significant. The effect of DAB as a defensive secondary metabolite is assumed to be environmentally relevant depending on the number of the copepods. The results show a potential function of DAB that can play an important role in defense mechanisms of T. pseudonana.
Collapse
Affiliation(s)
- Sea-Yong Kim
- Department of Ecology, Environment and Plant Sciences, Stockholm University, SE 10691 Stockholm, Sweden
| | - Per Hedberg
- Department of Ecology, Environment and Plant Sciences, Stockholm University, SE 10691 Stockholm, Sweden
| | - Monika Winder
- Department of Ecology, Environment and Plant Sciences, Stockholm University, SE 10691 Stockholm, Sweden
| | - Sara Rydberg
- Department of Ecology, Environment and Plant Sciences, Stockholm University, SE 10691 Stockholm, Sweden.
| |
Collapse
|
14
|
Kim SY, Rasmussen U, Rydberg S. Effect and function of β-N-methylamino-L-alanine in the diatom Phaeodactylum tricornutum. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 830:154778. [PMID: 35341850 DOI: 10.1016/j.scitotenv.2022.154778] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/17/2022] [Accepted: 03/19/2022] [Indexed: 06/14/2023]
Abstract
The neurotoxin β-N-methylamino-L-alanine (BMAA) is an environmental factor connected to neurodegenerative diseases. BMAA can be produced by various microorganisms (e.g. bacteria, cyanobacteria, dinoflagellates and diatoms) present in diverse ecosystems. No previous study has revealed the function of BMAA in diatoms. In the present study, we combined physiological data with metabolomic and transcriptional data in order to investigate the effect and function of BMAA in the diatom Phaeodactylum tricornutum. P. tricornutum, exposed to different concentrations of exogenous BMAA, showed concentration dependent responses. When the concentration of supplemented BMAA was sufficient to arrest the growth of P. tricornutum, oxidative stress and obstructed carbon fixation were obtained from the specific metabolite and transcriptional data. Results also indicated increased concentration of intracellular chlorophyll a and alterations in the GS-GOGAT cycle, whereas the urea cycle was suppressed. We therefore conclude that BMAA represents a toxic metabolite able to control the growth of P. tricornutum by triggering oxidative stress, and further influencing photosynthesis and nitrogen metabolisms.
Collapse
Affiliation(s)
- Sea-Yong Kim
- Department of Ecology, Environment and Plant Sciences, Stockholm University, SE 10691 Stockholm, Sweden
| | - Ulla Rasmussen
- Department of Ecology, Environment and Plant Sciences, Stockholm University, SE 10691 Stockholm, Sweden
| | - Sara Rydberg
- Department of Ecology, Environment and Plant Sciences, Stockholm University, SE 10691 Stockholm, Sweden.
| |
Collapse
|
15
|
Dias FRP, de Souza Almeida RR, Sovrani V, Thomaz NK, Gonçalves CA, Quincozes-Santos A, Bobermin LD. Glioprotective Effects of Resveratrol Against BMAA-Induced Astroglial Dysfunctions. Neurotox Res 2022; 40:530-541. [PMID: 35320508 DOI: 10.1007/s12640-022-00492-9] [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: 02/08/2022] [Revised: 03/13/2022] [Accepted: 03/17/2022] [Indexed: 11/28/2022]
Abstract
Astroglial cells play important roles in maintaining central nervous system (CNS) homeostasis. The neurotoxin β-N-methylamino-L-alanine (BMAA) has usually been associated with neurodegeneration due to its toxic effects on neurons. However, little is known about the effects of BMAA on astroglial cells. Resveratrol, a natural polyphenol, represents a potential protective strategy against brain injuries. In the present study, we sought to investigate BMAA-induced astroglial dysfunctions and the glioprotective roles of resveratrol. BMAA did not impair astroglial cellular viability, but increased glutamate uptake, glutamate metabolism into glutamine, and reactive oxygen species production, while decreased glutathione (GSH) and superoxide dismutase (SOD)-based antioxidant defenses and triggers an inflammatory response. In contrast, resveratrol was able to prevent most of these BMAA-induced functional changes in astroglial cells. Moreover, both BMAA and resveratrol modulated the gene expression of molecular pathways associated with glutamate metabolism, redox homeostasis, and inflammatory response, which characterize their roles on astroglial functions. In this regard, BMAA downregulated adenosine receptors, peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC-1α), phosphoinositide-3-kinase (PI3K), and Akt, while resveratrol prevented these effects and upregulated nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). Our study, for the first time, demonstrates that BMAA directly impacts key astroglial functions, contributing to elucidating the cellular and molecular mechanisms of this toxin in the CNS. In addition, we reinforce the glioprotective effects of resveratrol against BMAA-induced astroglial dysfunctions.
Collapse
Affiliation(s)
- Filipe Renato Pereira Dias
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rua Ramiro Barcelos, 2600 - Anexo, 90035-003, RS, Brazil
| | - Rômulo Rodrigo de Souza Almeida
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rua Ramiro Barcelos, 2600 - Anexo, 90035-003, RS, Brazil
| | - Vanessa Sovrani
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rua Ramiro Barcelos, 2600 - Anexo, 90035-003, RS, Brazil
| | - Natalie K Thomaz
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rua Ramiro Barcelos, 2600 - Anexo, 90035-003, RS, Brazil
| | - Carlos-Alberto Gonçalves
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rua Ramiro Barcelos, 2600 - Anexo, 90035-003, RS, Brazil
- Programa de Pós-Graduação Em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - André Quincozes-Santos
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rua Ramiro Barcelos, 2600 - Anexo, 90035-003, RS, Brazil
- Programa de Pós-Graduação Em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Larissa Daniele Bobermin
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rua Ramiro Barcelos, 2600 - Anexo, 90035-003, RS, Brazil.
- Programa de Pós-Graduação Em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
16
|
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: 1] [Impact Index Per Article: 0.5] [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
|
17
|
Beckers P, Lara O, Belo do Nascimento I, Desmet N, Massie A, Hermans E. Validation of a System xc– Functional Assay in Cultured Astrocytes and Nervous Tissue Samples. Front Cell Neurosci 2022; 15:815771. [PMID: 35095428 PMCID: PMC8793334 DOI: 10.3389/fncel.2021.815771] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
Disruption of the glutamatergic homeostasis is commonly observed in neurological diseases and has been frequently correlated with the altered expression and/or function of astrocytic high-affinity glutamate transporters. There is, however, a growing interest for the role of the cystine-glutamate exchanger system xc– in controlling glutamate transmission. This exchanger is predominantly expressed in glial cells, especially in microglia and astrocytes, and its dysregulation has been documented in diverse neurological conditions. While most studies have focused on measuring the expression of its specific subunit xCT by RT-qPCR or by Western blotting, the activity of this exchanger in tissue samples remains poorly examined. Indeed, the reported use of sulfur- and carbon-radiolabeled cystine in uptake assays shows several drawbacks related to its short radioactive half-life and its relatively high cost. We here report on the elaborate validation of a method using tritiated glutamate as a substrate for the reversed transport mediated by system xc–. The uptake assay was validated in primary cultured astrocytes, in transfected cells as well as in crude synaptosomes obtained from fresh nervous tissue samples. Working in buffers containing defined concentrations of Na+, allowed us to differentiate the glutamate uptake supported by system xc– or by high-affinity glutamate transporters, as confirmed by using selective pharmacological inhibitors. The specificity was further demonstrated in primary astrocyte cultures from transgenic mice lacking xCT or in cell lines where xCT expression was genetically induced or reduced. As such, this assay appears to be a robust and cost-efficient solution to investigate the activity of this exchanger in physiological and pathological conditions. It also provides a reliable tool for the screening and characterization of new system xc– inhibitors which have been frequently cited as valuable drugs for nervous disorders and cancer.
Collapse
Affiliation(s)
- Pauline Beckers
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Olaya Lara
- Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ines Belo do Nascimento
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Nathalie Desmet
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Ann Massie
- Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Emmanuel Hermans
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
- *Correspondence: Emmanuel Hermans,
| |
Collapse
|
18
|
Italiano CJ, Pu L, Violi JP, Duggin IG, Rodgers KJ. Cysteine biosynthesis contributes to β-methylamino-l-alanine tolerance in Escherichia coli. Res Microbiol 2021; 172:103852. [PMID: 34246779 DOI: 10.1016/j.resmic.2021.103852] [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: 03/14/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 11/25/2022]
Abstract
In contrast to mammalian cells, bacteria such as Escherichia coli have been shown to display tolerance towards the neurotoxin β-methylamino-l-alanine (BMAA) suggesting that these prokaryotes possess a way to metabolise BMAA or its products, resulting in their export, degradation, or detoxification. Single gene deletion mutants of E. coli K-12 with inactivated amino acid biosynthesis pathways were treated with 500 μg/ml BMAA and the resulting growth was monitored. Wild type E. coli and most of the gene deletion mutants displayed unaltered growth in the presence of BMAA over 12 h. Conversely, deletion of genes in the cysteine biosynthesis pathway, cysE, cysK or cysM resulted in a BMAA dose-dependent growth delay in minimal medium. Through further studies of the ΔcysE strain, we observed increased susceptibility to oxidative stress from H2O2 in minimal medium, and disruptions in glutathione levels and oxidation state. The cysteine biosynthesis pathway is therefore linked to the tolerance of BMAA and oxidative stress in E. coli, which potentially represents a mechanism of BMAA detoxification.
