Answered on April 11, 2025
Plasmalogen is a subclass of glycerophospholipids characterized by a vinyl-ether bond at the sn-1 position and typically a polyunsaturated fatty acid at the sn-2 position of the glycerol backbone. These lipids are integral components of cell membranes, particularly abundant in neuronal, immune, and cardiovascular tissues.[1-2]
Plasmalogens play several critical roles in cellular physiology. They are involved in membrane organization, fluidity, and the function of integral membrane proteins and lipid rafts.[2] Additionally, plasmalogens act as endogenous antioxidants, protecting cell membrane components from oxidative stress.[2][4] Their deficiency has been associated with various neurodegenerative and metabolic diseases, such as Alzheimer's disease and Parkinson's disease.[3][5]
The biosynthesis of plasmalogens begins in peroxisomes and is completed in the endoplasmic reticulum.[6-7] Dysregulation of plasmalogen homeostasis can impair cholesterol biosynthesis and is linked to several pathological conditions, including peroxisome biogenesis disorders and rhizomelic chondrodysplasia punctata.[6]
In summary, plasmalogens are essential glycerophospholipids with significant roles in membrane structure, antioxidative defense, and cellular signaling, with their dysregulation contributing to various diseases.

1.
Plasmalogens: A Potential Therapeutic Target for Neurodegenerative and Cardiometabolic Disease.

Paul S, Lancaster GI, Meikle PJ.

Progress in Lipid Research. 2019;74:186-195. doi:10.1016/j.plipres.2019.04.003.

Leading Journal

Plasmalogens are a class of membrane glycerophospholipids with unique properties. They contain a vinyl-ether linked alkyl chain at the sn-1 position of the glycerol backbone and, typically, a polyunsaturated fatty acyl chain at the sn-2 position. Plasmalogens are critical for human health and have established roles in neuronal development, the immune response and as endogenous antioxidants. However, the mechanistic bases of these and other biological functions of plasmalogens are not well defined. Lipidomic studies have characterised reduced levels of plasmalogens in a number of disease states, including neurodegenerative and cardiometabolic disease, highlighting the potential of plasmalogen modulation as a therapeutic strategy. A number of approaches have been proposed to upregulate plasmalogen levels in different clinical settings; these include dietary intervention with inositol or the naturally occurring metabolic precursors known as alkylglycerols. Plasmalogen modulation has been utilised in both preclinical and clinical studies to prevent onset and/or attenuate progression of neurodegenerative diseases, atherosclerosis, insulin resistance and hepatosteatosis. These studies are providing new insight into the mechanistic role of plasmalogens in disease and their therapeutic potential. In this review, we will examine the strategies for plasmalogen modulation and recent progress toward therapeutic applications with a focus on neurodegenerative and cardiometabolic disease.

2.
Potential Role of Plasmalogens in the Modulation of Biomembrane Morphology.

Almsherqi ZA.

Frontiers in Cell and Developmental Biology. 2021;9:673917. doi:10.3389/fcell.2021.673917.

Plasmalogens are a subclass of cell membrane glycerophospholipids that typically include vinyl- ether bond at the sn-1 position and polyunsaturated fatty acid at the sn-2 position. They are highly abundant in the neuronal, immune, and cardiovascular cell membranes. Despite the abundance of plasmalogens in a plethora of cells, tissues, and organs, the role of plasmalogens remains unclear. Plasmalogens are required for the proper function of integral membrane proteins, lipid rafts, cell signaling, and differentiation. More importantly, plasmalogens play a crucial role in the cell as an endogenous antioxidant that protects the cell membrane components such as phospholipids, unsaturated fatty acids, and lipoproteins from oxidative stress. The incorporation of vinyl-ether linked with alkyl chains in phospholipids alter the physicochemical properties (e.g., the hydrophilicity of the headgroup), packing density, and conformational order of the phospholipids within the biomembranes. Thus, plasmalogens play a significant role in determining the physical and chemical properties of the biomembrane such as its fluidity, thickness, and lateral pressure of the biomembrane. Insights on the important structural and functional properties of plasmalogens may help us to understand the molecular mechanism of membrane transformation, vesicle formation, and vesicular fusion, especially at the synaptic vesicles where plasmalogens are rich and essential for neuronal function. Although many aspects of plasmalogen phospholipid involvement in membrane transformation identified through experiments and membrane mimic systems, remain to be confirmed , the compiled data show many intriguing properties of vinyl-ether bonded lipids that may play a significant role in the structural and morphological changes of the biomembranes. In this review, we present the current limited knowledge of the emerging potential role of plasmalogens as a modulator of the biomembrane morphology.

3.
Advances in the Biosynthetic Pathways and Application Potential of Plasmalogens in Medicine.

Zhou Y, Yu N, Zhao J, et al.

Frontiers in Cell and Developmental Biology. 2020;8:765. doi:10.3389/fcell.2020.00765.

Plasmalogens are a special class of polar glycerolipids containing a vinyl-ether bond and an ester bond at sn-1 and sn-2 positions of the glycerol backbone, respectively. In animals, impaired biosynthesis and regulation of plasmalogens may lead to certain neurological and metabolic diseases. Plasmalogens deficiency was proposed to be strongly associated with neurodegenerative and metabolic diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), and appropriate supplement of plasmalogens could help to prevent and possibly provide therapy of these diseases. Plasmalogens evolved first in anaerobic bacteria with an anaerobic biosynthetic pathway. Later, an oxygen-dependent biosynthesis of plasmalogens appeared in animal cells. This review summarizes and updates current knowledge of anaerobic and aerobic pathways of plasmalogens biosynthesis, including the enzymes involved, steps and aspects of the regulation of these processes. Strategies for increasing the expression of plasmalogen synthetic genes using synthetic biology techniques under specific conditions are discussed. Deep understanding of plasmalogens biosynthesis will provide the bases for the use of plasmalogens and their precursors as potential therapeutic regimens for age-related degenerative and metabolic diseases.

4.

Plasmalogens are a specific glycerophospholipid class containing a vinyl ether moiety at the sn-1-position of the glycerol backbone. The high susceptibility of this vinyl ether bond to oxidative damage and traces of acids may indicate the possible function of plasmalogens in biological systems: The regarded cell-internal antioxidative defense of membranes by protecting other phospholipids or lipoprotein particles against oxidative stress is controversial. Reactive oxygen species preferably affect the vinyl ether function as well as the olefinic acyl residues at the sn-2-position of plasmalogens. This review is dedicated to the role of plasmalogens in different cells and tissues as spermatozoal cells or brain tissue. The first chapter of this review will discuss the molecular structure and chemistry of plasmalogen molecules, their distributions in cells and tissues and the species-specificity. In the second chapter their functions as lipid mediators will be considered and the controversial antioxidative function will be discussed. The supposed function of plasmalogens as "scavengers" for reactive oxygen species (ROS) in biological membranes is challenged by the finding that plasmalogen oxidation products as alpha-hydroxyaldehydes and plasmalogen epoxides accumulate in all chronic diseases as atherosclerosis and myocardial infarction, upon aging as well as in Alzheimers disease and other neuropathological conditions. All these conditions, characterized by increased membrane instability and oxidative damage, will be reviewed in chapter three. Chronically proceeding processes can be described by permanently invading polymorphonuclear neutrophils into inflammatory loci. The degranulation of the azurophilic granula in polymorphonuclear leukocytes causes the release of highly reactive substances, for instance the myeloperoxidase-generated hypochlorous acid (HOCl) acting as effective oxidant. Therefore, special attention will be paid to neutrophil-derived HOCl. The last chapter deals with currently used methods of detecting plasmalogens and their degradation products. Although chromatographic methods will be also discussed, special attention will be given to (31)P NMR spectroscopy and soft ionization techniques of mass spectrometry as electrospray ionization or matrix-assisted laser desorption and ionization time-of-flight mass spectrometry.

