Plasmalogens and Alzheimer's disease: a review

Xiao Q Su, Junming Wang, Andrew J Sinclair, Xiao Q Su, Junming Wang, Andrew J Sinclair

Abstract

Growing evidence suggests that ethanolamine plasmalogens (PlsEtns), a subtype of phospholipids, have a close association with Alzheimer's disease (AD). Decreased levels of PlsEtns have been commonly found in AD patients, and were correlated with cognition deficit and severity of disease. Limited studies showed positive therapeutic outcomes with plasmalogens interventions in AD subjects and in rodents. The potential mechanisms underlying the beneficial effects of PlsEtns on AD may be related to the reduction of γ-secretase activity, an enzyme that catalyzes the synthesis of β-amyloid (Aβ), a hallmark of AD. Emerging in vitro evidence also showed that PlsEtns prevented neuronal cell death by enhancing phosphorylation of AKT and ERK signaling through the activation of orphan G-protein coupled receptor (GPCR) proteins. In addition, PlsEtns have been found to suppress the death of primary mouse hippocampal neuronal cells through the inhibition of caspase-9 and caspase-3 cleavages. Further in-depth investigations are required to determine the signature molecular species of PlsEtns associated with AD, hence their potential role as biomarkers. Clinical intervention with plasmalogens is still in its infancy but may have the potential to be explored for a novel therapeutic approach to correct AD pathology and neural function.

Keywords: Alzheimer’s disease; Biomarker; Mechanisms of action; Plasmalogens; Therapeutic efficacy.

Conflict of interest statement

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Chemical structures of Plasmalogens. R1 = palmitic acid (16:0) or stearic acid (18:0) or oleic acid (18:1). R2 = arachidonic acid (20:4) or docosahexaenoic acid (22:6) or oleic acid (18:1) or linoleic acid (18:2). When there is no double bond in the alkyl chain in the 1-position, the plasmalogens are referred to as alkyl plasmalogens, in contrast to when there is a double bond, as shown here, and they are referred to as alkenyl plasmalogens
Fig. 2
Fig. 2
Biosynthetic pathway of Plasmalogens. Abbreviations: DHAP, dihydroxyacetone phosphate; DHAPAT, dihydroxyacetone phosphate acyltransferase; DHAP, dihydroxyacetone phosphate; ADHAP-S, alkyl dihydroxyacetone phosphate synthase; FAR1/2, acyl-CoA reductase 1 and 2
Fig. 3
Fig. 3
Proposed mechanistic association of ethanolamine plasmalogens deficiency and Alzheimer’s disease. Abbreviations: Aβ, β-amyloid; PlsEtns, ethanolamine plasmalogens; VLCFAs, very long chain fatty acids. PlsEtns have a close association with Alzheimer’s disease (AD). Decreased levels of PlsEtns have been commonly found in AD patients, and are correlated with cognition deficit and severity of disease, although it is not known whether it is the cause or the consequence of the disease. It has been suggested maybe it is both. A few possible mechanisms with regards to the decrease of PlsEtns in AD have been suggested: peroxisome dysfunction, oxidative stress, alterations in membrane lipid rafts and inflammatory responses. Decreased PlsEtns may further enhance oxidative damage and alter membrane properties in AD. This plus increased membrane free cholesterol associated with PlsEtns deficiency could increase the production of Aβ. Aβ and reactive oxygen species could further decrease PlsEtns level. PlsEtns are major lipids facilitating membrane fusion of synaptic vesicles associated with neurotransmitter release, thus loss of PlsEtns could potentially contribute to the synaptic dysfunction and neurotransmitter depletion in Alzheimer’s disease. The association of decreased level of PlsEtns and neuroinflammation may be related to antioxidant properties of plasmalogens that protect cells from oxidative stress. Neuroinflammation has been reported to be associated with Aβ accumulation

References

    1. Braverman NE, Moser AB. Functions of plasmalogen lipids in health and disease. Biochim Biophys Acta (BBA) - Mol Basis Dis. 2012;1822:1442–1452. doi: 10.1016/j.bbadis.2012.05.008.
