The non-protein amino acid BMAA is misincorporated into human proteins in place of L-serine causing protein misfolding and aggregation

Rachael Anne Dunlop, Paul Alan Cox, Sandra Anne Banack, Kenneth John Rodgers, Rachael Anne Dunlop, Paul Alan Cox, Sandra Anne Banack, Kenneth John Rodgers

Abstract

Mechanisms of protein misfolding are of increasing interest in the aetiology of neurodegenerative diseases characterized by protein aggregation and tangles including Amyotrophic Lateral Sclerosis (ALS), Alzheimer's disease (AD), Parkinson's disease (PD), Lewy Body Dementia (LBD), and Progressive Supranuclear Palsy (PSP). Some forms of neurodegenerative illness are associated with mutations in genes which control assembly of disease related proteins. For example, the mouse sticky mutation sti, which results in undetected mischarging of tRNA(Ala) with serine resulting in the substitution of serine for alanine in proteins causes cerebellar Purkinje cell loss and ataxia in laboratory animals. Replacement of serine 422 with glutamic acid in tau increases the propensity of tau aggregation associated with neurodegeneration. However, the possibility that environmental factors can trigger abnormal folding in proteins remains relatively unexplored. We here report that a non-protein amino acid, β-N-methylamino-L-alanine (BMAA), can be misincorporated in place of L-serine into human proteins. We also report that this misincorporation can be inhibited by L-serine. Misincorporation of BMAA into human neuroproteins may shed light on putative associations between human exposure to BMAA produced by cyanobacteria and an increased incidence of ALS.

Conflict of interest statement

Competing Interests: A patent application relating to the potential of L-serine to be used as a therapeutic agent for the treatment of neurodegenerative diseases has been submitted (PCT/US2012/066373). This patent does not alter the authors’ adherence to all the PLOS ONE policies on sharing data and materials and there are no consultancies or additional products in development associated with it.

Figures

Figure 1. Uptake and incorporation of 3…
Figure 1. Uptake and incorporation of 3H-BMAA into proteins by MRC-5 cells.
Panel A, uptake of radiolabel by cells was expressed as disintegrations per minute (DPM) per µg of cell protein. Panel B, radiolabel in the cell protein fraction was expressed as DPM per µg of total cell protein. Values are mean +/− SD for three independent experiments (n = 3).
Figure 2. Inhibition of incorporation of radiolabel…
Figure 2. Inhibition of incorporation of radiolabel into cell protein by cycloheximide.
MRC-5, HUVEC and SH-SY5Y cells were incubated with 3H-BMAA with or without CHX (2 µg/ml). Cells proteins were isolated and the amount of radiolabel present in proteins (DPM per cell protein) determined and expressed as a percentage of control where control treatments were designated 100%. Parallel cultures of MRC-5 cells were incubated with 3H-leucine (41 nM) with or without CHX and the effects of CHX similarly determined (open bars). Values are mean +/− SD for three independent experiments (n = 3). Radiolabel incorporation in each cell type was compared with and without CHX using Student’s two-tailed t-test (***P<0.001 ****P<0.0001).