5.
Quantitative Analysis of Ethanolamine Plasmalogen Species in Red Blood Cells Using Liquid Chromatography Tandem Mass Spectrometry for Diagnosing Peroxisome Biogenesis Disorders.

De Biase I, Yuzyuk T, Cui W, et al.

Clinica Chimica Acta; International Journal of Clinical Chemistry. 2023;542:117295. doi:10.1016/j.cca.2023.117295.

Plasmalogens are glycerophospholipids characterized by a vinyl-ether bond with a fatty alcohol at the sn-1 position, a polyunsaturated fatty acid at the sn-2 position, and a polar head at the sn-3 position, commonly phosphoethanolamine. Plasmalogens play crucial roles in several cellular processes. Reduced levels have been associated with Alzheimer's and Parkinson's disease progression. Markedly reduced plasmalogens are a classic feature of peroxisome biogenesis disorders (PBD) because plasmalogen synthesis requires functional peroxisomes. Particularly, severe plasmalogen deficiency is the biochemical hallmark of rhizomelic chondrodysplasia punctata (RCDP). Traditionally, plasmalogens are evaluated in red blood cells (RBCs) by gas-chromatography/mass-spectrometry (GC-MS), which cannot distinguish individual species. We developed a liquid-chromatography/tandem mass-spectrometry (LC-MS/MS) method to quantify eighteen phosphoethanolamine plasmalogens in RBCs to diagnose PBD patients, especially RCDP. Validation results showed a specific, robust, and precise method with broad analytical range. Age-specific reference intervals were established; control medians were used to assess plasmalogen deficiency in patients' RBCs. Clinical utility was also confirmed in Pex7 deficient mouse models recapitulating severe and milder RCDP clinical phenotypes. To our knowledge, this is the first attempt to replace the GC-MS method in the clinical laboratory. In addition to diagnosing PBDs, structure-specific plasmalogen quantitation could help understand disease pathogenesis and monitor therapy.

6.
Plasmalogen Homeostasis - Regulation of Plasmalogen Biosynthesis and Its Physiological Consequence in Mammals.

Honsho M, Fujiki Y.

FEBS Letters. 2017;591(18):2720-2729. doi:10.1002/1873-3468.12743.

Plasmalogens, mostly ethanolamine-containing alkenyl ether phospholipids, are a major subclass of glycerophospholipids. Plasmalogen synthesis is initiated in peroxisomes and completed in the endoplasmic reticulum. The absence of plasmalogens in several organs of peroxisome biogenesis-defective patients suggests that the de novo synthesis of plasmalogens plays a pivotal role in its homeostasis in tissues. Plasmalogen synthesis is regulated by modulating the stability of fatty acyl-CoA reductase 1 on peroxisomal membranes, a rate-limiting enzyme in plasmalogen synthesis, by sensing plasmalogens in the inner leaflet of plasma membranes. Dysregulation of plasmalogen homeostasis impairs cholesterol biosynthesis by altering the stability of squalene monooxygenase, a key enzyme in cholesterol biosynthesis, implying physiological consequences of plasmalogen homeostasis with respect to cholesterol metabolism in cells, as well as in organs such as the liver.

7.
Regulation of Plasmalogen Biosynthesis in Mammalian Cells and Tissues.

Honsho M, Fujiki Y.

Brain Research Bulletin. 2023;194:118-123. doi:10.1016/j.brainresbull.2023.01.011.

Plasmalogens are a unique family of cellular glycerophospholipids that contain a vinyl-ether bond. Synthesis of plasmalogens is initiated in peroxisomes and completed in the endoplasmic reticulum. The absence of plasmalogens in several organs of patients with deficiency in peroxisome biogenesis suggests that de novo synthesis of plasmalogens contributes significantly to plasmalogen homeostasis in humans. Plasmalogen biosynthesis is spatiotemporally regulated by a feedback mechanism that senses the amount of plasmalogens in the inner leaflet of the plasma membrane and regulates the stability of fatty acyl-CoA reductase 1 (FAR1), the rate-limiting enzyme for plasmalogen biosynthesis. Dysregulation of plasmalogen synthesis impairs cholesterol synthesis in cells and brain, resulting in the reduced expression of genes such as mRNA encoding myelin basic protein, a phenotype found in the cerebellum of plasmalogen-deficient mice. In this review, we summarize the current knowledge of molecular mechanisms underlying the regulation of plasmalogen biosynthesis and the link between plasmalogen homeostasis and cholesterol biosynthesis, and address the pathogenesis of impaired plasmalogen homeostasis in rodent and humans.

Expanded question: What causes dysregulation in plasmalogens, a subclass of glycerophospholipids, and how does this dysregulation impact cellular functions and contribute to diseases such as Alzheimer's disease and Parkinson's disease?

Answered on April 11, 2025
Dysregulation in plasmalogens can be caused by several factors, including genetic mutations, oxidative stress, and inflammatory stimuli. Genetic mutations affecting enzymes involved in plasmalogen biosynthesis, such as glycerone phosphate O-acyltransferase (Gnpat) and alkyl-dihydroxyacetonephosphate synthase (AGPS), can lead to reduced plasmalogen levels. Additionally, oxidative stress and inflammatory stimuli can downregulate these enzymes through pathways involving NF-κB and c-Myc, further reducing plasmalogen synthesis.[1]
Impact on cellular functions: Plasmalogens are crucial for maintaining membrane integrity, fluidity, and function. They also act as antioxidants, protecting cells from oxidative damage. Dysregulation of plasmalogens can impair these functions, leading to increased oxidative stress, disrupted membrane dynamics, and altered signaling pathways.[3-4]
Contribution to diseases: In Alzheimer's disease (AD), reduced plasmalogen levels are associated with increased amyloid-beta production and neuroinflammation. Plasmalogens modulate γ-secretase activity, which is involved in amyloid-beta synthesis, and their deficiency can exacerbate amyloid plaque formation. In Parkinson's disease (PD), plasmalogen deficiency is linked to mitochondrial dysfunction and increased oxidative stress, contributing to neuronal degeneration.[5-6]
In summary, dysregulation in plasmalogens is caused by genetic mutations, oxidative stress, and inflammatory stimuli, leading to impaired cellular functions and contributing to neurodegenerative diseases such as Alzheimer's and Parkinson's disease through mechanisms involving increased oxidative stress, disrupted membrane dynamics, and altered signaling pathways.[1][3][5-6]

1.
Reduction of Ether-Type Glycerophospholipids, Plasmalogens, by NF-κB Signal Leading to Microglial Activation.

Hossain MS, Abe Y, Ali F, et al.

The Journal of Neuroscience : The Official Journal of the Society for Neuroscience. 2017;37(15):4074-4092. doi:10.1523/JNEUROSCI.3941-15.2017.