Collapse
Affiliation(s)
- Carly J Italiano
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| | - Lisa Pu
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| | - Jake P Violi
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| | - Iain G Duggin
- The iThree Institute, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| | - Kenneth J Rodgers
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| |
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: 25] [Impact Index Per Article: 8.3] [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
|
Quinn AW, Phillips CR, Violi JP, Steele JR, Johnson MS, Westerhausen MT, Rodgers KJ. β-Methylamino-L-alanine-induced protein aggregation in vitro and protection by L-serine. Amino Acids 2021; 53:1351-1359. [PMID: 34283312 DOI: 10.1007/s00726-021-03049-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 07/12/2021] [Indexed: 10/20/2022]
Abstract
The cyanobacterial non-protein amino acid α-amino-β-methylaminopropionic acid, more commonly known as BMAA, was first discovered in the seeds of the ancient gymnosperm Cycad circinalis (now Cycas micronesica Hill). BMAA was linked to the high incidence of neurological disorders on the island of Guam first reported in the 1950s. BMAA still attracts interest as a possible causative factor in amyotrophic lateral sclerosis (ALS) following the identification of ALS disease clusters associated with living in proximity to lakes with regular cyanobacterial blooms. Since its discovery, BMAA toxicity has been the subject of many in vivo and in vitro studies. A number of mechanisms of toxicity have been proposed including an agonist effect at glutamate receptors, competition with cysteine for transport system xc_ and other mechanisms capable of generating cellular oxidative stress. In addition, a wide range of studies have reported effects related to disturbances in proteostasis including endoplasmic reticulum stress and activation of the unfolded protein response. In the present studies we examine the effects of BMAA on the ubiquitin-proteasome system (UPS) and on chaperone-mediated autophagy (CMA) by measuring levels of ubiquitinated proteins and lamp2a protein levels in a differentiated neuronal cell line exposed to BMAA. The BMAA induced increases in oxidised proteins and the increase in CMA activity reported could be prevented by co-administration of L-serine but not by the two antioxidants examined. These data provide further evidence of a protective role for L-serine against the deleterious effects of BMAA.
Collapse
Affiliation(s)
- Adam W Quinn
- Neurotoxin Research Group, School of Life Sciences, University of Technology Syd, ney, Faculty of Science, Building 4, Level 7, room 329. Thomas Street, Sydney, NSW, 2007, Australia
| | - Connor R Phillips
- Neurotoxin Research Group, School of Life Sciences, University of Technology Syd, ney, Faculty of Science, Building 4, Level 7, room 329. Thomas Street, Sydney, NSW, 2007, Australia
| | - Jake P Violi
- Neurotoxin Research Group, School of Life Sciences, University of Technology Syd, ney, Faculty of Science, Building 4, Level 7, room 329. Thomas Street, Sydney, NSW, 2007, Australia
| | - Joel R Steele
- Neurotoxin Research Group, School of Life Sciences, University of Technology Syd, ney, Faculty of Science, Building 4, Level 7, room 329. Thomas Street, Sydney, NSW, 2007, Australia
| | - Michael S Johnson
- Neurotoxin Research Group, School of Life Sciences, University of Technology Syd, ney, Faculty of Science, Building 4, Level 7, room 329. Thomas Street, Sydney, NSW, 2007, Australia
| | - Mika T Westerhausen
- Neurotoxin Research Group, School of Life Sciences, University of Technology Syd, ney, Faculty of Science, Building 4, Level 7, room 329. Thomas Street, Sydney, NSW, 2007, Australia
| | - Kenneth J Rodgers
- Neurotoxin Research Group, School of Life Sciences, University of Technology Syd, ney, Faculty of Science, Building 4, Level 7, room 329. Thomas Street, Sydney, NSW, 2007, Australia.
| |
Collapse
|
21
|
Koksharova OA, Butenko IO, Pobeguts OV, Safronova NA, Govorun VM. β-N-Methylamino-L-Alanine (BMAA) Causes Severe Stress in Nostoc sp. PCC 7120 Cells under Diazotrophic Conditions: A Proteomic Study. Toxins (Basel) 2021; 13:325. [PMID: 33946501 PMCID: PMC8147232 DOI: 10.3390/toxins13050325] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 04/07/2021] [Accepted: 04/28/2021] [Indexed: 12/24/2022] Open
Abstract
Non-proteinogenic neurotoxic amino acid β-N-methylamino-L-alanine (BMAA) is synthesized by cyanobacteria, diatoms, and dinoflagellates, and is known to be a causative agent of human neurodegenerative diseases. Different phytoplankton organisms' ability to synthesize BMAA could indicate the importance of this molecule in the interactions between microalgae in nature. We were interested in the following: what kinds of mechanisms underline BMAA's action on cyanobacterial cells in different nitrogen supply conditions. Herein, we present a proteomic analysis of filamentous cyanobacteria Nostoc sp. PCC 7120 cells that underwent BMAA treatment in diazotrophic conditions. In diazotrophic growth conditions, to survive, cyanobacteria can use only biological nitrogen fixation to obtain nitrogen for life. Note that nitrogen fixation is an energy-consuming process. In total, 1567 different proteins of Nostoc sp. PCC 7120 were identified by using LC-MS/MS spectrometry. Among them, 123 proteins belonging to different functional categories were selected-due to their notable expression differences-for further functional analysis and discussion. The presented proteomic data evidences that BMAA treatment leads to very strong (up to 80%) downregulation of α (NifD) and β (NifK) subunits of molybdenum-iron protein, which is known to be a part of nitrogenase. This enzyme is responsible for catalyzing nitrogen fixation. The genes nifD and nifK are under transcriptional control of a global nitrogen regulator NtcA. In this study, we have found that BMAA impacts in a total of 22 proteins that are under the control of NtcA. Moreover, BMAA downregulates 18 proteins that belong to photosystems I or II and light-harvesting complexes; BMAA treatment under diazotrophic conditions also downregulates five subunits of ATP synthase and enzyme NAD(P)H-quinone oxidoreductase. Therefore, we can conclude that the disbalance in energy and metabolite amounts leads to severe intracellular stress that induces the upregulation of stress-activated proteins, such as starvation-inducible DNA-binding protein, four SOS-response enzymes, and DNA repair enzymes, nine stress-response enzymes, and four proteases. The presented data provide new leads into the ecological impact of BMAA on microalgal communities that can be used in future investigations.
Collapse
Affiliation(s)
- Olga A. Koksharova
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Leninskie Gory, 1-40, 119991 Moscow, Russia;
- Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, Kurchatov Square, 2, 123182 Moscow, Russia
| | - Ivan O. Butenko
- Scientific-Research Institute of Physical-Chemical Medicine, 119435 Moscow, Russia; (I.O.B.); (O.V.P.); (V.M.G.)
| | - Olga V. Pobeguts
- Scientific-Research Institute of Physical-Chemical Medicine, 119435 Moscow, Russia; (I.O.B.); (O.V.P.); (V.M.G.)
| | - Nina A. Safronova
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Leninskie Gory, 1-40, 119991 Moscow, Russia;
| | - Vadim M. Govorun
- Scientific-Research Institute of Physical-Chemical Medicine, 119435 Moscow, Russia; (I.O.B.); (O.V.P.); (V.M.G.)
| |
Collapse
|
22
|
Samardzic K, Steele JR, Violi JP, Colville A, Mitrovic SM, Rodgers KJ. Toxicity and bioaccumulation of two non-protein amino acids synthesised by cyanobacteria, β-N-Methylamino-L-alanine (BMAA) and 2,4-diaminobutyric acid (DAB), on a crop plant. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111515. [PMID: 33099142 DOI: 10.1016/j.ecoenv.2020.111515] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 10/02/2020] [Accepted: 10/13/2020] [Indexed: 06/11/2023]
Abstract
In order to study the toxicity of the cyanobacterial non-protein amino acids (NPAAs) L-β-N-methylamino-L-alanine (BMAA) and its structural isomer L-2,4-diaminobutyric acid (DAB) in the forage crop plant alfalfa (Medicago sativa), seedlings were exposed to NPAA-containing media for four days. Root growth was significantly inhibited by both treatments. The content of derivatised free and protein-bound BMAA and DAB in seedlings was then analysed by LC-MS/MS. Both NPAAs were detected in free and protein-bound fractions with higher levels detected in free fractions. Compared to shoots, there was approximately tenfold more BMAA and DAB in alfalfa roots. These results suggest that NPAAs might be taken up into crop plants from contaminated irrigation water and enter the food chain. This may present an exposure pathway for NPAAs in humans.