    1. Brites P, Waterham HR, Wanders RJ. Functions and biosynthesis of plasmalogens in health and disease. Biochim Biophys Acta Mol Cell Biol Lipids. 2004;1636:219–231. doi: 10.1016/j.bbalip.2003.12.010.
    1. Wood PL. Lipidomics of Alzheimer's disease: current status. Alzheimers Res Ther. 2012;4:5. doi: 10.1186/alzrt103.
    1. Hoerrmann W, Donis J, Sluga E, Stütz H, Paltauf F. Serum plasmalogens in ischemic cerebrovascular disease. VASA Z Gefasskrankheiten. 1991;20:319–322.
    1. Graessler J, Schwudke D, Schwarz PE, Herzog R, Shevchenko A, Bornstein SR. Top-down lipidomics reveals ether lipid deficiency in blood plasma of hypertensive patients. PLoS One. 2009;4:e6261. doi: 10.1371/journal.pone.0006261.
    1. Brosche T. Plasmalogen levels in serum from patients with impaired carbohydrate or lipid metabolism and in elderly subjects with normal metabolic values. Arch Gerontol Geriatr. 2001;32:283–294. doi: 10.1016/S0167-4943(01)00105-4.
    1. Messias MCF, Mecatti GC, Priolli DG, de Oliveira CP. Plasmalogen lipids: functional mechanism and their involvement in gastrointestinal cancer. Lipids Health Dis. 2018;17:41. doi: 10.1186/s12944-018-0685-9.
    1. Mandel H, Sharf R, Berant M, Wanders RJ, Vreken P, Aviram M. Plasmalogen phospholipids are involved in HDL-mediated cholesterol efflux: insights from investigations with plasmalogen-deficient cells. Biochem Biophys Res Commun. 1998;250:369–373. doi: 10.1006/bbrc.1998.9321.
    1. Farooqui AA, Horrocks LA, Farooqui T. Glycerophospholipids in brain: their metabolism, incorporation into membranes, functions, and involvement in neurological disorders. Chem Phys Lipids. 2000;106:1–29. doi: 10.1016/S0009-3084(00)00128-6.
    1. Zoeller RA, Nagan N, Gaposchkin DP, Legner MA, Lieberthal W. Plasmalogens as endogenous antioxidants: somatic cell mutants reveal the importance of the vinyl ether. Biochem J. 1999;338:769–776. doi: 10.1042/bj3380769.
    1. Broniec A, Klosinski R, Pawlak A, Wrona-Krol M, Thompson D, Sarna T. Interactions of plasmalogens and their diacyl analogs with singlet oxygen in selected model systems. Free Radic Biol Med. 2011;50:892–898. doi: 10.1016/j.freeradbiomed.2011.01.002.
    1. Sindelar PJ, Guan Z, Dallner G, Ernster L. The protective role of plasmalogens in iron-induced lipid peroxidation. Free Radic Biol Med. 1999;26:318–324. doi: 10.1016/S0891-5849(98)00221-4.
    1. Stables MJ, Gilroy DW. Old and new generation lipid mediators in acute inflammation and resolution. Prog Lipid Res. 2011;50:35–51. doi: 10.1016/j.plipres.2010.07.005.
    1. Jan AT, Azam M, Rahman S, Almigeiti A, Choi DH, Lee EJ, Haq QMR, Choi I. Perspective insights into disease progression, diagnostics, and therapeutic approaches in Alzheimer’s disease: a judicious update. Front Aging Neurosci. 2017;9:356. doi: 10.3389/fnagi.2017.00356.
    1. Fujino T, Yamada T, Asada T, Tsuboi Y, Wakana C, Mawatari S, Kono S. Efficacy and blood Plasmalogen changes by Oral Administration of Plasmalogen in patients with mild Alzheimer's disease and mild cognitive impairment: a multicenter, randomized, double-blind, placebo-controlled trial. EBioMedicine. 2017;17:199–205. doi: 10.1016/j.ebiom.2017.02.012.