Neuroinflammation characterized by activation of glial cells is observed in various neurodegenerative diseases including Alzheimer's disease (AD). Although the reduction of ether-type glycerophospholipids, plasmalogens (Pls), in the brain is reported in AD patients, the mechanism of the reduction and its impact on neuroinflammation remained elusive. In the present study, we found for the first time that various inflammatory stimuli reduced Pls levels in murine glial cells via NF-κB activation, which then downregulated a Pls-synthesizing enzyme, glycerone phosphate O-acyltransferase (Gnpat) through increased c-Myc recruitment onto the promoter. We also found that systemic injection of lipopolysaccharide, aging, and chronic restraint stress reduced brain Pls contents that were associated with glial NF-κB activation, an increase in c-Myc expression, and downregulation of in the mouse cortex and hippocampus. More interestingly, the reduction of Pls contents in the murine cortex itself could increase the activated phenotype of microglial cells and the expression of proinflammatory cytokines, suggesting further acceleration of neuroinflammation by reduction of brain Pls. A similar mechanism of reduction was also found in human cell lines, triple-transgenic AD mouse brain, and postmortem human AD brain tissues. These findings suggest a novel mechanism of neuroinflammation that may explain prolonged progression of AD and help us to explore preventive and therapeutic strategies to treat neurodegenerative diseases. Ether-type glycerophospholipids, plasmalogens (Pls), are reduced in the brain of Alzheimer disease (AD) patients. We found that inflammatory stimuli reduced Pls contents by downregulation of the Pls-synthesizing enzyme glycerone phosphate O-acyltransferase (Gnpat) through NF-κB-mediated recruitment of c-Myc onto the promoter in both murine and human cell lines. Murine brains after systemic lipopolysaccharide, chronic stress, and aging, as well as triple-transgenic AD mice and postmortem human AD brain tissues all showed increased c-Myc and reduced expression. Interestingly, knockdown of itself activated NF-κB in glial cell lines and microglia in mouse cortex. Our findings provide a new insight into the mechanism of neuroinflammation and may help to develop a novel therapeutic approach for neurodegenerative diseases such as AD.

2.
Plasmalogen Synthesis Is Regulated via Alkyl-Dihydroxyacetonephosphate-Synthase by Amyloid Precursor Protein Processing and Is Affected in Alzheimer's Disease.

Grimm MO, Kuchenbecker J, Rothhaar TL, et al.

Journal of Neurochemistry. 2011;116(5):916-25. doi:10.1111/j.1471-4159.2010.07070.x.

Lipids play an important role as risk or protective factors in Alzheimer's disease, which is characterized by amyloid plaques composed of aggregated amyloid-beta. Plasmalogens are major brain lipids and controversially discussed to be altered in Alzheimer's disease (AD) and whether changes in plasmalogens are cause or consequence of AD pathology. Here, we reveal a new physiological function of the amyloid precursor protein (APP) in plasmalogen metabolism. The APP intracellular domain was found in vivo and in vitro to increase the expression of the alkyl-dihydroxyacetonephosphate-synthase (AGPS), a rate limiting enzyme in plasmalogen synthesis. Alterations in APP dependent changes of AGPS expression result in reduced protein and plasmalogen levels. Under the pathological situation of AD, increased amyloid-beta level lead to increased reactive oxidative species production, reduced AGPS protein and plasmalogen level. Accordingly, phosphatidylethanol plasmalogen was decreased in the frontal cortex of AD compared to age matched controls. Our findings elucidate that plasmalogens are decreased as a consequence of AD and regulated by APP processing under physiological conditions.

3.
Plasmalogens: A Potential Therapeutic Target for Neurodegenerative and Cardiometabolic Disease.

Paul S, Lancaster GI, Meikle PJ.

Progress in Lipid Research. 2019;74:186-195. doi:10.1016/j.plipres.2019.04.003.

Leading Journal

Plasmalogens are a class of membrane glycerophospholipids with unique properties. They contain a vinyl-ether linked alkyl chain at the sn-1 position of the glycerol backbone and, typically, a polyunsaturated fatty acyl chain at the sn-2 position. Plasmalogens are critical for human health and have established roles in neuronal development, the immune response and as endogenous antioxidants. However, the mechanistic bases of these and other biological functions of plasmalogens are not well defined. Lipidomic studies have characterised reduced levels of plasmalogens in a number of disease states, including neurodegenerative and cardiometabolic disease, highlighting the potential of plasmalogen modulation as a therapeutic strategy. A number of approaches have been proposed to upregulate plasmalogen levels in different clinical settings; these include dietary intervention with inositol or the naturally occurring metabolic precursors known as alkylglycerols. Plasmalogen modulation has been utilised in both preclinical and clinical studies to prevent onset and/or attenuate progression of neurodegenerative diseases, atherosclerosis, insulin resistance and hepatosteatosis. These studies are providing new insight into the mechanistic role of plasmalogens in disease and their therapeutic potential. In this review, we will examine the strategies for plasmalogen modulation and recent progress toward therapeutic applications with a focus on neurodegenerative and cardiometabolic disease.

4.
Plasmalogens, Platelet-Activating Factor and Beyond - Ether Lipids in Signaling and Neurodegeneration.

Dorninger F, Forss-Petter S, Wimmer I, Berger J.

Neurobiology of Disease. 2020;145:105061. doi:10.1016/j.nbd.2020.105061.

Glycerol-based ether lipids including ether phospholipids form a specialized branch of lipids that in mammals require peroxisomes for their biosynthesis. They are major components of biological membranes and one particular subgroup, the plasmalogens, is widely regarded as a cellular antioxidant. Their vast potential to influence signal transduction pathways is less well known. Here, we summarize the literature showing associations with essential signaling cascades for a wide variety of ether lipids, including platelet-activating factor, alkylglycerols, ether-linked lysophosphatidic acid and plasmalogen-derived polyunsaturated fatty acids. The available experimental evidence demonstrates links to several common players like protein kinase C, peroxisome proliferator-activated receptors or mitogen-activated protein kinases. Furthermore, ether lipid levels have repeatedly been connected to some of the most abundant neurological diseases, particularly Alzheimer's disease and more recently also neurodevelopmental disorders like autism. Thus, we critically discuss the potential role of these compounds in the etiology and pathophysiology of these diseases with an emphasis on signaling processes. Finally, we review the emerging interest in plasmalogens as treatment target in neurological diseases, assessing available data and highlighting future perspectives. Although many aspects of ether lipid involvement in cellular signaling identified in vitro still have to be confirmed in vivo, the compiled data show many intriguing properties and contributions of these lipids to health and disease that will trigger further research.

5.
Plasmalogens and Alzheimer's Disease: A Review.

Su XQ, Wang J, Sinclair AJ.

Lipids in Health and Disease. 2019;18(1):100. doi:10.1186/s12944-019-1044-1.

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.

6.
Therapeutic Efficacy of Plasmalogens for Alzheimer's Disease, Mild Cognitive Impairment, and Parkinson's Disease in Conjunction With a New Hypothesis for the Etiology of Alzheimer's Disease.

Fujino T, Hossain MS, Mawatari S.

Advances in Experimental Medicine and Biology. 2020;1299:195-212. doi:10.1007/978-3-030-60204-8_14.

It has been reported in recent years that blood levels of plasmalogens (Pls) are decreased in various diseases. None of those reports, however, conducted any clinical trials to examine the effect of Pls on those diseases. This article describes our recent report on a therapeutic efficacy of orally administered Pls in mild cognitive impairment (MCI), mild to severe Alzheimer's disease (AD), and Parkinson's disease (PD). A 24-week, multicenter, randomized, double-blind, placebo-controlled trial was performed in patients with MCI (n = 178) and mild AD (n = 98). The study design for moderate AD (n = 57) and severe AD (n = 18) was 12-week open-labeled, and the design for patients with PD (n = 10) was 24-week open-labeled. They showed a significant improvement in cognitive function and other clinical symptoms with elevation of the blood Pls levels. No adverse events were reported. The baseline levels of plasma ethanolamine plasmalogen and erythrocyte ethanolamine plasmalogen in MCI, AD, and PD were significantly lower than those of normal aged. The degree of reduction in the blood Pls levels was in the order of MCI ≺ mild AD ≺ moderate AD ≺ severe AD ≺ PD. The findings suggest that the blood levels of Pls may be a beneficial biomarker for assessing AD severity. Based on these results, we have proposed a new hypothesis for the etiology of AD and other neuropsychiatric disorders.

Expanded question: What other environmental toxins can cause dysregulation in the development of plasmalogens, similar to the effects of genetic mutations, oxidative stress, and inflammatory stimuli?