Collapse
Affiliation(s)
- Kate Samardzic
- School of Life Sciences, University of Technology Sydney, Ultimo, Australia.
| | - Joel R Steele
- School of Life Sciences, University of Technology Sydney, Ultimo, Australia
| | - Jake P Violi
- School of Life Sciences, University of Technology Sydney, Ultimo, Australia
| | - Anne Colville
- School of Life Sciences, University of Technology Sydney, Ultimo, Australia
| | - Simon M Mitrovic
- School of Life Sciences, University of Technology Sydney, Ultimo, Australia
| | - Kenneth J Rodgers
- School of Life Sciences, University of Technology Sydney, Ultimo, Australia
| |
Collapse
|
23
|
Kazama M, Kato Y, Kakita A, Noguchi N, Urano Y, Masui K, Niida-Kawaguchi M, Yamamoto T, Watabe K, Kitagawa K, Shibata N. Astrocytes release glutamate via cystine/glutamate antiporter upregulated in response to increased oxidative stress related to sporadic amyotrophic lateral sclerosis. Neuropathology 2020; 40:587-598. [PMID: 33305472 DOI: 10.1111/neup.12716] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/29/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023]
Abstract
A vast body of evidence implicates increased oxidative stress and extracellular glutamate accumulation in the pathomechanism of sporadic amyotrophic lateral sclerosis (ALS). Cystine/glutamate antiporter (xCT) carries extracellular cystine uptake and intracellular glutamate release (cystine/glutamate exchange) in the presence of oxidative stress. The aim of the present study was to determine the involvement of xCT in ALS. Immunohistochemical observations in the spinal cord sections demonstrated that xCT was mainly expressed in astrocytes, with staining more intense in 12 sporadic ALS patients as compared to 12 age-matched control individuals. Western blot and densitometric analyses of the spinal cord samples revealed that the relative value of xCT/β-actin optical density ratio was significantly higher in the ALS group as compared to the control group. Next, we conducted cell culture experiments using a human astrocytoma-derived cell line (1321N1) and a mouse motor neuron/neuroblastoma hybrid cell line (NSC34). In 1321N1 cells, the normalized xCT expression levels in cell lysates were significantly increased by H2 O2 treatment. Glutamate concentrations in 1321 N1 cell culture-conditioned media were significantly elevated by H2 O2 treatment, and the H2 O2 -driven elevations were completely canceled by the xCT inhibitor erastin pretreatment. In motor neuron-differentiated NSC34 cells (NSC34d cells), both the normalized xCT expression levels in the cell lysates and glutamate concentrations in the cell-conditioned media were constant with or without H2 O2 treatment. The present results provide in vivo and in vitro evidence that astrocytes upregulate xCT expression to release glutamate in response to increased oxidative stress associated with ALS, contributing to extracellular glutamate accumulation.
Collapse
Affiliation(s)
- Miku Kazama
- Faculty of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Yoichiro Kato
- Division of Pathological Neuroscience, Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Noriko Noguchi
- Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Yasuomi Urano
- Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Kenta Masui
- Division of Pathological Neuroscience, Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Motoko Niida-Kawaguchi
- Division of Pathological Neuroscience, Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Tomoko Yamamoto
- Division of Pathological Neuroscience, Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Kazuhiko Watabe
- Department of Medical Technology, Kyorin University, Tokyo, Japan
| | - Kazuo Kitagawa
- Department of Neurology, Tokyo Women's Medical University, Tokyo, Japan
| | - Noriyuki Shibata
- Faculty of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
24
|
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: 0.8] [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
|
25
|
Western Pacific ALS-PDC: Evidence implicating cycad genotoxins. J Neurol Sci 2020; 419:117185. [PMID: 33190068 DOI: 10.1016/j.jns.2020.117185] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 09/20/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022]
Abstract
Amyotrophic Lateral Sclerosis and Parkinsonism-Dementia Complex (ALS-PDC) is a disappearing neurodegenerative disorder of apparent environmental origin formerly hyperendemic among Chamorros of Guam-USA, Japanese residents of the Kii Peninsula, Honshu Island, Japan and Auyu-Jakai linguistic groups of Papua-Indonesia on the island of New Guinea. The most plausible etiology is exposure to genotoxins in seed of neurotoxic cycad plants formerly used for food and/or medicine. Primary suspicion falls on methylazoxymethanol (MAM), the aglycone of cycasin and on the non-protein amino acid β-N-methylamino-L-alanine, both of which are metabolized to formaldehyde. Human and animal studies suggest: (a) exposures occurred early in life and sometimes during late fetal brain development, (b) clinical expression of neurodegenerative disease appeared years or decades later, and (c) pathological changes in various tissues indicate the disease was not confined to the CNS. Experimental evidence points to toxic molecular mechanisms involving DNA damage, epigenetic changes, transcriptional mutagenesis, neuronal cell-cycle reactivation and perturbation of the ubiquitin-proteasome system that led to polyproteinopathy and culminated in neuronal degeneration. Lessons learned from research on ALS-PDC include: (a) familial disease may reflect common toxic exposures across generations, (b) primary disease prevention follows cessation of exposure to culpable environmental triggers; and (c) disease latency provides a prolonged period during which to intervene therapeutically. Exposure to genotoxic chemicals ("slow toxins") in the early stages of life should be considered in the search for the etiology of ALS-PDC-related neurodegenerative disorders, including sporadic forms of ALS, progressive supranuclear palsy and Alzheimer's disease.
Collapse
|
26
|
Chen Q, Konrad C, Sandhu D, Roychoudhury D, Schwartz BI, Cheng RR, Bredvik K, Kawamata H, Calder EL, Studer L, Fischer SM, Manfredi G, Gross SS. Accelerated transsulfuration metabolically defines a discrete subclass of amyotrophic lateral sclerosis patients. Neurobiol Dis 2020; 144:105025. [PMID: 32745521 PMCID: PMC7491150 DOI: 10.1016/j.nbd.2020.105025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/30/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis is a disease characterized by progressive paralysis and death. Most ALS-cases are sporadic (sALS) and patient heterogeneity poses challenges for effective therapies. Applying metabolite profiling on 77-sALS patient-derived-fibroblasts and 43-controls, we found ~25% of sALS cases (termed sALS-1) are characterized by transsulfuration pathway upregulation, where methionine-derived-homocysteine is channeled into cysteine for glutathione synthesis. sALS-1 fibroblasts selectively exhibited a growth defect under oxidative conditions, fully-rescued by N-acetylcysteine (NAC). [U–13C]-glucose tracing showed transsulfuration pathway activation with accelerated glucose flux into the Krebs cycle. We established a four-metabolite support vector machine model predicting sALS-1 metabotype with 97.5% accuracy. Both sALS-1 metabotype and growth phenotype were validated in an independent cohort of sALS cases. Importantly, plasma metabolite profiling identified a system-wide cysteine metabolism perturbation as a hallmark of sALS-1. Findings reveal that sALS patients can be stratified into distinct metabotypes with differential sensitivity to metabolic stress, providing novel insights for personalized therapy.
Collapse
Affiliation(s)
- Qiuying Chen
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Csaba Konrad
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Davinder Sandhu
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | | | | | - Roger R Cheng
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Kirsten Bredvik
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Hibiki Kawamata
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Elizabeth L Calder
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Center, New York, NY, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Center, New York, NY, USA
| | | | - Giovanni Manfredi
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Steven S Gross
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
27
|
Koksharova OA, Butenko IO, Pobeguts OV, Safronova NA, Govorun VM. The First Proteomics Study of Nostoc sp. PCC 7120 Exposed to Cyanotoxin BMAA under Nitrogen Starvation. Toxins (Basel) 2020; 12:E310. [PMID: 32397431 PMCID: PMC7290344 DOI: 10.3390/toxins12050310] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 01/10/2023] Open
Abstract
The oldest prokaryotic photoautotrophic organisms, cyanobacteria, produce many different metabolites. Among them is the water-soluble neurotoxic non-protein amino acid beta-N-methylamino-L-alanine (BMAA), whose biological functions in cyanobacterial metabolism are of fundamental scientific and practical interest. An early BMAA inhibitory effect on nitrogen fixation and heterocyst differentiation was shown in strains of diazotrophic cyanobacteria Nostoc sp. PCC 7120, Nostocpunctiforme PCC 73102 (ATCC 29133), and Nostoc sp. strain 8963 under conditions of nitrogen starvation. Herein, we present a comprehensive proteomic study of Nostoc (also called Anabaena) sp. PCC 7120 in the heterocyst formation stage affecting by BMAA treatment under nitrogen starvation conditions. BMAA disturbs proteins involved in nitrogen and carbon metabolic pathways, which are tightly co-regulated in cyanobacteria cells. The presented evidence shows that exogenous BMAA affects a key nitrogen regulatory protein, PII (GlnB), and some of its protein partners, as well as glutamyl-tRNA synthetase gltX and other proteins that are involved in protein synthesis, heterocyst differentiation, and nitrogen metabolism. By taking into account the important regulatory role of PII, it becomes clear that BMAA has a severe negative impact on the carbon and nitrogen metabolism of starving Nostoc sp. PCC 7120 cells. BMAA disturbs carbon fixation and the carbon dioxide concentrating mechanism, photosynthesis, and amino acid metabolism. Stress response proteins and DNA repair enzymes are upregulated in the presence of BMAA, clearly indicating severe intracellular stress. This is the first proteomic study of the effects of BMAA on diazotrophic starving cyanobacteria cells, allowing a deeper insight into the regulation of the intracellular metabolism of cyanobacteria by this non-protein amino acid.