Figure 3. Recovery of radiolabel from cell…
Figure 3. Recovery of radiolabel from cell proteins following incubation of cells with 3H-BMAA.
SH-SY5Y cells were incubated with 3H-BMAA, cell proteins precipitated by TCA (10%) precipitation and the amount of radiolabel released from proteins (i.e. not TCA precipitable) after incubation with DTT or DTT and SDS quantified. Percentage of radiolabel remaining in proteins following treatment is expressed as a % of buffer alone. Cell proteins were also incubated with pronase or HCl and the release of radiolabel quantified relative to that of buffer alone (for Pronase) or water (for Acid). Values are mean +/− SD, P<0.0001 using Student’s two-tailed t-test, three independent tests (n = 3).
Figure 4. Inhibition of incorporation or radiolabel…
Figure 4. Inhibition of incorporation or radiolabel into cell proteins by l-serine.
Panel A, Incorporation of radiolabeled BMAA was inhibited by l-serine in a concentration dependent manner. Panel B, d-serine (D-SER) had no significant impact on the incorporation of BMAA (NS, P = 0.4419). l-serine significantly inhibited the incorporation of BMAA compared to control cells (CTRL, ***P = 0.0002) and d-serine (D-SER, **P<0.01). Panel C, there was a significantly (****P<0.001) greater incorporation of BMAA when all protein amino acids were omitted from the culture medium (ALL) compared to when none were (NONE). When only l-serine was omitted (l-SERINE) incorporation was restored to approximately 80% (****P = 0.0009). Student’s two-tailed T-test, values are mean +/− SD for three independent experiments (n = 3).
Figure 5. Identification of BMAA in cell…
Figure 5. Identification of BMAA in cell proteins using tandem mass spectrometry.
Panel A, a BMAA standard and a hydrolyzed protein sample (from MRC-5 cells treated with 250 µM BMAA for 24 hours) were run on triple quadrupole LC/MS/MS. Retention times, unique daughter ion (m/z 258), and ratios of m/z transitions during collision-induced dissociation of the m/z 459 parent ion matched those of an authenticated BMAA standard. Panel B, linear (r2 = 0.99012) recovery of BMAA from the hydrolyzed proteins as determined by triple quad LC/MS/MS. Values are mean +/− SD from three independent incubations (n = 3).
Figure 6. Incubation with BMAA results in…
Figure 6. Incubation with BMAA results in the formation of autofluorescent bodies and apoptotic changes in cells.
Panel A, Autofluorescence was observed in the cytosol and perinuclear regions of MRC-5 cells incubated with 300 µM BMAA. Panel B, autofluorescence was reduced when cells were co-incubated with 300 µM l-serine. Cells were incubated in 6 well plates and showed consistent perinuclear and cytosolic autofluorescence of varying degrees of intensity in BMAA treated cultures. The overall intensity of the signal was reduced when cells were co-incubated with l-serine. Images are representative of at least 12 fields of view from triplicate cultures. Scale bar is 100 um. Panel C, incubation with BMAA induced significant (**P = 0.0044) apoptosis in SH-SY5Y cells compared to control cells, which could be abrogated when co-incubated with l-serine (***P = 0.0005) or CHX (**P = 0.002). Student’s two-tailed T-test, values are mean + SD from three independent incubations (n = 3).