Answered on April 11, 2025
Environmental toxins that can cause dysregulation in the development of plasmalogens include hypochlorous acid (HOCl) and lipopolysaccharide (LPS).
Hypochlorous acid, generated by myeloperoxidase (MPO) from hydrogen peroxide and chloride ions, targets plasmalogens due to their vinyl ether bond, leading to oxidative modification and degradation. This process results in the generation of 2-chloro fatty aldehydes and lysophospholipids, compromising normal neuronal function and synaptic transmission.[1]
Lipopolysaccharide, a component of the outer membrane of Gram-negative bacteria, can induce systemic inflammation and neuroinflammation. LPS exposure leads to NF-κB activation, which downregulates the plasmalogen-synthesizing enzyme glycerone phosphate O-acyltransferase (Gnpat) through increased c-Myc recruitment onto the promoter, thereby reducing plasmalogen levels.[2]
These environmental toxins, similar to genetic mutations, oxidative stress, and inflammatory stimuli, contribute to the dysregulation of plasmalogen synthesis and homeostasis, impacting cellular functions and potentially exacerbating neurodegenerative diseases.

1.
Mouse Brain Plasmalogens Are Targets for Hypochlorous Acid-Mediated Modification in Vitro and in Vivo.

Ullen A, Fauler G, Köfeler H, et al.

Free Radical Biology & Medicine. 2010;49(11):1655-65. doi:10.1016/j.freeradbiomed.2010.08.025.

Leading Journal

Plasmalogens, 1-O-alk-1'-enyl-2-acyl-sn-glycerophospholipids, are significant constituents of cellular membranes and are essential for normal brain development. Plasmalogens, which contain a vinyl ether bond at the sn-1 position, are preferential targets for hypochlorous acid (HOCl), generated by myeloperoxidase (MPO) from H(2)O(2) and chloride ions. Because MPO is implicated in neurodegeneration, this study pursued two aims: (i) to investigate the reactivity of mouse brain plasmalogens toward HOCl in vitro and (ii) to obtain in vivo evidence for MPO-mediated brain plasmalogen modification. Liquid chromatography coupled to hybrid linear ion trap-Fourier transform-ion cyclotron resonance mass spectrometry revealed plasmalogen modification in mouse brain lipid extracts at lower HOCl concentrations as observed for diacylphospholipids, resulting in the generation of 2-chloro fatty aldehydes and lysophospholipids. Lysophosphatidylethanolamine accumulation was transient, whereas lysophosphatidylcholine species containing saturated acyl residues remained stable. In vivo, a single, systemic endotoxin injection resulted in upregulation of cerebral MPO mRNA levels to a range comparable to that observed for tumor necrosis factor-α and cyclooxygenase-2. This inflammatory response was accompanied by a significant decrease in several brain plasmalogen species and concomitant in vivo generation of 2-chlorohexadecanal. The present findings demonstrate that activation of the MPO-H(2)O(2)-chloride system under neuroinflammatory conditions results in oxidative attack of the total cerebral plasmalogen pool. As this lipid class is indispensable for normal neuronal function, HOCl-mediated plasmalogen modification is likely to compromise normal synaptic transmission.

2.
Reduction of Ether-Type Glycerophospholipids, Plasmalogens, by NF-κB Signal Leading to Microglial Activation.

Hossain MS, Abe Y, Ali F, et al.

The Journal of Neuroscience : The Official Journal of the Society for Neuroscience. 2017;37(15):4074-4092. doi:10.1523/JNEUROSCI.3941-15.2017.

Neuroinflammation characterized by activation of glial cells is observed in various neurodegenerative diseases including Alzheimer's disease (AD). Although the reduction of ether-type glycerophospholipids, plasmalogens (Pls), in the brain is reported in AD patients, the mechanism of the reduction and its impact on neuroinflammation remained elusive. In the present study, we found for the first time that various inflammatory stimuli reduced Pls levels in murine glial cells via NF-κB activation, which then downregulated a Pls-synthesizing enzyme, glycerone phosphate O-acyltransferase (Gnpat) through increased c-Myc recruitment onto the promoter. We also found that systemic injection of lipopolysaccharide, aging, and chronic restraint stress reduced brain Pls contents that were associated with glial NF-κB activation, an increase in c-Myc expression, and downregulation of in the mouse cortex and hippocampus. More interestingly, the reduction of Pls contents in the murine cortex itself could increase the activated phenotype of microglial cells and the expression of proinflammatory cytokines, suggesting further acceleration of neuroinflammation by reduction of brain Pls. A similar mechanism of reduction was also found in human cell lines, triple-transgenic AD mouse brain, and postmortem human AD brain tissues. These findings suggest a novel mechanism of neuroinflammation that may explain prolonged progression of AD and help us to explore preventive and therapeutic strategies to treat neurodegenerative diseases. Ether-type glycerophospholipids, plasmalogens (Pls), are reduced in the brain of Alzheimer disease (AD) patients. We found that inflammatory stimuli reduced Pls contents by downregulation of the Pls-synthesizing enzyme glycerone phosphate O-acyltransferase (Gnpat) through NF-κB-mediated recruitment of c-Myc onto the promoter in both murine and human cell lines. Murine brains after systemic lipopolysaccharide, chronic stress, and aging, as well as triple-transgenic AD mice and postmortem human AD brain tissues all showed increased c-Myc and reduced expression. Interestingly, knockdown of itself activated NF-κB in glial cell lines and microglia in mouse cortex. Our findings provide a new insight into the mechanism of neuroinflammation and may help to develop a novel therapeutic approach for neurodegenerative diseases such as AD.

Expanded question: What impact do metals or preservatives in the blood have on the dysregulation of plasmalogen synthesis and homeostasis, and how might this contribute to neurodegenerative diseases such as Alzheimer's and Parkinson's disease?

Answered on April 11, 2025
Metals and preservatives in the blood can significantly impact the dysregulation of plasmalogen synthesis and homeostasis, contributing to neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD).
Metals:
1. Iron (Fe) and Copper (Cu): These redox-active metals can catalyze the formation of reactive oxygen species (ROS), leading to oxidative stress and lipid peroxidation, which disrupts plasmalogen synthesis and homeostasis. Elevated levels of Fe and Cu have been associated with increased oxidative damage and amyloid-beta aggregation in AD, as well as α-synuclein aggregation in PD.[1-4]
2. Manganese (Mn): Excess Mn can lead to mitochondrial dysfunction and oxidative stress, contributing to the dysregulation of plasmalogen synthesis. Mn accumulation has been linked to neurodegenerative processes in PD.[3-4]
3. Aluminum (Al): Al exposure has been implicated in the disruption of metal homeostasis and increased oxidative stress, which can impair plasmalogen synthesis and contribute to neurofibrillary tangle formation in AD.[3][5]
Preservatives:
1. Lipopolysaccharide (LPS): LPS, a bacterial endotoxin, can induce systemic inflammation and activate the NF-κB pathway, leading to the downregulation of plasmalogen-synthesizing enzymes such as glycerone phosphate O-acyltransferase (Gnpat). This results in reduced plasmalogen levels and exacerbates neuroinflammation, contributing to the progression of AD and PD.[6]
Mechanisms:
Oxidative Stress: Metals like Fe and Cu catalyze ROS production, leading to lipid peroxidation and plasmalogen degradation.[2-3]
Inflammation: LPS and other inflammatory stimuli activate NF-κB, reducing plasmalogen synthesis and increasing neuroinflammation.[6]
Protein Aggregation: Metals can induce conformational changes in amyloidogenic proteins, promoting aggregation and neurotoxicity.[3][5]
In summary, metals and preservatives in the blood can disrupt plasmalogen synthesis and homeostasis through oxidative stress, inflammation, and protein aggregation, contributing to the pathogenesis of neurodegenerative diseases such as Alzheimer's and Parkinson's disease.