Collapse
Affiliation(s)
- Olga A. Koksharova
- Lomonosov Moscow State University, Belozersky Institute of Physical-Chemical Biology, Leninskie Gory, 1-40, 119992 Moscow, Russia;
- Institute of Molecular Genetics, Russian Academy of Sciences, Kurchatov Square, 2, 123182 Moscow, Russia
| | - Ivan O. Butenko
- Federal Research and Clinical Centre of Physical-Chemical Medicine, 119435 Moscow, Russia; (I.O.B.); (O.V.P.); (V.M.G.)
| | - Olga V. Pobeguts
- Federal Research and Clinical Centre of Physical-Chemical Medicine, 119435 Moscow, Russia; (I.O.B.); (O.V.P.); (V.M.G.)
| | - Nina A. Safronova
- Lomonosov Moscow State University, Belozersky Institute of Physical-Chemical Biology, Leninskie Gory, 1-40, 119992 Moscow, Russia;
| | - Vadim M. Govorun
- Federal Research and Clinical Centre of Physical-Chemical Medicine, 119435 Moscow, Russia; (I.O.B.); (O.V.P.); (V.M.G.)
| |
Collapse
|
28
|
Vallerga CL, Zhang F, Fowdar J, McRae AF, Qi T, Nabais MF, Zhang Q, Kassam I, Henders AK, Wallace L, Montgomery G, Chuang YH, Horvath S, Ritz B, Halliday G, Hickie I, Kwok JB, Pearson J, Pitcher T, Kennedy M, Bentley SR, Silburn PA, Yang J, Wray NR, Lewis SJG, Anderson T, Dalrymple-Alford J, Mellick GD, Visscher PM, Gratten J. Analysis of DNA methylation associates the cystine-glutamate antiporter SLC7A11 with risk of Parkinson's disease. Nat Commun 2020; 11:1238. [PMID: 32144264 PMCID: PMC7060318 DOI: 10.1038/s41467-020-15065-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 02/17/2020] [Indexed: 11/09/2022] Open
Abstract
An improved understanding of etiological mechanisms in Parkinson's disease (PD) is urgently needed because the number of affected individuals is projected to increase rapidly as populations age. We present results from a blood-based methylome-wide association study of PD involving meta-analysis of 229 K CpG probes in 1,132 cases and 999 controls from two independent cohorts. We identify two previously unreported epigenome-wide significant associations with PD, including cg06690548 on chromosome 4. We demonstrate that cg06690548 hypermethylation in PD is associated with down-regulation of the SLC7A11 gene and show this is consistent with an environmental exposure, as opposed to medications or genetic factors with effects on DNA methylation or gene expression. These findings are notable because SLC7A11 codes for a cysteine-glutamate anti-porter regulating levels of the antioxidant glutathione, and it is a known target of the environmental neurotoxin β-methylamino-L-alanine (BMAA). Our study identifies the SLC7A11 gene as a plausible biological target in PD.
Collapse
Affiliation(s)
- Costanza L Vallerga
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Futao Zhang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Javed Fowdar
- Griffith Institute for Drug Discovery (GRIDD), Griffith University, Brisbane, Australia
| | - Allan F McRae
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Ting Qi
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Marta F Nabais
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.,University of Exeter Medical School, Exeter EX2 5DW, Devon, UK
| | - Qian Zhang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Irfahan Kassam
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Anjali K Henders
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Leanne Wallace
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Grant Montgomery
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Yu-Hsuan Chuang
- Department of Epidemiology, Fielding School of Public Health, UCLA, Los Angeles, CA, USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA.,Department of Biostatistics, Fielding School of Public Health, UCLA, Los Angeles, CA, USA
| | - Beate Ritz
- Department of Epidemiology, Fielding School of Public Health, UCLA, Los Angeles, CA, USA.,Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.,Department of Environmental Health, Fielding School of Public Health, UCLA, Los Angeles, CA, USA
| | - Glenda Halliday
- Brain and Mind Centre & Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Ian Hickie
- Brain and Mind Centre & Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - John B Kwok
- Brain and Mind Centre & Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - John Pearson
- Department of Pathology, University of Otago, Christchurch, New Zealand
| | - Toni Pitcher
- New Zealand Brain Research Institute, Christchurch, New Zealand.,Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Martin Kennedy
- Department of Pathology, University of Otago, Christchurch, New Zealand
| | - Steven R Bentley
- Griffith Institute for Drug Discovery (GRIDD), Griffith University, Brisbane, Australia
| | - Peter A Silburn
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Jian Yang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Naomi R Wray
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Simon J G Lewis
- Brain and Mind Centre & Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Tim Anderson
- New Zealand Brain Research Institute, Christchurch, New Zealand.,Department of Medicine, University of Otago, Christchurch, New Zealand
| | - John Dalrymple-Alford
- New Zealand Brain Research Institute, Christchurch, New Zealand.,Department of Psychology, University of Canterbury, Christchurch, New Zealand
| | - George D Mellick
- Griffith Institute for Drug Discovery (GRIDD), Griffith University, Brisbane, Australia
| | - Peter M Visscher
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia. .,Queensland Brain Institute, The University of Queensland, Brisbane, Australia.
| | - Jacob Gratten
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia. .,Mater Research Institute, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
29
|
Yan B, Liu Z, Huang R, Xu Y, Liu D, Wang W, Zhao Z, Cui F, Shi W. Impact factors on the production of β-methylamino-L-alanine (BMAA) by cyanobacteria. CHEMOSPHERE 2020; 243:125355. [PMID: 31759214 DOI: 10.1016/j.chemosphere.2019.125355] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/17/2019] [Accepted: 11/11/2019] [Indexed: 06/10/2023]
Abstract
Cyanobacteria produce a series of secondary metabolites, one of which is beta-N-methylamino-l-alanine (BMAA). BMAA is considered to be the cause of human neurodegeneration. Compared with other cyanotoxins, the role of BMAA in cyanobacteria remains unclear. To investigate this question, six strains of cyanobacteria were cultured and tested in this experiment with an optimized and validated BMAA determination method. The results show that four strains can produce BMAA. The effects of nutrient levels on the production of BMAA by Anabaena sp. FACHB-418 were studied by changing the initial concentrations of nitrate (NaNO3) and phosphate (K2HPO4) in mediums. Bound BMAA was detected in all samples and the concentrations were within 50-100 ng/g. Free BMAA was presence when the concentration of nitrogen was lower than 1.7 mg/L (121.43 μM). Free BMAA was released from the dead and ruptured cells during the cell decline period, so dissolved BMAA cannot be detectable in the adaptation and logarithmic periods, but could be abundant in the decline periods. Statistical analyses show that free BMAA concentrations were negatively correlated with nitrogen strongly (p = 2.334 × 10-10 and r = -0.842), but positively correlated with phosphorus weakly (p = 0.016 and r = 0.405). Moreover, the results of culture experiments indicated that exogenous BMAA could inhibit the growth of cyanobacteria that cannot produce BMAA, and the effect was enhanced as the concentration of exogenous BMAA increased. This phenomenon implies that the production of BMAA may be the stress response by some cyanobacteria to low nitrogen conditions to kill other cyanobacteria, i.e., their competitors.