    1. Lim WLF, Martins IJ, Martins RN. The involvement of lipids in Alzheimer's disease. J Genet Genomics. 2014;41:261–274. doi: 10.1016/j.jgg.2014.04.003.
    1. Mawatari S, Katafuchi T, Miake K, Fujino T. Dietary plasmalogen increases erythrocyte membrane plasmalogen in rats. Lipids Health Dis. 2012;11:161. doi: 10.1186/1476-511X-11-161.
    1. Farooqui AA, Horrocks LA. Book review: plasmalogens: workhorse lipids of membranes in normal and injured neurons and glia. Neuroscientist. 2001;7:232–245. doi: 10.1177/107385840100700308.
    1. Garcia C, Lutz NW, Confort-Gouny S, Cozzone PJ, Armand M, Bernard M. Phospholipid fingerprints of milk from different mammalians determined by 31 P NMR: towards specific interest in human health. Food Chem. 2012;135:1777–1783. doi: 10.1016/j.foodchem.2012.05.111.
    1. Fave G, Coste T, Armand M. Physicochemical properties of lipids: new strategies to manage fatty acid bioavailability. Cell Mol Biol. 2004;50:815–832.
    1. Nishimukai M, Wakisaka T, Hara H. Ingestion of plasmalogen markedly increased plasmalogen levels of blood plasma in rats. Lipids. 2003;38:1227–1235. doi: 10.1007/s11745-003-1183-9.
    1. Cohn JS, Kamili A, Wat E, Chung RW, Tandy S. Dietary phospholipids and intestinal cholesterol absorption. Nutrients. 2010;2:116–127. doi: 10.3390/nu2020116.
    1. Küllenberg D, Taylor LA, Schneider M, Massing U. Health effects of dietary phospholipids. Lipids Health Dis. 2012;11:3. doi: 10.1186/1476-511X-11-3.
    1. Astarita G, Jung K-M, Berchtold NC, Nguyen VQ, Gillen DL, Head E, Cotman CW, Piomelli D. Deficient liver biosynthesis of docosahexaenoic acid correlates with cognitive impairment in Alzheimer's disease. PLoS One. 2010;5:e12538. doi: 10.1371/journal.pone.0012538.
    1. Roels F, Espeel M, Pauwels M, De Craemer D, Egberts H, Van der Spek P. Different types of peroxisomes in human duodenal epithelium. Gut. 1991;32:858–865. doi: 10.1136/gut.32.8.858.
    1. Wiesner P, Leidl K, Boettcher A, Schmitz G, Liebisch G. Lipid profiling of FPLC-separated lipoprotein fractions by electrospray ionization tandem mass spectrometry. J Lipid Res. 2009;50:574–585. doi: 10.1194/jlr.D800028-JLR200.
    1. Candela P, Gosselet F, Miller F, Buee-Scherrer V, Torpier G, Cecchelli R, Fenart L. Physiological pathway for low-density lipoproteins across the blood-brain barrier: transcytosis through brain capillary endothelial cells in vitro. Endothelium. 2008;15:254–264. doi: 10.1080/10623320802487759.
    1. Honsho M, Yagita Y, Kinoshita N, Fujiki Y. Isolation and characterization of mutant animal cell line defective in alkyl-dihydroxyacetonephosphate synthase: localization and transport of plasmalogens to post-Golgi compartments. Biochimica et Biophysica Acta (BBA)-Molecular. Cell Res. 2008;1783:1857–1865.
    1. Pike LJ, Han X, Chung K-N, Gross RW. Lipid rafts are enriched in arachidonic acid and plasmenylethanolamine and their composition is independent of caveolin-1 expression: a quantitative electrospray ionization/mass spectrometric analysis. Biochemistry. 2002;41:2075–2088. doi: 10.1021/bi0156557.
    1. Wood PL, Mankidy R, Ritchie S, Heath D, Wood JA, Flax J, Goodenowe DB. Circulating plasmalogen levels and Alzheimer disease assessment scale–cognitive scores in Alzheimer patients. J Psychiatry Neurosci. 2010;35:59. doi: 10.1503/jpn.090059.