1.
Common and Trace Metals in Alzheimer's and Parkinson's Diseases.

Doroszkiewicz J, Farhan JA, Mroczko J, et al.

International Journal of Molecular Sciences. 2023;24(21):15721. doi:10.3390/ijms242115721.

Trace elements and metals play critical roles in the normal functioning of the central nervous system (CNS), and their dysregulation has been implicated in neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD). In a healthy CNS, zinc, copper, iron, and manganese play vital roles as enzyme cofactors, supporting neurotransmission, cellular metabolism, and antioxidant defense. Imbalances in these trace elements can lead to oxidative stress, protein aggregation, and mitochondrial dysfunction, thereby contributing to neurodegeneration. In AD, copper and zinc imbalances are associated with amyloid-beta and tau pathology, impacting cognitive function. PD involves the disruption of iron and manganese levels, leading to oxidative damage and neuronal loss. Toxic metals, like lead and cadmium, impair synaptic transmission and exacerbate neuroinflammation, impacting CNS health. The role of aluminum in AD neurofibrillary tangle formation has also been noted. Understanding the roles of these elements in CNS health and disease might offer potential therapeutic targets for neurodegenerative disorders. The Codex Alimentarius standards concerning the mentioned metals in foods may be one of the key legal contributions to safeguarding public health. Further research is needed to fully comprehend these complex mechanisms and develop effective interventions.

2.
Metal-Catalyzed Disruption of Membrane Protein and Lipid Signaling in the Pathogenesis of Neurodegenerative Disorders.

Mattson MP.

Annals of the New York Academy of Sciences. 2004;1012:37-50. doi:10.1196/annals.1306.004.

Membrane lipid peroxidation and oxidative modification of various membrane and associated proteins (e.g., receptors, ion transporters and channels, and signal transduction and cytoskeletal proteins) occur in a range of neurodegenerative disorders. This membrane-associated oxidative stress (MAOS) is promoted by redox-active metals, most notably iron and copper. The mechanisms whereby different genetic and environmental factors initiate MAOS in specific neurological disorders are being elucidated. In Alzheimer's disease (AD), the amyloid beta-peptide generates reactive oxygen species and induces MAOS, resulting in disruption of cellular calcium homeostasis. In Parkinson's disease (PD), mitochondrial toxins and perturbed ubiquitin-dependent proteolysis may impair ATP production and increase oxyradical production and MAOS. The inheritance of polyglutamine-expanded huntingtin may promote neuronal degeneration in Huntington's disease (HD), in part, by increasing MAOS. Increased MAOS occurs in amyotrophic lateral sclerosis (ALS) as the result of genetic abnormalities (e.g., Cu/Zn-superoxide dismutase mutations) or exposure to environmental toxins. Levels of iron are increased in vulnerable neuronal populations in AD and PD, and dietary and pharmacological manipulations of iron and copper modify the course of the disease in mouse models of AD and PD in ways that suggest a role for these metals in disease pathogenesis. An increasing number of pharmacological and dietary interventions are being identified that can suppress MAOS and neuronal damage and improve functional outcome in animal models of AD, PD, HD, and ALS. Novel preventative and therapeutic approaches for neurodegenerative disorders are emerging from basic research on the molecular and cellular actions of metals and MAOS in neural cells.

3.
Dietary Trace Elements and the Pathogenesis of Neurodegenerative Diseases.

Kawahara M, Kato-Negishi M, Tanaka KI.

Nutrients. 2023;15(9):2067. doi:10.3390/nu15092067.

Trace elements such as iron (Fe), zinc (Zn), copper (Cu), and manganese (Mn) are absorbed from food via the gastrointestinal tract, transported into the brain, and play central roles in normal brain functions. An excess of these trace elements often produces reactive oxygen species and damages the brain. Moreover, increasing evidence suggests that the dyshomeostasis of these metals is involved in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease, prion diseases, and Lewy body diseases. The disease-related amyloidogenic proteins can regulate metal homeostasis at the synapses, and thus loss of the protective functions of these amyloidogenic proteins causes neurodegeneration. Meanwhile, metal-induced conformational changes of the amyloidogenic proteins contribute to enhancing their neurotoxicity. Moreover, excess Zn and Cu play central roles in the pathogenesis of vascular-type senile dementia. Here, we present an overview of the intake, absorption, and transport of four essential elements (Fe, Zn, Cu, Mn) and one non-essential element (aluminum: Al) in food and their connections with the pathogenesis of neurodegenerative diseases based on metal-protein, and metal-metal cross-talk.

4.
Metals Linked With the Most Prevalent Primary Neurodegenerative Dementias in the Elderly: A Narrative Review.

Mateo D, Marquès M, Torrente M.

Environmental Research. 2023;236(Pt 1):116722. doi:10.1016/j.envres.2023.116722.

Leading Journal

The ageing population has been steadily increasing worldwide, leading to a higher risk of cognitive decline and dementia. Environmental toxicants, particularly metals, have been identified as modifiable risk factors for cognitive impairment. Continuous exposure to metals occurs mainly through dietary sources, with older adults being particularly vulnerable. However, imbalances in the gut microbiota, known as dysbiosis, have also been associated with dementia. A literature review was conducted to explore the potential role of metals in the development of cognitive decline and the most prevalent primary neurodegenerative dementias, as well as their interaction with the gut microbiota. High levels of iron (Fe) and copper (Cu) are associated with mild cognitive impairment (MCI) and Alzheimer's disease (AD), while low selenium (Se) levels are linked to poor cognitive status. Parkinson's disease dementia (PDD) is associated with elevated levels of iron (Fe), manganese (Mn), and zinc (Zn), but the role of copper (Cu) remains unclear. The relationship between metals and Lewy body dementia (LBD) requires further investigation. High aluminium (Al) exposure is associated with frontotemporal dementia (FTD), and elevated selenium (Se) levels may be linked to its onset. Challenges in comparing studies arise from the heterogeneity of metal analysis matrices and analytical techniques, as well as the limitations of small study cohorts. More research is needed to understand the influence of metals on cognition through the gut microbiota (GMB) and its potential relevance in the development of these diseases.

5.
Environmental Pollutants as Risk Factors for Neurodegenerative Disorders: Alzheimer and Parkinson Diseases.

Chin-Chan M, Navarro-Yepes J, Quintanilla-Vega B.

Frontiers in Cellular Neuroscience. 2015;9:124. doi:10.3389/fncel.2015.00124.

Neurodegenerative diseases including Alzheimer (AD) and Parkinson (PD) have attracted attention in last decades due to their high incidence worldwide. The etiology of these diseases is still unclear; however the role of the environment as a putative risk factor has gained importance. More worryingly is the evidence that pre- and post-natal exposures to environmental factors predispose to the onset of neurodegenerative diseases in later life. Neurotoxic metals such as lead, mercury, aluminum, cadmium and arsenic, as well as some pesticides and metal-based nanoparticles have been involved in AD due to their ability to increase beta-amyloid (Aβ) peptide and the phosphorylation of Tau protein (P-Tau), causing senile/amyloid plaques and neurofibrillary tangles (NFTs) characteristic of AD. The exposure to lead, manganese, solvents and some pesticides has been related to hallmarks of PD such as mitochondrial dysfunction, alterations in metal homeostasis and aggregation of proteins such as α-synuclein (α-syn), which is a key constituent of Lewy bodies (LB), a crucial factor in PD pathogenesis. Common mechanisms of environmental pollutants to increase Aβ, P-Tau, α-syn and neuronal death have been reported, including the oxidative stress mainly involved in the increase of Aβ and α-syn, and the reduced activity/protein levels of Aβ degrading enzyme (IDE)s such as neprilysin or insulin IDE. In addition, epigenetic mechanisms by maternal nutrient supplementation and exposure to heavy metals and pesticides have been proposed to lead phenotypic diversity and susceptibility to neurodegenerative diseases. This review discusses data from epidemiological and experimental studies about the role of environmental factors in the development of idiopathic AD and PD, and their mechanisms of action.