Collapse
Affiliation(s)
- Boyin Yan
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150090, PR China
| | - Zhiquan Liu
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150090, PR China; Institute of Environmental Research at Greater Bay, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, 510006, PR China.
| | - Rui Huang
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150090, PR China
| | - Yongpeng Xu
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150090, PR China
| | - Dongmei Liu
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150090, PR China
| | - Wei Wang
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150090, PR China
| | - Zhiwei Zhao
- School of Environment and Ecology, Chongqing University, Chongqing, 400044, PR China
| | - Fuyi Cui
- School of Environment and Ecology, Chongqing University, Chongqing, 400044, PR China
| | - Wenxin Shi
- School of Environment and Ecology, Chongqing University, Chongqing, 400044, PR China.
| |
Collapse
|
30
|
Metabolism of the neurotoxic amino acid β-N-methylamino-L-alanine in human cell culture models. Toxicon 2019; 168:131-139. [PMID: 31330193 DOI: 10.1016/j.toxicon.2019.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 12/12/2022]
Abstract
Human dietary exposure to the environmental neurotoxin β-N-methylamino-L-alanine (BMAA) has been implicated in an increased risk of developing sporadic neurodegenerative diseases like Alzheimer's and amyotrophic lateral sclerosis. Evidence suggests that humans are exposed to BMAA globally, but very little is known about BMAA metabolism in mammalian systems, let alone in humans. The most plausible, evidence-based mechanisms of BMAA toxicity rely on the metabolic stability of the amino acid and that, following ingestion, it enters the circulatory system unmodified. BMAA crosses from the intestinal lumen into the circulatory system, and the small intestine and liver are the first sites for dietary amino acid metabolism. Both tissues have substantial amino acid metabolic needs, which are largely fulfilled by dietary amino acids. Metabolism of BMAA in these tissues has been largely overlooked, yet is important in gauging the true human exposure risk. Here we investigate the potential for BMAA metabolism by the human liver and small intestine, using in vitro cell systems. Data show that BMAA metabolism via common proteinogenic amino acid metabolic pathways is negligible, and that in the presence of other amino acids cellular uptake of BMAA is substantially reduced. These data suggest that the majority of ingested BMAA remains unmodified following passage through the small intestine and liver. This not only supports oral BMAA exposure as a plausible exposure route to toxic doses of BMAA, but also supports previous notions that protein deficient diets or malnutrition may increase an individual's susceptibility to BMAA absorption and subsequent toxicity.
Collapse
|
31
|
Patel D, Kharkar PS, Gandhi NS, Kaur E, Dutt S, Nandave M. Novel analogs of sulfasalazine as system x c - antiporter inhibitors: Insights from the molecular modeling studies. Drug Dev Res 2019; 80:758-777. [PMID: 31199023 DOI: 10.1002/ddr.21557] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 04/16/2019] [Accepted: 05/27/2019] [Indexed: 02/05/2023]
Abstract
System xc - (Sxc - ), a cystine-glutamate antiporter, is established as an interesting target for the treatment of several pathologies including epileptic seizures, glioma, neurodegenerative diseases, and multiple sclerosis. Erastin, sorafenib, and sulfasalazine (SSZ) are a few of the established inhibitors of Sxc - . However, its pharmacological inhibition with novel and potent agents is still very much required due to potential issues, for example, potency, bioavailability, and blood-brain barrier (BBB) permeability, with the current lead molecules such as SSZ. Therefore, in this study, we report the synthesis and structure-activity relationships (SAR) of SSZ derivatives along with molecular docking and dynamics simulations using the developed homology model of xCT chain of Sxc - antiporter. The generated homology model attempted to address the limitations of previously reported comparative protein models, thereby increasing the confidence in the computational modeling studies. The main objective of the present study was to derive a suitable lead structure from SSZ eliminating its potential issues for the treatment of glioblastoma multiforme (GBM), a deadly and malignant grade IV astrocytoma. The designed compounds with favorable Sxc - inhibitory activity following in vitro Sxc - inhibition studies, showed moderately potent cytotoxicity in patient-derived human glioblastoma cells, thereby generating potential interest in these compounds. The xCT-ligand model can be further optimized in search of potent lead molecules for novel drug discovery and development studies.
Collapse
Affiliation(s)
- Dhavalkumar Patel
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS (Deemed to be University), Vile Parle (West), Mumbai, Maharashtra, India
| | - Prashant S Kharkar
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS (Deemed to be University), Vile Parle (West), Mumbai, Maharashtra, India
| | - Neha S Gandhi
- School of Mathematical Sciences and Institute for Health and Biomedical Innovation, Queensland University of Technology, Gardens Point Campus, Brisbane, Queensland, Australia
| | - Ekjot Kaur
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Shilpee Dutt
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Mukesh Nandave
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS (Deemed to be University), Vile Parle (West), Mumbai, Maharashtra, India.,Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| |
Collapse
|
32
|
Schneider TR, Hakami-Tafreshi R, Tomasino-Perez A, Tayebi L, Lobner D. Effects of dental composite resin monomers on dental pulp cells. Dent Mater J 2019; 38:579-583. [PMID: 31105159 DOI: 10.4012/dmj.2018-163] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Methacrylate monomers found in many dental materials cause toxicity to dental pulp cells but the mechanism of the toxicity is poorly understood. We used cultured human dental pulp cells to test the effects of three commonly used monomers; bisphenol-A-glycidyl methacrylate (Bis-GMA), urethane dimethacrylate (UDMA), and triethyleneglycol dimethacrylate (TEGDMA). The order of toxicity was Bis-GMA>UDMA>TEGDMA. The toxicity correlated inversely with cystine uptake, with TEGDMA stimulating uptake and BisGMA and UDMA inhibiting uptake. Bis-GMA and UDMA induced oxidative stress, while TEGDMA did not. Toxicity correlated poorly with glutathione levels, as all compounds decreased cellular glutathione. TEGDMA is less toxic than Bis-GMA and UDMA likely because it stimulates cystine uptake and does not induce oxidative stress, the enhanced uptake of cystine appears to compensate for TEGDMA's direct interaction with glutathione. Bis-GMA and UDMA both deplete glutathione and inhibit cystine uptake leading to oxidative stress and cell death.
Collapse
Affiliation(s)
| | | | | | | | - Doug Lobner
- Department of Biomedical Sciences, Marquette University
| |
Collapse
|
33
|
Manolidi K, Triantis TM, Kaloudis T, Hiskia A. Neurotoxin BMAA and its isomeric amino acids in cyanobacteria and cyanobacteria-based food supplements. JOURNAL OF HAZARDOUS MATERIALS 2019; 365:346-365. [PMID: 30448548 DOI: 10.1016/j.jhazmat.2018.10.084] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 10/26/2018] [Accepted: 10/27/2018] [Indexed: 06/09/2023]
Abstract
Cyanobacteria are photosynthetic microorganisms distributed globally in aquatic and terrestrial environments. They are also industrially cultivated to be used as dietary supplements, as they have a high nutritional value; however, they are also known to produce a wide range of toxic secondary metabolites, called cyanotoxins. BMAA (β-methylamino-l-alanine) and its most common structural isomers, DAB (2,4-diaminobutyric acid) and AEG (N-2-aminoethylglycine) produced by cyanobacteria, are non-proteinogenic amino acids that have been associated with neurodegenerative diseases. A possible route of exposure to those amino acids is through consumption of food supplements based on cyanobacteria. The review critically discusses existing reports regarding the occurrence of BMAA, DAB and AEG in cyanobacteria and cyanobacteria-based food supplements. It is shown that inconsistencies in reported results could be attributed to performance of different methods of extraction and analysis applied and in ambiguities regarding determination of soluble and bound fractions of the compounds. The critical aspect of this review aims to grow awareness of human intake of neurotoxic amino acids, while results presented in literature concerning dietary supplements aim to promote further research, quality control as well as development of guidelines for cyanotoxins in food products.
Collapse
Affiliation(s)
- Korina Manolidi
- Institute of Nanoscience and Nanotechnology, National Center for Scientific Research "DEMOKRITOS", Patriarchou Grigoriou E' & Neapoleos 27, 15341, Athens, Greece; National and Kapodistrian University of Athens, Faculty of Chemistry, 15784, Panepistimiopolis, Athens, Greece.
| | - Theodoros M Triantis
- Institute of Nanoscience and Nanotechnology, National Center for Scientific Research "DEMOKRITOS", Patriarchou Grigoriou E' & Neapoleos 27, 15341, Athens, Greece.
| | - Triantafyllos Kaloudis
- Institute of Nanoscience and Nanotechnology, National Center for Scientific Research "DEMOKRITOS", Patriarchou Grigoriou E' & Neapoleos 27, 15341, Athens, Greece; Water Quality Control Department, Athens Water Supply and Sewerage Company - EYDAP SA, Athens, Greece.
| | - Anastasia Hiskia
- Institute of Nanoscience and Nanotechnology, National Center for Scientific Research "DEMOKRITOS", Patriarchou Grigoriou E' & Neapoleos 27, 15341, Athens, Greece.
| |
Collapse
|
34
|
van Onselen R, Downing TG. β- N-methylamino-L-alanine Inhibits Human Catalase Activity: Possible Implications for Neurodegenerative Disease Development. Int J Toxicol 2019; 38:129-134. [PMID: 30663459 DOI: 10.1177/1091581818821921] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The naturally produced, nonprotein amino acid β- N-methylamino-l-alanine (BMAA) has been proposed as a significant contributor to sporadic neurodegenerative disease development worldwide. However, the existing hypothesized mechanisms of toxicity do not adequately explain the role of BMAA in neurodegenerative disease development. There is evidence for BMAA-induced enzyme inhibition, but the effect of BMAA on human stress response enzymes has received little attention, despite the well-described role of oxidative stress in neurodegenerative disease development. The aim of this study was therefore to investigate the effect of BMAA on human catalase activity and compare it to the known inhibitor 3-amino-1,2,4-triazole. BMAA inhibited human erythrocyte catalase in a cell-free exposure to the same extent as the known inhibitor. Based on enzyme kinetics, the inhibition appears to be noncompetitive, possibly as a result of BMAA binding in the nicotinamide adenine dinucleotide phosphate (NADPH) binding site. BMAA-induced catalase inhibition was also observed in a human cell line culture. We therefore propose that BMAA-induced enzyme inhibition, specifically catalase inhibition, is a mechanism of toxicity that may contribute to the neurotoxicity of BMAA, further supporting the role of BMAA in neurodegenerative disease development.