    1. Wallner S, Schmitz G. Plasmalogens the neglected regulatory and scavenging lipid species. Chem Phys Lipids. 2011;164:573–589. doi: 10.1016/j.chemphyslip.2011.06.008.
    1. Onodera T, Futai E, Kan E, Abe N, Uchida T, Kamio Y, Kaneko J. Phosphatidylethanolamine plasmalogen enhances the inhibiting effect of phosphatidylethanolamine on γ-secretase activity. J Biochem. 2014;157:301–309. doi: 10.1093/jb/mvu074.
    1. Jaunmuktane Z, Mead S, Ellis M, Wadsworth JD, Nicoll AJ, Kenny J, Launchbury F, Linehan J, Richard-Loendt A, Walker AS. Evidence for human transmission of amyloid-[bgr] pathology and cerebral amyloid angiopathy. Nature. 2015;525:247–250. doi: 10.1038/nature15369.
    1. Jucker M, Walker LC. Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature. 2013;501:45–51. doi: 10.1038/nature12481.
    1. Swerdlow RH, Burns JM, Khan SM. The Alzheimer's disease mitochondrial cascade hypothesis: progress and perspectives. Biochim Biophys Acta (BBA) - Mol Basis Dis. 2014;1842:1219–1231. doi: 10.1016/j.bbadis.2013.09.010.
    1. Hetz C, Martinon F, Rodriguez D, Glimcher LH. The unfolded protein response: integrating stress signals through the stress sensor IRE1α. Physiol Rev. 2011;91:1219–1243. doi: 10.1152/physrev.00001.2011.
    1. Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007;8:519–529. doi: 10.1038/nrm2199.
    1. Prince MJ, Wimo A, Guerchet MM, Ali GC, Wu Y-T, Prina M. World Alzheimer Report 2015 - The Global Impact of Dementia: An analysis of prevalence, incidence, cost and trends. London: Alzheimer's Disease International; 2015. p. 84.
    1. Coart E, Barrado LG, Duits FH, Scheltens P, van der Flier WM, Teunissen CE, van der Vies SM, Burzykowski T. Correcting for the absence of a gold standard improves diagnostic accuracy of biomarkers in Alzheimer’s disease. J Alzheimers Dis. 2015;46:889–899. doi: 10.3233/JAD-142886.
    1. Tyas SL, Manfreda J, Strain LA, Montgomery PR. Risk factors for Alzheimer's disease: a population-based, longitudinal study in Manitoba, Canada. Int J Epidemiol. 2001;30:590–597. doi: 10.1093/ije/30.3.590.
    1. Cummings JL, Mackell J, Kaufer D. Behavioral effects of current Alzheimer’s disease treatments: a descriptive review. Alzheimers Dement. 2008;4:49–60. doi: 10.1016/j.jalz.2007.10.011.
    1. Ginsberg L, Rafique S, Xuereb JH, Rapoport SI, Gershfeld NL. Disease and anatomic specificity of ethanolamine plasmalogen deficiency in Alzheimer's disease brain. Brain Res. 1995;698:223–226. doi: 10.1016/0006-8993(95)00931-F.
    1. Wood PL, Barnette BL, Kaye JA, Quinn JF, Woltjer RL. Non-targeted lipidomics of CSF and frontal cortex grey and white matter in control, mild cognitive impairment, and Alzheimer’s disease subjects. Acta Neuropsychiatr. 2015;27:270–278. doi: 10.1017/neu.2015.18.
    1. Han X, Holtzman DM, McKeel DW. Plasmalogen deficiency in early Alzheimer's disease subjects and in animal models: molecular characterization using electrospray ionization mass spectrometry. J Neurochem. 2001;77:1168–1180. doi: 10.1046/j.1471-4159.2001.00332.x.
    1. Ellison DW, Beal MF, Martin JB. Phosphoethanolamine and ethanolamine are decreased in Alzheimer's disease and Huntington's disease. Brain Res. 1987;417:389–392. doi: 10.1016/0006-8993(87)90471-9.