References

    1. Zaher HS, Green R (2009) Fidelity at the molecular level: lessons from protein synthesis. Cell 136: 746–762.
    1. Lee JW, Beebe K, Nangle LA, Jang J, Longo-Guess CM, et al. (2006) Editing-defective tRNA synthetase causes protein misfolding and neurodegeneration. Nature 443: 50–55.
    1. Haase C, Stieler JT, Arendt T, Holzer M (2004) Pseudophosphorylation of tau protein alters its ability for self-aggregation. Journal of Neurochemistry 88: 1509–1520.
    1. Bell EA (2003) Nonprotein amino acids of plants: significance in medicine, nutrition, and agriculture. Journal of Agricultural and Food Chemistry 51: 2854–2865.
    1. Bell EA (2009) The discovery of BMAA, and examples of biomagnification and protein incorporation involving other non-protein amino acids. Amyotrophic Lateral Sclerosis 10 Supplement 221–25.
    1. Rubenstein E (2000) Biologic effects of and clinical disorders caused by nonprotein amino acids. Medicine 79: 80–89.
    1. Rodgers KJ, Wang H, Fu S, Dean RT (2002) Biosynthetic incorporation of oxidized amino acids into proteins and their cellular proteolysis. Free Radical Biology & Medicine 32: 766–775.
    1. Rodgers KJ, Shiozawa N (2008) Misincorporation of amino acid analogues into proteins by biosynthesis. The International Journal of Biochemistry & Cell Biology 40: 1452–1466.
    1. Allende CC, Allende JE (1964) Purification and substrate specificity of arginyl-ribonucleic acid synthetase from rat liver. The Journal of Biological Chemistry 239: 1102–1106.
    1. Rosenthal GA, Janzen DH, Dahlman DL (1977) Degradation and detoxification of canavanine by a specialized seed predator. Science 196: 658–660.
    1. Banack SA, Johnson HE, Cheng R, Cox PA (2007) Production of the neurotoxin BMAA by a marine cyanobacterium. Marine Drugs 5: 180–196.
    1. Cox PA, Banack SA, Murch SJ, Rasmussen U, Tien G, et al. (2005) Diverse taxa of cyanobacteria produce beta-N-methylamino-L-alanine, a neurotoxic amino acid. Proceedings of the National Academy of Sciences of the United States of America 102: 5074–5078.
    1. Murch SJ, Cox PA, Banack SA (2004) A mechanism for slow release of biomagnified cyanobacterial neurotoxins and neurodegenerative disease in Guam. Proceedings of the National Academy of Sciences of the United States of America 101: 12228–12231.
    1. Karlsson O, Berg C, Brittebo EB, Lindquist NG (2009) Retention of the cyanobacterial neurotoxin beta-N-methylamino-l-alanine in melanin and neuromelanin-containing cells – a possible link between Parkinson-dementia complex and pigmentary retinopathy. Pigment Cell & Melanoma Research 22: 120–130.
    1. Tso C, Rye K-A, Barter P (2012) Phenotypic and functional changes in blood monocytes following adherence to endothelium. PLOS ONE 7: e37091 doi:
    1. Minter AJ, Dawes J, Chesterman CN (1992) Effects of heparin and endothelial cell growth supplement on haemostatic functions of vascular endothelium. Thrombosis and Haemostasis 67: 718–723.
    1. Smith PK, Krohn RI, Hermanson GT, Mallia AK, Gartner FH, et al. (1985) Measurement of protein using bicinchoninic acid. Analytical Biochemistry 150: 76–85.
    1. Mondo K, Hammerschlag N, Basile M, Pablo J, Banack SA, et al. (2012) Cyanobacterial neurotoxin β-N-methylamino-L-alanine (BMAA) in shark fins. Marine Drugs 10: 509–520.
    1. Rodgers KJ, Hume PM, Dunlop RA, Dean RT (2004) Biosynthesis and turnover of DOPA-containing proteins by human cells. Free Radical Biology & Medicine 37: 1756–1764.
    1. Banack SA, Metcalf JS, Spácil Z, Downing TG, Downing S, et al. (2011) Distinguishing the cyanobacterial neurotoxin β-N-methylamino-L-alanine (BMAA) from other diamino acids. Toxicon 57: 730–738.
    1. Dunlop RA, Dean RT, Rodgers KJ (2008) The impact of specific oxidized amino acids on protein turnover in J774 cells. The Biochemical Journal 410: 131–140.
    1. Tang J, Dunlop RA, Rowe A, Rodgers KJ, Ramzan I (2011) Kavalactones Yangonin and Methysticin induce apoptosis in human hepatocytes (HepG2) in vitro . Phytotherapy Research 25: 417–423.
    1. Dunlop RA, Brunk UT, Rodgers KJ (2011) Proteins containing oxidized amino acids induce apoptosis in human monocytes. Biochemical Journal 435: 207–16.
    1. Luk KC, Kehm V, Carroll J, Zhang B, O’Brien P, et al. (2012) Pathological a-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science 338: 949–953.