Collapse
Affiliation(s)
- Rianita van Onselen
- 1 Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth, South Africa
| | - Tim G Downing
- 1 Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth, South Africa
| |
Collapse
|
35
|
Gerić M, Gajski G, Domijan AM, Garaj-Vrhovac V, Filipič M, Žegura B. Genotoxic effects of neurotoxin ß-N-methylamino-l-alanine in human peripheral blood cells. CHEMOSPHERE 2019; 214:623-632. [PMID: 30290362 DOI: 10.1016/j.chemosphere.2018.09.155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 09/21/2018] [Accepted: 09/26/2018] [Indexed: 06/08/2023]
Abstract
The non-proteinogenic amino acid ß-N-methylamino-l-alanine (BMAA) is associated with the development of neurodegenerative diseases such as Alzheimer's disease, amyotrophic lateral sclerosis/parkinsonism-dementia complex (ALS-PDC) and amyotrophic lateral sclerosis. BMAA is known to induce neurotoxic effects leading to neurodegeneration via multiple mechanisms including misfolded protein accumulation, glutamate induced excitotoxicity, calcium dyshomeostasis, endoplasmic reticulum stress and oxidative stress. In the present study, for the first time, genotoxic activity of BMAA (2.5, 5, 10 and 20 μg/mL) was studied in human peripheral blood cells (HPBCs) using the comet and cytokinesis-block micronucleus cytome assays. In addition, the influence of BMAA on the oxidative stress was assessed. At non-cytotoxic concentrations BMAA did not induce formation of DNA strand breaks in HPBCs after 4 and 24 h exposure; however, it significantly increased the number of micronuclei after 24 and 48 h at 20 μg/mL and nucleoplasmic bridges after 48 h at 20 μg/mL. The frequency of nuclear buds was slightly though non-significantly increased after 48 h. Altogether, this indicates that in HPBCs BMAA is clastogenic and induces complex genomic alterations including structural chromosomal rearrangements and gene amplification. No influence on oxidative stress markers was noticed. These findings provide new evidence that environmental neurotoxin BMAA, in addition to targeting common pathways involved in neurodegeneration, can also induce genomic instability in non-target HPBCs suggesting that it might be involved in cancer development. Therefore, these data are important in advancing our current knowledge and opening new questions in the understanding of the mechanisms of BMAA toxicity, particularly in the context of genotoxicity.
Collapse
Affiliation(s)
- Marko Gerić
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia.
| | - Goran Gajski
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia.
| | - Ana-Marija Domijan
- Department of Pharmaceutical Botany, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia.
| | - Vera Garaj-Vrhovac
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia.
| | - Metka Filipič
- Department for Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia.
| | - Bojana Žegura
- Department for Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia.
| |
Collapse
|
36
|
β-N-methylamino-L-alanine (BMAA) suppresses cell cycle progression of non-neuronal cells. Sci Rep 2018; 8:17995. [PMID: 30573743 PMCID: PMC6301973 DOI: 10.1038/s41598-018-36418-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 11/21/2018] [Indexed: 12/25/2022] Open
Abstract
β-N-methylamino-L-alanine (BMAA), a natural non-proteinaceous amino acid, is a neurotoxin produced by a wide range of cyanobacteria living in various environments. BMAA is a candidate environmental risk factor for neurodegenerative diseases such as amyotrophic lateral sclerosis and Parkinson-dementia complex. Although BMAA is known to exhibit weak neuronal excitotoxicity via glutamate receptors, the underlying mechanism of toxicity has yet to be fully elucidated. To examine the glutamate receptor-independent toxicity of BMAA, we investigated the effects of BMAA in non-neuronal cell lines. BMAA potently suppressed the cell cycle progression of NIH3T3 cells at the G1/S checkpoint without inducing plasma membrane damage, apoptosis, or overproduction of reactive oxygen species, which were previously reported for neurons and neuroblastoma cells treated with BMAA. We found no evidence that activation of glutamate receptors was involved in the suppression of the G1/S transition by BMAA. Our results indicate that BMAA affects cellular functions, such as the division of non-neuronal cells, through glutamate receptor-independent mechanisms.
Collapse
|
37
|
Cyanobacterial Neurotoxin Beta-Methyl-Amino-l-Alanine Affects Dopaminergic Neurons in Optic Ganglia and Brain of Daphnia magna. Toxins (Basel) 2018; 10:toxins10120527. [PMID: 30544796 PMCID: PMC6315693 DOI: 10.3390/toxins10120527] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 12/02/2018] [Accepted: 12/06/2018] [Indexed: 12/31/2022] Open
Abstract
The non-proteinogenic amino acid beta-methyl-amino-l-alanine (BMAA) is a neurotoxin produced by cyanobacteria. BMAA accumulation in the brain of animals via biomagnification along the food web can contribute to the development of neurodegenerative diseases such as Amyotrophic lateral sclerosis/Parkinsonism dementia complex (ALS/PDC), the latter being associated with a loss of dopaminergic neurons. Daphnia magna is an important microcrustacean zooplankton species that plays a key role in aquatic food webs, and BMAA-producing cyanobacteria often form part of their diet. Here, we tested the effects of BMAA on putative neurodegeneration of newly identified specific dopaminergic neurons in the optic ganglia/brain complex of D. magna using quantitative tyrosine-hydroxylase immunohistochemistry and fluorescence cytometry. The dopaminergic system was analysed in fed and starved isogenic D. magna adults incubated under different BMAA concentrations over 4 days. Increased BMAA concentration showed significant decrease in the stainability of dopaminergic neurons of D. magna, with fed animals showing a more extreme loss. Furthermore, higher BMAA concentrations tended to increase offspring mortality during incubation. These results are indicative of ingested BMAA causing neurodegeneration of dopaminergic neurons in D. magna and adversely affecting reproduction. This may imply similar effects of BMAA on known human neurodegenerative diseases involving dopaminergic neurons.
Collapse
|
38
|
Ubiquity of the neurotoxin β-N-methylamino-L-alanine and its isomers confirmed by two different mass spectrometric methods in diverse marine mollusks. Toxicon 2018; 151:129-136. [DOI: 10.1016/j.toxicon.2018.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/24/2018] [Accepted: 07/05/2018] [Indexed: 12/11/2022]
|
39
|
van Onselen R, Downing T. BMAA-protein interactions: A possible new mechanism of toxicity. Toxicon 2018; 143:74-80. [DOI: 10.1016/j.toxicon.2018.01.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 01/16/2018] [Accepted: 01/18/2018] [Indexed: 02/08/2023]
|
40
|
Cellular and Molecular Aspects of the β-N-Methylamino-l-alanine (BMAA) Mode of Action within the Neurodegenerative Pathway: Facts and Controversy. Toxins (Basel) 2017; 10:toxins10010006. [PMID: 29271898 PMCID: PMC5793093 DOI: 10.3390/toxins10010006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/19/2017] [Accepted: 12/20/2017] [Indexed: 12/12/2022] Open
Abstract
The implication of the cyanotoxin β-N-methylamino-l-alanine (BMAA) in long-lasting neurodegenerative disorders is still a matter of controversy. It has been alleged that chronic ingestion of BMAA through the food chain could be a causative agent of amyotrophic lateral sclerosis (ALS) and several related pathologies including Parkinson syndrome. Both in vitro and in vivo studies of the BMAA mode of action have focused on different molecular targets, demonstrating its toxicity to neuronal cells, especially motoneurons, and linking it to human neurodegenerative diseases. Historically, the hypothesis of BMAA-induced excitotoxicity following the stimulation of glutamate receptors has been established. However, in this paradigm, most studies have shown acute, rather than chronic effects of BMAA. More recently, the interaction of this toxin with neuromelanin, a pigment present in the nervous system, has opened a new research perspective. The issues raised by this toxin are related to its kinetics of action, and its possible incorporation into cellular proteins. It appears that BMAA neurotoxic activity involves different targets through several mechanisms known to favour the development of neurodegenerative processes.