    1. Rothhaar TL, Grösgen S, Haupenthal VJ, Burg VK, Hundsdörfer B, Mett J, Riemenschneider M, Grimm HS, Hartmann T, Grimm MO. Plasmalogens inhibit APP processing by directly affecting γ-secretase activity in Alzheimer’s disease. Sci World J. 2012;2012. Article ID 141240. 10.1100/2012/141240.
    1. Molina J, Jimenez-Jimenez F, Vargas C, Gomez P, De Bustos F, Orti-Pareja M, Tallon-Barranco A, Benito-Leon J, Arenas J, Enriquez-de-Salamanca R. Cerebrospinal fluid levels of non-neurotransmitter amino acids in patients with Alzheimer's disease. J Neural Transm. 1998;105:279–286. doi: 10.1007/s007020050057.
    1. Yamashita S, Kiko T, Fujiwara H, Hashimoto M, Nakagawa K, Kinoshita M, Furukawa K, Arai H, Miyazawa T. Alterations in the levels of amyloid-β, phospholipid hydroperoxide, and plasmalogen in the blood of patients with Alzheimer’s disease: possible interactions between amyloid-β and these lipids. J Alzheimers Dis. 2016;50:527–537. doi: 10.3233/JAD-150640.
    1. Goodenowe DB, Cook LL, Liu J, Lu Y, Jayasinghe DA, Ahiahonu PW, Heath D, Yamazaki Y, Flax J, Krenitsky KF. Peripheral ethanolamine plasmalogen deficiency: a logical causative factor in Alzheimer's disease and dementia. J Lipid Res. 2007;48:2485–2498. doi: 10.1194/jlr.P700023-JLR200.
    1. Wood PL, Locke VA, Herling P, Passaro A, Vigna GB, Volpato S, Valacchi G, Cervellati C, Zuliani G. Targeted lipidomics distinguishes patient subgroups in mild cognitive impairment (MCI) and late onset Alzheimer's disease (LOAD) BBA Clinical. 2016;5:25–28. doi: 10.1016/j.bbacli.2015.11.004.
    1. Han X. Lipid alterations in the earliest clinically recognizable stage of Alzheimer's disease: implication of the role of lipids in the pathogenesis of Alzheimer's disease. Curr Alzheimer Res. 2005;2:65–77. doi: 10.2174/1567205052772786.
    1. Grimm MO, Mett J, Stahlmann CP, Haupenthal VJ, Blümel T, Stötzel H, Grimm HS, Hartmann T. Eicosapentaenoic acid and docosahexaenoic acid increase the degradation of amyloid-β by affecting insulin-degrading enzyme. Biochem Cell Biol. 2016;94:534–542. doi: 10.1139/bcb-2015-0149.
    1. Hopperton KE, Trépanier M-O, Giuliano V, Bazinet RP. Brain omega-3 polyunsaturated fatty acids modulate microglia cell number and morphology in response to intracerebroventricular amyloid-β 1-40 in mice. J Neuroinflammation. 2016;13:257. doi: 10.1186/s12974-016-0721-5.
    1. Ren H, Luo C, Feng Y, Yao X, Shi Z, Liang F, Kang JX, Wan J-B, Pei Z, Su H. Omega-3 polyunsaturated fatty acids promote amyloid-β clearance from the brain through mediating the function of the glymphatic system. FASEB J. 2017;31:282–293. doi: 10.1096/fj.201600896.
    1. Astarita G, Piomelli D. Towards a whole-body systems [multi-organ] lipidomics in Alzheimer’s disease. Prostaglandins. Leukot Essent Fatty Acids (PLEFA) 2011;85:197–203. doi: 10.1016/j.plefa.2011.04.021.
    1. Crawford MA, Bazinet RP, Sinclair AJ. Fat intake and CNS functioning: ageing and disease. Ann Nutr Metab. 2009;55:202–228. doi: 10.1159/000229003.