    1. Costanzo M, Zurzolo C (2013) The cell biology of prion-like spread of protein aggregates: mechanisms and implication in neurodegeneration. Biochemical Journal 452: 1–17.
    1. Neumann M, Kwong LK, Lee EB, Kremmer E, Flatley A, et al. (2009) Phosphorylation of S409/410 of TDP-43 is a consistent feature in all sporadic and familial forms of TDP-43 proteinopathies. Acta Neuropathologica 117: 137–149.
    1. Polsky FI, Nunn PB, Bell EA (1972) Distribution and toxicity of alpha-amino-beta-methylaminopropionic acid. Federation Proceedings 31: 1473–1475.
    1. Cheng R, Banack SA (2009) Previous studies underestimate BMAA concentrations in cycad flour. Amyotrophic Lateral Sclerosis 10 Suppl 241–43.
    1. Cox PA, Banack SA, Murch SJ (2003) Biomagnification of cyanobacterial neurotoxins and neurodegenerative disease among the Chamorro people of Guam. Proceedings of the National Academy of Sciences of the United States of America 100: 13380–13383.
    1. Pablo J, Banack SA, Cox PA, Johnson TE, Papapetropoulos S, et al. (2009) Cyanobacterial neurotoxin BMAA in ALS and Alzheimer’s disease. Acta Neurologica Scandinavica 120: 216–225.
    1. Hendrickson TL, De Crécy-Lagard V, Schimmel P (2004) Incorporation of non-natural amino acids into proteins. Annual Review of Biochemistry 73: 147–176.
    1. Ozawa K, Headlam MJ, Mouradov D, Watt SJ, Beck JL, et al. (2005) Translational incorporation of L-3,4-dihydroxyphenylalanine into proteins. The FEBS Journal 272: 3162–3171.
    1. Jucker M, Walker LC (2011) Pathogenic protein seeding in Alzheimer’s disease and other neurodegenerative disorders. Annals of Neurology 70: 532–540.
    1. Okle O, Stemmer K, Deschl U, Dietrich DR (2012) L-BMAA induced ER stress and enhanced caspase 12 cleavage in human neuroblastoma SH-SY5Y cells at low nonexcitotoxic concentrations. Toxicological Sciences 131: 217–224.
    1. Cox PA, Richer R, Metcalf JS, Banack SA, Codd GA, et al. (2009) Cyanobacteria and BMAA exposure from desert dust: a possible link to sporadic ALS among Gulf War veterans. Amyotrophic Lateral Sclerosis 10 Suppl 2109–117.
    1. Xie X, Basile M, Mash DC (2013) Cerebral uptake and protein incorporation of cyanobacterial toxin BMAA. NeuroReport: doi .
    1. Karlsson O, Kultima K, Wadensten H, Nilsson A, Roman E, et al. (2013) Neurotoxin-induced neuropeptide perturbations in striatum of neonatal rats. Journal of Proteome Research 12: 1678–1690.
    1. Okle O, Rath L, Galizia CG, Dietrich DR (2013) The cyanobacterial neurotoxin beta-N-methylamino-l-alanine (BMAA) induces neuronal and behavioral changes in honeybees. Toxicology and Applied Pharmacology 270: 9–15.
    1. de Munck E, Muñoz-Sáez E, Miguel BG, Solas MT, Ojeda I, et al. (2013) β-N-methylamino-l-alanine causes neurological and pathological phenotypes mimicking Amyotrophic Lateral Sclerosis (ALS): The first step towards an experimental model for sporadic ALS. Environmental Toxicology and Pharmacology 36: 243–255.
    1. Chan SW, Dunlop RA, Rowe A, Double KL, Rodgers KJ (2012) L-DOPA is incorporated into brain proteins of patients treated for Parkinson’s disease, inducing toxicity in human neuroblastoma cells in vitro . Experimental Neurology 238: 29–37.
    1. Lobner D, Piana PMT, Salous AK, Peoples RW (2007) Beta-N-methylamino-L-alanine enhances neurotoxicity through multiple mechanisms. Neurobiology of Disease 25: 360–366.
    1. Chiu AS, Gehringer MM, Welch JH, Neilan BA (2011) Does α-amino-β-methylaminopropionic acid (BMAA) play a role in neurodegeneration? International Journal of Environmental Research and Public Health 8: 3728–3746.
    1. Chiu AS, Gehringer MM, Braidy N, Guillemin GJ, Welch JH, et al. (2013) Gliotoxicity of the cyanotoxin, β-methyl-amino-L-alanine (BMAA). Scientific Reports 3: 1482.
    1. Caller TA, Doolin JW, Haney JF, Murby AJ, West KG, et al. (2009) A cluster of amyotrophic lateral sclerosis in New Hampshire: a possible role for toxic cyanobacteria blooms. Amyotrophic Lateral Sclerosis 10 Suppl 2101–108.
    1. Field NC, Metcalf JS, Caller TA, Banack SA, Cox PA, et al. (2013) Linking β-methylamino-l-alanine exposure to sporadic amyotrophic lateral sclerosis in Annapolis, MD. Toxicon 70: 179–183.
    1. Bradley WG, Borenstein AR, Nelson LM, Codd GA, Rosen BH, et al. (2013) Is exposure to cyanobacteria an environmental risk factor for amyotrophic lateral sclerosis and other neurodegenerative diseases? Amyotrophic Lateral Sclerosis & Frontotemporal Degeneration 14: 325–33 doi:

Source: PubMed

3
Se inscrever