6.
Reduction of Ether-Type Glycerophospholipids, Plasmalogens, by NF-κB Signal Leading to Microglial Activation.

Hossain MS, Abe Y, Ali F, et al.

The Journal of Neuroscience : The Official Journal of the Society for Neuroscience. 2017;37(15):4074-4092. doi:10.1523/JNEUROSCI.3941-15.2017.

Neuroinflammation characterized by activation of glial cells is observed in various neurodegenerative diseases including Alzheimer's disease (AD). Although the reduction of ether-type glycerophospholipids, plasmalogens (Pls), in the brain is reported in AD patients, the mechanism of the reduction and its impact on neuroinflammation remained elusive. In the present study, we found for the first time that various inflammatory stimuli reduced Pls levels in murine glial cells via NF-κB activation, which then downregulated a Pls-synthesizing enzyme, glycerone phosphate O-acyltransferase (Gnpat) through increased c-Myc recruitment onto the promoter. We also found that systemic injection of lipopolysaccharide, aging, and chronic restraint stress reduced brain Pls contents that were associated with glial NF-κB activation, an increase in c-Myc expression, and downregulation of in the mouse cortex and hippocampus. More interestingly, the reduction of Pls contents in the murine cortex itself could increase the activated phenotype of microglial cells and the expression of proinflammatory cytokines, suggesting further acceleration of neuroinflammation by reduction of brain Pls. A similar mechanism of reduction was also found in human cell lines, triple-transgenic AD mouse brain, and postmortem human AD brain tissues. These findings suggest a novel mechanism of neuroinflammation that may explain prolonged progression of AD and help us to explore preventive and therapeutic strategies to treat neurodegenerative diseases. Ether-type glycerophospholipids, plasmalogens (Pls), are reduced in the brain of Alzheimer disease (AD) patients. We found that inflammatory stimuli reduced Pls contents by downregulation of the Pls-synthesizing enzyme glycerone phosphate O-acyltransferase (Gnpat) through NF-κB-mediated recruitment of c-Myc onto the promoter in both murine and human cell lines. Murine brains after systemic lipopolysaccharide, chronic stress, and aging, as well as triple-transgenic AD mice and postmortem human AD brain tissues all showed increased c-Myc and reduced expression. Interestingly, knockdown of itself activated NF-κB in glial cell lines and microglia in mouse cortex. Our findings provide a new insight into the mechanism of neuroinflammation and may help to develop a novel therapeutic approach for neurodegenerative diseases such as AD.

Expanded question: Create a patient handout explaining how disruption of plasmalogen synthesis can lead to autism, and what are some of the toxicities and their mechanisms.

Answered on April 12, 2025
This patient handout will explain how the disruption of plasmalogen synthesis can lead to autism spectrum disorder (ASD) and outline some of the toxicities and their mechanisms. The information is grounded in current medical literature, which highlights the role of plasmalogens in brain function and development, and how their dysregulation can contribute to neurodevelopmental disorders like autism.[1-5]

Plasmalogen Disruption and Autism

What are Plasmalogens?
Plasmalogens are a special type of fat found in the brain and other parts of the body. They help protect cells from damage and are important for brain function and development.[1-2]
How Does Disruption of Plasmalogen Synthesis Lead to Autism?
When the body cannot make enough plasmalogens, it can affect brain development and function. This disruption can lead to problems with how brain cells communicate and protect themselves, which may contribute to autism spectrum disorder (ASD).[1-3]
Mechanisms of Disruption:
Oxidative Stress: Plasmalogens help protect brain cells from damage caused by harmful molecules called reactive oxygen species (ROS). Without enough plasmalogens, brain cells are more vulnerable to damage.[2-3]
Inflammation: Low levels of plasmalogens can lead to increased inflammation in the brain, which can affect brain development and function.[2][5]
Cell Signaling: Plasmalogens are involved in important signaling pathways in the brain. Disruption in their levels can interfere with these pathways, affecting brain function and development.[2-3]
Toxicities and Their Mechanisms:
Propionic Acid: This substance, produced by certain gut bacteria, can alter brain and blood plasmalogen levels. High levels of propionic acid have been linked to increased activity and changes in brain lipid composition, which may contribute to ASD.[5]
Environmental Toxins: Exposure to certain metals and preservatives can disrupt plasmalogen synthesis. For example, metals like iron and copper can cause oxidative stress, while lipopolysaccharides (LPS) from bacteria can increase inflammation, both of which can affect plasmalogen levels and brain function.[2][5]
Conclusion:
Disruption in plasmalogen synthesis can lead to problems in brain development and function, contributing to autism spectrum disorder. Understanding these mechanisms can help in developing potential treatments and preventive strategies.[1-5]

References

This patient handout provides a clear and concise explanation of how plasmalogen disruption can lead to autism and the mechanisms involved. It is grounded in current medical literature to ensure accuracy and reliability.

1.
Plasmalogen in the Brain: Effects on Cognitive Functions and Behaviors Attributable to Its Properties.

Udagawa J, Hino K.

Brain Research Bulletin. 2022;188:197-202. doi:10.1016/j.brainresbull.2022.08.008.

Ether phospholipid compositions are altered in the plasma or brain of patients with brain disorders, such as Alzheimer and Parkinson's disease, including those with psychiatric disorders like schizophrenia and bipolar disorders. Notably, plasmenyl ethanolamine has a unique chemical structure, i.e., a vinyl-ether bond at the sn-1 position, which mainly links with polyunsaturated fatty acids (PUFAs) at the sn-2 position. Those characteristic moieties give plasmalogen molecules unique biophysical and chemical properties that modulate membrane trafficking, lipid rafts, intramolecular PUFA moieties, and oxidative states. Previous reports suggested that a deficiency in plasmenyl ethanolamine leads to disturbances of the myelin structure, synaptic neurotransmission and intracellular signaling, apoptosis of neurons, and neuroinflammation, accompanied by cognitive disturbances and aberrant behaviors like hyperactivity in mice. Therefore, this review summarizes the relationship between the biological functions of plasmalogen. We also proposed biophysical properties that alter brain phospholipid compositions related to aberrant behaviors and cognitive dysfunction. Finally, a brief review of possible remedial plasmalogen replacement therapies for neurological, psychiatric, and developmental disorders attributable to disturbed plasmalogen compositions in the organs and cells was conducted.

2.
Plasmalogens, Platelet-Activating Factor and Beyond - Ether Lipids in Signaling and Neurodegeneration.

Dorninger F, Forss-Petter S, Wimmer I, Berger J.

Neurobiology of Disease. 2020;145:105061. doi:10.1016/j.nbd.2020.105061.

Glycerol-based ether lipids including ether phospholipids form a specialized branch of lipids that in mammals require peroxisomes for their biosynthesis. They are major components of biological membranes and one particular subgroup, the plasmalogens, is widely regarded as a cellular antioxidant. Their vast potential to influence signal transduction pathways is less well known. Here, we summarize the literature showing associations with essential signaling cascades for a wide variety of ether lipids, including platelet-activating factor, alkylglycerols, ether-linked lysophosphatidic acid and plasmalogen-derived polyunsaturated fatty acids. The available experimental evidence demonstrates links to several common players like protein kinase C, peroxisome proliferator-activated receptors or mitogen-activated protein kinases. Furthermore, ether lipid levels have repeatedly been connected to some of the most abundant neurological diseases, particularly Alzheimer's disease and more recently also neurodevelopmental disorders like autism. Thus, we critically discuss the potential role of these compounds in the etiology and pathophysiology of these diseases with an emphasis on signaling processes. Finally, we review the emerging interest in plasmalogens as treatment target in neurological diseases, assessing available data and highlighting future perspectives. Although many aspects of ether lipid involvement in cellular signaling identified in vitro still have to be confirmed in vivo, the compiled data show many intriguing properties and contributions of these lipids to health and disease that will trigger further research.