    1. L. Paul, Amin M., Mankidy Rishikesh, Smith Tara, B. Dayan. Alzheimer's Disease Pathogenesis-Core Concepts, Shifting Paradigms and Therapeutic Targets. 2011. Plasmalogen Deficit: A New and Testable Hypothesis for the Etiology of Alzheimer’s Disease.
    1. Kou J, Kovacs GG, Höftberger R, Kulik W, Brodde A, Forss-Petter S, Hönigschnabl S, Gleiss A, Brügger B, Wanders R. Peroxisomal alterations in Alzheimer’s disease. Acta Neuropathol. 2011;122:271–283. doi: 10.1007/s00401-011-0836-9.
    1. Katafuchi T, Ifuku M, Mawatari S, Noda M, Miake K, Sugiyama M, Fujino T. Effects of plasmalogens on systemic lipopolysaccharide-induced glial activation and β-amyloid accumulation in adult mice. Ann N Y Acad Sci. 2012;1262:85–92. doi: 10.1111/j.1749-6632.2012.06641.x.
    1. Grimm MO, Kuchenbecker J, Rothhaar TL, Grösgen S, Hundsdörfer B, Burg VK, Friess P, Müller U, Grimm HS, Riemenschneider M. Plasmalogen synthesis is regulated via alkyl-dihydroxyacetonephosphate-synthase by amyloid precursor protein processing and is affected in Alzheimer’s disease. J Neurochem. 2011;116:916–925. doi: 10.1111/j.1471-4159.2010.07070.x.
    1. Mangold HK, Weber N. Biosynthesis and biotransformation of ether lipids. Lipids. 1987;22:789–799. doi: 10.1007/BF02535533.
    1. Reiss D, Beyer K, Engelmann B. Delayed oxidative degradation of polyunsaturated diacyl phospholipids in the presence of plasmalogen phospholipids in vitro. Biochem J. 1997;323:807–814. doi: 10.1042/bj3230807.
    1. Hartmann T, Kuchenbecker J, Grimm MO. Alzheimer’s disease: the lipid connection. J Neurochem. 2007;103:159–170. doi: 10.1111/j.1471-4159.2007.04715.x.
    1. Lee J, Culyba EK, Powers ET, Kelly JW. Amyloid-β forms fibrils by nucleated conformational conversion of oligomers. Nat Chem Biol. 2011;7:602–609. doi: 10.1038/nchembio.624.
    1. Farooqui AA, Horrocks LA. Plasmalogens, phospholipase A2, and docosahexaenoic acid turnover in brain tissue. J Mol Neurosci. 2001;16:263–272. doi: 10.1385/JMN:16:2-3:263.
    1. Latorre E, Collado MP, Fernández I, Aragonés MD, Catalán RE. Signaling events mediating activation of brain ethanolamine plasmalogen hydrolysis by ceramide. FEBS J. 2003;270:36–46.
    1. Di Paolo G, Kim T-W. Linking lipids to Alzheimer's disease: cholesterol and beyond. Nat Rev Neurosci. 2011;12:284–296. doi: 10.1038/nrn3012.
    1. Florent-Bechard S, Desbene C, Garcia P, Allouche A, Youssef I, Escanye M-C, Koziel V, Hanse M, Malaplate-Armand C, Stenger C. The essential role of lipids in Alzheimer's disease. Biochimie. 2009;91:804–809. doi: 10.1016/j.biochi.2009.03.004.
    1. Selkoe DJ. Resolving controversies on the path to Alzheimer's therapeutics. Nat Med. 2011;17:1060–1065. doi: 10.1038/nm.2460.
    1. Hossain MS, Mineno K, Katafuchi T. Neuronal orphan G-protein coupled receptor proteins mediate plasmalogens-induced activation of ERK and Akt signaling. PLoS One. 2016;11:e0150846. doi: 10.1371/journal.pone.0150846.
    1. Hossain MS, Ifuku M, Take S, Kawamura J, Miake K, Katafuchi T. Plasmalogens rescue neuronal cell death through an activation of AKT and ERK survival signaling. PLoS One. 2013;8:e83508. doi: 10.1371/journal.pone.0083508.

Source: PubMed

3
Subskrybuj