Collapse
|
41
|
Roy U, Conklin L, Schiller J, Matysik J, Berry JP, Alia A. Metabolic profiling of zebrafish (Danio rerio) embryos by NMR spectroscopy reveals multifaceted toxicity of β-methylamino-L-alanine (BMAA). Sci Rep 2017; 7:17305. [PMID: 29230019 PMCID: PMC5725574 DOI: 10.1038/s41598-017-17409-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/21/2017] [Indexed: 11/09/2022] Open
Abstract
β-methylamino-L-alanine (BMAA) has been linked to several interrelated neurodegenerative diseases. Despite considerable research, specific contributions of BMAA toxicity to neurodegenerative diseases remain to be fully resolved. In the present study, we utilized state-of-the-art high-resolution magic-angle spinning nuclear magnetic resonance (HRMAS NMR), applied to intact zebrafish (Danio rerio) embryos, as a model of vertebrate development, to elucidate changes in metabolic profiles associated with BMAA exposure. Complemented by several alternative analytical approaches (i.e., in vivo visualization and in vitro assay), HRMAS NMR identified robust and dose-dependent effect of BMAA on several relevant metabolic pathways suggesting a multifaceted toxicity of BMAA including: (1) localized production of reactive oxygen species (ROS), in the developing brain, consistent with excitotoxicity; (2) decreased protective capacity against excitotoxicity and oxidative stress including reduced taurine and glutathione; (3) inhibition of several developmentally stereotypical energetic and metabolic transitions, i.e., metabolic reprogramming; and (4) inhibition of lipid biosynthetic pathways. Matrix-assisted laser desorption time-of-flight (MALDI-ToF) mass spectrometry further identified specific effects on phospholipids linked to both neural development and neurodegeneration. Taken together, a unified model of the neurodevelopmental toxicity of BMAA in the zebrafish embryo is presented in relation to the potential contribution of BMAA to neurodegenerative disease.
Collapse
Affiliation(s)
- Upasana Roy
- Institute for Medical Physics and Biophysics, University of Leipzig, D-04107, Leipzig, Germany.,Institute of Analytical Chemistry, University of Leipzig, D-04103, Leipzig, Germany
| | - Laura Conklin
- Department of Chemistry and Biochemistry, Florida International University, North Miami, FL, 33181, USA
| | - Jürgen Schiller
- Institute for Medical Physics and Biophysics, University of Leipzig, D-04107, Leipzig, Germany
| | - Jörg Matysik
- Institute of Analytical Chemistry, University of Leipzig, D-04103, Leipzig, Germany
| | - John P Berry
- Department of Chemistry and Biochemistry, Florida International University, North Miami, FL, 33181, USA.
| | - A Alia
- Institute for Medical Physics and Biophysics, University of Leipzig, D-04107, Leipzig, Germany. .,Leiden Institute of Chemistry, 2333, Leiden, The Netherlands.
| |
Collapse
|
42
|
Dunlop RA, Powell J, Guillemin GJ, Cox PA. Mechanisms of L-Serine Neuroprotection in vitro Include ER Proteostasis Regulation. Neurotox Res 2017; 33:123-132. [PMID: 29098664 DOI: 10.1007/s12640-017-9829-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 09/27/2017] [Accepted: 10/06/2017] [Indexed: 01/14/2023]
Abstract
β-N-methylamino-L-alanine (L-BMAA) is a neurotoxic non-protein amino acid produced by cyanobacteria. Recently, chronic dietary exposure to L-BMAA was shown to trigger neuropathology in nonhuman primates consistent with Guamanian ALS/PDC, a paralytic disease that afflicts Chamorro villagers who consume traditional food items contaminated with L-BMAA. However, the addition of the naturally occurring amino acid L-serine to the diet of the nonhuman primates resulted in a significant reduction in ALS/PDC neuropathology. L-serine is a dietary amino acid that plays a crucial role in central nervous system development, neuronal signaling, and synaptic plasticity and has been shown to impart neuroprotection from L-BMAA-induced neurotoxicity both in vitro and in vivo. We have previously shown that L-serine prevents the formation of autofluorescent aggregates and death by apoptosis in human cell lines and primary cells. These effects are likely imparted by L-serine blocking incorporation of L-BMAA into proteins hence preventing proteotoxic stress. However, there are likely other mechanisms for L-serine-mediated neuroprotection. Here, we explore the molecular mechanisms of L-serine neuroprotection using a human unfolded protein response real-time PCR array with genes from the ER stress and UPR pathways, and western blotting. We report that L-serine caused the differential expression of many of the same genes as L-BMAA, even though concentrations of L-serine in the culture medium were ten times lower than that of L-BMAA. We propose that L-serine may be functioning as a small proteostasis regulator, in effect altering the cells to quickly respond to a possible oxidative insult, thus favoring a return to homeostasis.
Collapse
Affiliation(s)
- R A Dunlop
- Brain Chemistry Labs, The Institute for Ethnomedicine, Jackson, WY, USA.,Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - J Powell
- Brain Chemistry Labs, The Institute for Ethnomedicine, Jackson, WY, USA
| | - G J Guillemin
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - P A Cox
- Brain Chemistry Labs, The Institute for Ethnomedicine, Jackson, WY, USA.
| |
Collapse
|
43
|
Laugeray A, Oummadi A, Jourdain C, Feat J, Meyer-Dilhet G, Menuet A, Plé K, Gay M, Routier S, Mortaud S, Guillemin GJ. Perinatal Exposure to the Cyanotoxin β-N-Méthylamino-L-Alanine (BMAA) Results in Long-Lasting Behavioral Changes in Offspring-Potential Involvement of DNA Damage and Oxidative Stress. Neurotox Res 2017; 33:87-112. [PMID: 28879461 DOI: 10.1007/s12640-017-9802-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/25/2017] [Accepted: 08/18/2017] [Indexed: 12/13/2022]
Abstract
We recently demonstrated that perinatal exposure to the glutamate-related herbicide, glufosinate ammonium, has deleterious effects on neural stem cell (NSC) homeostasis within the sub-ventricular zone (SVZ), probably leading to ASD-like symptoms in offspring later in life. In the present study, we aimed to investigate whether perinatal exposure to another glutamate-related toxicant, the cyanobacterial amino acid β-N-methylamino-L-alanine (BMAA), might also trigger neurodevelopmental disturbances. With this aim, female mice were intranasally exposed to low doses of BMAA, 50 mg kg-1 three times a week from embryonic days 7-10 to postnatal day 21. Behavioral analyses were performed during the offspring's early life and during adulthood. Developmental analyses revealed that perinatal exposure to BMAA hastened the appearance of some reflexes and communicative skills. BMAA-exposed offspring displayed sex-dependent changes in emotional cognition shortly after exposure. Later in life, the female offspring continued to express emotional defects and to display abnormal sociability, while males were less affected. To assess whether early exposure to BMAA had deleterious effects on NSC homeostasis, we exposed mice NSCs to 1 and 3 mM BMAA during 24 h. We found that BMAA-exposed NSCs produced high levels of ROS, highlighting the ability of BMAA to induce oxidative stress. We also showed that BMAA exposure increased the number of γH2AX/53BP1 foci per nucleus, suggesting that BMAA-induced DNA damage in NSCs. Collectively, this data strongly suggests that perinatal exposure to the cyanobacteria BMAA, even at low doses, results in neurobehavioral disturbances during both the postnatal period and adulthood. This is considered to be underpinned at the cellular level through dysregulation of NSC homeostasis in the developing brain.
Collapse
Affiliation(s)
- Anthony Laugeray
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355, Centre National de la Recherche Scientifique, 3b, rue de la Férollerie, 45071, Orléans, France. .,University of Orléans, Orléans, France.
| | | | | | - Justyne Feat
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355, Centre National de la Recherche Scientifique, 3b, rue de la Férollerie, 45071, Orléans, France
| | - Géraldine Meyer-Dilhet
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355, Centre National de la Recherche Scientifique, 3b, rue de la Férollerie, 45071, Orléans, France
| | - Arnaud Menuet
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355, Centre National de la Recherche Scientifique, 3b, rue de la Férollerie, 45071, Orléans, France.,University of Orléans, Orléans, France
| | - Karen Plé
- University of Orléans, Orléans, France.,Institute de Chimie Organique et Analytique, UMR 7311, Center National de la Recherche Scientifique, Orléans, France
| | - Marion Gay
- University of Orléans, Orléans, France.,Institute de Chimie Organique et Analytique, UMR 7311, Center National de la Recherche Scientifique, Orléans, France
| | - Sylvain Routier
- University of Orléans, Orléans, France.,Institute de Chimie Organique et Analytique, UMR 7311, Center National de la Recherche Scientifique, Orléans, France
| | - Stéphane Mortaud
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355, Centre National de la Recherche Scientifique, 3b, rue de la Férollerie, 45071, Orléans, France. .,University of Orléans, Orléans, France.
| | - Gilles J Guillemin
- Neuroinflammation Group, MND and Neurodegenerative Diseases Research Center, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
44
|
Tan VX, Mazzocco C, Varney B, Bodet D, Guillemin TA, Bessede A, Guillemin GJ. Detection of the Cyanotoxins L-BMAA Uptake and Accumulation in Primary Neurons and Astrocytes. Neurotox Res 2017; 33:55-61. [PMID: 28852990 DOI: 10.1007/s12640-017-9787-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 07/14/2017] [Accepted: 07/21/2017] [Indexed: 12/12/2022]
Abstract
We show for the first time that a newly developed polyclonal antibody (pAb) can specifically target the cyanotoxin β-methylamino-L-alanine (BMAA) and can be used to enable direct visualization of BMAA entry and accumulation in primary brain cells. We used this pAb to investigate the effect of acute and chronic accumulation, and toxicity of both BMAA and its natural isomer 2,4-diaminobutyric acid (DAB), separately or in combination, on primary cultures of rat neurons. We further present evidence that co-treatment with BMAA and DAB increased neuronal death, as measured by MAP2 fluorescence level, and appeared to reduce BMAA accumulation. DAB is likely to be acting synergistically with BMAA resulting in higher level of cellular toxicity. We also found that glial cells such as microglia and astrocytes are also able to directly uptake BMAA indicating that additional brain cell types are affected by BMAA-induced toxicity. Therefore, BMAA clearly acts at multiple cellular levels to possibly increase the risk of developing neurodegenerative diseases, including neuro- and gliotoxicity and synergetic exacerbation with other cyanotoxins.