3.
Regulation of Plasmalogen Biosynthesis in Mammalian Cells and Tissues.

Honsho M, Fujiki Y.

Brain Research Bulletin. 2023;194:118-123. doi:10.1016/j.brainresbull.2023.01.011.

Plasmalogens are a unique family of cellular glycerophospholipids that contain a vinyl-ether bond. Synthesis of plasmalogens is initiated in peroxisomes and completed in the endoplasmic reticulum. The absence of plasmalogens in several organs of patients with deficiency in peroxisome biogenesis suggests that de novo synthesis of plasmalogens contributes significantly to plasmalogen homeostasis in humans. Plasmalogen biosynthesis is spatiotemporally regulated by a feedback mechanism that senses the amount of plasmalogens in the inner leaflet of the plasma membrane and regulates the stability of fatty acyl-CoA reductase 1 (FAR1), the rate-limiting enzyme for plasmalogen biosynthesis. Dysregulation of plasmalogen synthesis impairs cholesterol synthesis in cells and brain, resulting in the reduced expression of genes such as mRNA encoding myelin basic protein, a phenotype found in the cerebellum of plasmalogen-deficient mice. In this review, we summarize the current knowledge of molecular mechanisms underlying the regulation of plasmalogen biosynthesis and the link between plasmalogen homeostasis and cholesterol biosynthesis, and address the pathogenesis of impaired plasmalogen homeostasis in rodent and humans.

4.
Interactions Between the Lipidome and Genetic and Environmental Factors in Autism.

Yap CX, Henders AK, Alvares GA, et al.

Nature Medicine. 2023;29(4):936-949. doi:10.1038/s41591-023-02271-1.

Leading Journal

Autism omics research has historically been reductionist and diagnosis centric, with little attention paid to common co-occurring conditions (for example, sleep and feeding disorders) and the complex interplay between molecular profiles and neurodevelopment, genetics, environmental factors and health. Here we explored the plasma lipidome (783 lipid species) in 765 children (485 diagnosed with autism spectrum disorder (ASD)) within the Australian Autism Biobank. We identified lipids associated with ASD diagnosis (n = 8), sleep disturbances (n = 20) and cognitive function (n = 8) and found that long-chain polyunsaturated fatty acids may causally contribute to sleep disturbances mediated by the FADS gene cluster. We explored the interplay of environmental factors with neurodevelopment and the lipidome, finding that sleep disturbances and unhealthy diet have a convergent lipidome profile (with potential mediation by the microbiome) that is also independently associated with poorer adaptive function. In contrast, ASD lipidome differences were accounted for by dietary differences and sleep disturbances. We identified a large chr19p13.2 copy number variant genetic deletion spanning the LDLR gene and two high-confidence ASD genes (ELAVL3 and SMARCA4) in one child with an ASD diagnosis and widespread low-density lipoprotein-related lipidome derangements. Lipidomics captures the complexity of neurodevelopment, as well as the biological effects of conditions that commonly affect quality of life among autistic people.

5.
Leading Journal

Gastrointestinal symptoms and altered blood phospholipid profiles have been reported in patients with autism spectrum disorders (ASD). Most of the phospholipid analyses have been conducted on the fatty acid composition of isolated phospholipid classes following hydrolysis. A paucity of information exists on how the intact phospholipid molecular species are altered in ASD. We applied ESI/MS to determine how brain and blood intact phospholipid species were altered during the induction of ASD-like behaviors in rats following intraventricular infusions with the enteric bacterial metabolite propionic acid. Animals were infused daily for 8 days, locomotor activity assessed, and animals killed during the induced behaviors. Propionic acid infusions increased locomotor activity. Lipid analysis revealed treatment altered 21 brain and 30 blood phospholipid molecular species. Notable alterations were observed in the composition of brain SM, diacyl mono and polyunsaturated PC, PI, PS, PE, and plasmalogen PC and PE molecular species. These alterations suggest that the propionic acid rat model is a useful tool to study aberrations in lipid metabolism known to affect membrane fluidity, peroxisomal function, gap junction coupling capacity, signaling, and neuroinflammation, all of which may be associated with the pathogenesis of ASD.

Answered on April 12, 2025
The doctor has requested a more detailed explanation of specific toxicities and their mechanisms related to plasmalogen disruption and autism. The following document will elaborate on these aspects, grounded in the medical literature provided.[1-13]

Plasmalogen Disruption and Autism

Plasmalogens are a special type of fat found in the brain and other parts of the body. They help protect cells from damage and are important for brain function and development.[1-2]
How Does Disruption of Plasmalogen Synthesis Lead to Autism?
When the body cannot make enough plasmalogens, it can affect brain development and function. This disruption can lead to problems with how brain cells communicate and protect themselves, which may contribute to autism spectrum disorder (ASD).[1-3]
Mechanisms of Disruption:
Oxidative Stress: Plasmalogens help protect brain cells from damage caused by harmful molecules called reactive oxygen species (ROS). Without enough plasmalogens, brain cells are more vulnerable to damage.[2-3]
Inflammation: Low levels of plasmalogens can lead to increased inflammation in the brain, which can affect brain development and function.[2][5]
Cell Signaling: Plasmalogens are involved in important signaling pathways in the brain. Disruption in their levels can interfere with these pathways, affecting brain function and development.[2-3]
Specific Toxicities and Their Mechanisms:
Propionic Acid: This substance, produced by certain gut bacteria, can alter brain and blood plasmalogen levels. High levels of propionic acid have been linked to increased activity and changes in brain lipid composition, which may contribute to ASD.[5]
Hypochlorous Acid (HOCl): Generated by myeloperoxidase (MPO) from hydrogen peroxide and chloride ions, HOCl targets plasmalogens due to their vinyl ether bond, leading to oxidative modification and degradation. This process results in the generation of 2-chloro fatty aldehydes and lysophospholipids, compromising normal neuronal function and synaptic transmission.[13]
Lipopolysaccharide (LPS): LPS, a bacterial endotoxin, can induce systemic inflammation and activate the NF-κB pathway, leading to the downregulation of plasmalogen-synthesizing enzymes such as glycerone phosphate O-acyltransferase (Gnpat). This results in reduced plasmalogen levels and exacerbates neuroinflammation, contributing to the progression of ASD.[6]
Metals: Exposure to metals like iron (Fe) and copper (Cu) can catalyze the formation of ROS, leading to oxidative stress and lipid peroxidation, which disrupts plasmalogen synthesis and homeostasis. Elevated levels of Fe and Cu have been associated with increased oxidative damage and neuroinflammation.[2-4]
Conclusion:
Disruption in plasmalogen synthesis can lead to problems in brain development and function, contributing to autism spectrum disorder. Understanding these mechanisms can help in developing potential treatments and preventive strategies.[1-13]

References

The document now includes a detailed explanation of specific toxicities and their mechanisms related to plasmalogen disruption and autism. This should provide a comprehensive understanding for the intended audience.

1.
Plasmalogen in the Brain: Effects on Cognitive Functions and Behaviors Attributable to Its Properties.

Udagawa J, Hino K.

Brain Research Bulletin. 2022;188:197-202. doi:10.1016/j.brainresbull.2022.08.008.