Collapse
Affiliation(s)
- Vanessa X Tan
- Macquarie University Centre for MND Research, Department of Biological Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | | | - Bianca Varney
- Macquarie University Centre for MND Research, Department of Biological Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | | | - Tristan A Guillemin
- Macquarie University Centre for MND Research, Department of Biological Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | | | - Gilles J Guillemin
- Macquarie University Centre for MND Research, Department of Biological Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia. .,Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
45
|
Mello FD, Braidy N, Marçal H, Guillemin G, Nabavi SM, Neilan BA. Mechanisms and Effects Posed by Neurotoxic Products of Cyanobacteria/Microbial Eukaryotes/Dinoflagellates in Algae Blooms: a Review. Neurotox Res 2017; 33:153-167. [PMID: 28836116 DOI: 10.1007/s12640-017-9780-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 06/28/2017] [Accepted: 07/05/2017] [Indexed: 12/12/2022]
Abstract
Environmental toxins produced by cyanobacteria and dinoflagellates have increasingly become a public health concern due to their ability to damage several tissues in humans. In particular, emerging evidence has called attention to the neurodegenerative effects of the cyanobacterial toxin β-N-methylamino-L-alanine (BMAA). Furthermore, other toxins such as anatoxin, saxitoxin, microcystin, nodularin and ciguatoxin also have a different range of effects on human tissues, including hepatotoxicity, neurotoxicity and gastrointestinal irritation. However, the vast majority of known environmental toxins have not yet been examined in the context of neurodegenerative disease. This review aims to investigate whether neurotoxic mechanisms can be demonstrated in all aforementioned toxins, and whether there exists a link to neurodegeneration. Management of toxin exposure and potential neuroprotective compounds is also discussed. Collectively, all aforementioned microbial toxins are likely to exert some form of neuronal damage, with many of their modes of action consistent with neurodegeneration. This is important in advancing our current understanding of the cytotoxic potential of environmental toxins upon human brain function, particularly in the context of age-related neurodegenerative disease.
Collapse
Affiliation(s)
- Fiona D Mello
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, The University of New South Wales, Sydney, Australia
| | - Helder Marçal
- Graduate School of Biomedical Engineering, Faculty of Engineering, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Gilles Guillemin
- Neuropharmacology group, MND and Neurodegenerative diseases Research Centre, Macquarie University, Sydney, NSW, Australia
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Brett A Neilan
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
46
|
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: 2.6] [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
|
47
|
Caller T, Henegan P, Stommel E. The Potential Role of BMAA in Neurodegeneration. Neurotox Res 2017; 33:222-226. [PMID: 28612294 DOI: 10.1007/s12640-017-9752-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/09/2017] [Accepted: 05/12/2017] [Indexed: 01/09/2023]
Abstract
Neurodegenerative diseases are a major public health issue throughout the world with devastating effects on patients and families. Sporadic forms of neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis are generally thought to develop as a consequence of genetic susceptibility and environmental influences. A number of environmental triggers have been identified in association with amyotrophic lateral sclerosis and Parkinson's disease. We discuss the role of β-methylamino-L-alanine in the development of neurodegeneration and the potential importance of this neurotoxin as a risk for neurodegeneration.
Collapse
Affiliation(s)
- Tracie Caller
- Department of Neurology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA. .,Cheyenne Regional Medical Group, Cheyenne, WY, 82001, USA. .,Institute for Ethnomedicine, PO Box 3464, Jackson, WY, 83001, USA.
| | - Patricia Henegan
- Department of Neurology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | - Elijah Stommel
- Department of Neurology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| |
Collapse
|
48
|
Banack SA, Cox PA. Creating a Simian Model of Guam ALS/PDC Which Reflects Chamorro Lifetime BMAA Exposures. Neurotox Res 2017; 33:24-32. [PMID: 28478528 DOI: 10.1007/s12640-017-9745-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/03/2017] [Accepted: 04/21/2017] [Indexed: 12/26/2022]
Abstract
The theory that β-N-methylamino-L-alanine (BMAA), a cyanobacterial toxin, contaminates traditional food supplies of the Chamorro people of Guam is supported by the recent finding that chronic dietary exposure to L-BMAA in vervets (Chlorocebus sabaeus) triggers the formation of neurofibrillary tangles (NFT) and β-amyloid plaques in the brain. In the first experiment, we found that all four vervets receiving a 210 mg/kg dose for 140 days developed NFT and sparse amyloid deposits. In the second experiment, all eight vervets receiving a 210 mg/kg dose for 140 days developed NFT and amyloid deposits, as well as all eight vervets that received only 21 mg/kg. Based on dietary surveys of the Chamorro people, we estimated lifetime chronic BMAA exposure at a high and a low level: 1) adult male Chamorros eating two flying foxes per month plus one 30 g serving of cycad flour per week; and 2) adult male Chamorros eating one 30 g serving of cycad flour per day combined with the consumption of eight flying foxes per month. The resultant cumulative lifetime Chamorro exposures ranged from 1 to 41 g/kg and are comparable to the total lifetime vervet exposures in our experiments of 2 and 22 g/kg, respectively. Furthermore, measured protein-bound BMAA concentrations of vervets fed L-BMAA powder are comparable to measured protein-bound BMAA concentrations in postmortem brain tissues of Chamorros who died with ALS/PDC.
Collapse
Affiliation(s)
- Sandra Anne Banack
- Brain Chemistry Labs, Institute for Ethnomedicine, Box 3464, Jackson, WY, 83001, USA.
| | - Paul Alan Cox
- Brain Chemistry Labs, Institute for Ethnomedicine, Box 3464, Jackson, WY, 83001, USA
| |
Collapse
|
49
|
Albano R, Lobner D. Transport of BMAA into Neurons and Astrocytes by System x c. Neurotox Res 2017; 33:1-5. [PMID: 28470569 DOI: 10.1007/s12640-017-9739-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/30/2017] [Accepted: 04/21/2017] [Indexed: 01/13/2023]
Abstract
The study of the mechanism of β-N-methylamino-L-alanine (BMAA) neurotoxicity originally focused on its effects at the N-methyl-D-aspartate (NMDA) receptor. In recent years, it has become clear that its mechanism of action is more complicated. First, there are certain cell types, such as motor neurons and cholinergic neurons, where the dominate mechanism of toxicity is through action at AMPA receptors. Second, even in cortical neurons where the primary mechanism of toxicity appears to be activation of NMDA receptors, there are other mechanisms involved. We found that along with NMDA receptors, activation of mGLuR5 receptors and effects on the cystine/glutamate antiporter (system xc-) were involved in the toxicity. The effects on system xc- are of particular interest. System xc- mediates the transport of cystine into the cell in exchange for releasing glutamate into the extracellular fluid. By releasing glutamate, system xc- can potentially cause excitotoxicity. However, through providing cystine to the cell, it regulates the levels of cellular glutathione (GSH), the main endogenous intracellular antioxidant, and in this way may protect cells against oxidative stress. We have previously published that BMAA inhibits cystine uptake leading to GSH depletion and had indirect evidence that BMAA is transported into the cells by system xc-. We now present direct evidence that BMAA is transported into both astrocytes and neurons through system xc-. The fact that BMAA is transported by system xc- also provides a mechanism for BMAA to enter brain cells potentially leading to misincorporation into proteins and protein misfolding.
Collapse
Affiliation(s)
- Rebecca Albano
- Department of Biomedical Sciences, Marquette University, 561 N. 15th Street, Rm 426, Milwaukee, WI, 53233, USA
| | - Doug Lobner
- Department of Biomedical Sciences, Marquette University, 561 N. 15th Street, Rm 426, Milwaukee, WI, 53233, USA.
| |
Collapse
|
50
|
β-N-Methylamino-L-Alanine Toxicity in PC12: Excitotoxicity vs. Misincorporation. Neurotox Res 2017; 33:15-23. [DOI: 10.1007/s12640-017-9743-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/12/2017] [Accepted: 04/21/2017] [Indexed: 10/19/2022]
|