Ether phospholipid compositions are altered in the plasma or brain of patients with brain disorders, such as Alzheimer and Parkinson's disease, including those with psychiatric disorders like schizophrenia and bipolar disorders. Notably, plasmenyl ethanolamine has a unique chemical structure, i.e., a vinyl-ether bond at the sn-1 position, which mainly links with polyunsaturated fatty acids (PUFAs) at the sn-2 position. Those characteristic moieties give plasmalogen molecules unique biophysical and chemical properties that modulate membrane trafficking, lipid rafts, intramolecular PUFA moieties, and oxidative states. Previous reports suggested that a deficiency in plasmenyl ethanolamine leads to disturbances of the myelin structure, synaptic neurotransmission and intracellular signaling, apoptosis of neurons, and neuroinflammation, accompanied by cognitive disturbances and aberrant behaviors like hyperactivity in mice. Therefore, this review summarizes the relationship between the biological functions of plasmalogen. We also proposed biophysical properties that alter brain phospholipid compositions related to aberrant behaviors and cognitive dysfunction. Finally, a brief review of possible remedial plasmalogen replacement therapies for neurological, psychiatric, and developmental disorders attributable to disturbed plasmalogen compositions in the organs and cells was conducted.

2.
Plasmalogens, Platelet-Activating Factor and Beyond - Ether Lipids in Signaling and Neurodegeneration.

Dorninger F, Forss-Petter S, Wimmer I, Berger J.

Neurobiology of Disease. 2020;145:105061. doi:10.1016/j.nbd.2020.105061.

Glycerol-based ether lipids including ether phospholipids form a specialized branch of lipids that in mammals require peroxisomes for their biosynthesis. They are major components of biological membranes and one particular subgroup, the plasmalogens, is widely regarded as a cellular antioxidant. Their vast potential to influence signal transduction pathways is less well known. Here, we summarize the literature showing associations with essential signaling cascades for a wide variety of ether lipids, including platelet-activating factor, alkylglycerols, ether-linked lysophosphatidic acid and plasmalogen-derived polyunsaturated fatty acids. The available experimental evidence demonstrates links to several common players like protein kinase C, peroxisome proliferator-activated receptors or mitogen-activated protein kinases. Furthermore, ether lipid levels have repeatedly been connected to some of the most abundant neurological diseases, particularly Alzheimer's disease and more recently also neurodevelopmental disorders like autism. Thus, we critically discuss the potential role of these compounds in the etiology and pathophysiology of these diseases with an emphasis on signaling processes. Finally, we review the emerging interest in plasmalogens as treatment target in neurological diseases, assessing available data and highlighting future perspectives. Although many aspects of ether lipid involvement in cellular signaling identified in vitro still have to be confirmed in vivo, the compiled data show many intriguing properties and contributions of these lipids to health and disease that will trigger further research.

3.
Regulation of Plasmalogen Biosynthesis in Mammalian Cells and Tissues.

Honsho M, Fujiki Y.

Brain Research Bulletin. 2023;194:118-123. doi:10.1016/j.brainresbull.2023.01.011.

Plasmalogens are a unique family of cellular glycerophospholipids that contain a vinyl-ether bond. Synthesis of plasmalogens is initiated in peroxisomes and completed in the endoplasmic reticulum. The absence of plasmalogens in several organs of patients with deficiency in peroxisome biogenesis suggests that de novo synthesis of plasmalogens contributes significantly to plasmalogen homeostasis in humans. Plasmalogen biosynthesis is spatiotemporally regulated by a feedback mechanism that senses the amount of plasmalogens in the inner leaflet of the plasma membrane and regulates the stability of fatty acyl-CoA reductase 1 (FAR1), the rate-limiting enzyme for plasmalogen biosynthesis. Dysregulation of plasmalogen synthesis impairs cholesterol synthesis in cells and brain, resulting in the reduced expression of genes such as mRNA encoding myelin basic protein, a phenotype found in the cerebellum of plasmalogen-deficient mice. In this review, we summarize the current knowledge of molecular mechanisms underlying the regulation of plasmalogen biosynthesis and the link between plasmalogen homeostasis and cholesterol biosynthesis, and address the pathogenesis of impaired plasmalogen homeostasis in rodent and humans.

4.
Interactions Between the Lipidome and Genetic and Environmental Factors in Autism.

Yap CX, Henders AK, Alvares GA, et al.

Nature Medicine. 2023;29(4):936-949. doi:10.1038/s41591-023-02271-1.

Autism omics research has historically been reductionist and diagnosis centric, with little attention paid to common co-occurring conditions (for example, sleep and feeding disorders) and the complex interplay between molecular profiles and neurodevelopment, genetics, environmental factors and health. Here we explored the plasma lipidome (783 lipid species) in 765 children (485 diagnosed with autism spectrum disorder (ASD)) within the Australian Autism Biobank. We identified lipids associated with ASD diagnosis (n = 8), sleep disturbances (n = 20) and cognitive function (n = 8) and found that long-chain polyunsaturated fatty acids may causally contribute to sleep disturbances mediated by the FADS gene cluster. We explored the interplay of environmental factors with neurodevelopment and the lipidome, finding that sleep disturbances and unhealthy diet have a convergent lipidome profile (with potential mediation by the microbiome) that is also independently associated with poorer adaptive function. In contrast, ASD lipidome differences were accounted for by dietary differences and sleep disturbances. We identified a large chr19p13.2 copy number variant genetic deletion spanning the LDLR gene and two high-confidence ASD genes (ELAVL3 and SMARCA4) in one child with an ASD diagnosis and widespread low-density lipoprotein-related lipidome derangements. Lipidomics captures the complexity of neurodevelopment, as well as the biological effects of conditions that commonly affect quality of life among autistic people.

5.

Gastrointestinal symptoms and altered blood phospholipid profiles have been reported in patients with autism spectrum disorders (ASD). Most of the phospholipid analyses have been conducted on the fatty acid composition of isolated phospholipid classes following hydrolysis. A paucity of information exists on how the intact phospholipid molecular species are altered in ASD. We applied ESI/MS to determine how brain and blood intact phospholipid species were altered during the induction of ASD-like behaviors in rats following intraventricular infusions with the enteric bacterial metabolite propionic acid. Animals were infused daily for 8 days, locomotor activity assessed, and animals killed during the induced behaviors. Propionic acid infusions increased locomotor activity. Lipid analysis revealed treatment altered 21 brain and 30 blood phospholipid molecular species. Notable alterations were observed in the composition of brain SM, diacyl mono and polyunsaturated PC, PI, PS, PE, and plasmalogen PC and PE molecular species. These alterations suggest that the propionic acid rat model is a useful tool to study aberrations in lipid metabolism known to affect membrane fluidity, peroxisomal function, gap junction coupling capacity, signaling, and neuroinflammation, all of which may be associated with the pathogenesis of ASD.

6.

Plasmalogens are a specific glycerophospholipid class containing a vinyl ether moiety at the sn-1-position of the glycerol backbone. The high susceptibility of this vinyl ether bond to oxidative damage and traces of acids may indicate the possible function of plasmalogens in biological systems: The regarded cell-internal antioxidative defense of membranes by protecting other phospholipids or lipoprotein particles against oxidative stress is controversial. Reactive oxygen species preferably affect the vinyl ether function as well as the olefinic acyl residues at the sn-2-position of plasmalogens. This review is dedicated to the role of plasmalogens in different cells and tissues as spermatozoal cells or brain tissue. The first chapter of this review will discuss the molecular structure and chemistry of plasmalogen molecules, their distributions in cells and tissues and the species-specificity. In the second chapter their functions as lipid mediators will be considered and the controversial antioxidative function will be discussed. The supposed function of plasmalogens as "scavengers" for reactive oxygen species (ROS) in biological membranes is challenged by the finding that plasmalogen oxidation products as alpha-hydroxyaldehydes and plasmalogen epoxides accumulate in all chronic diseases as atherosclerosis and myocardial infarction, upon aging as well as in Alzheimers disease and other neuropathological conditions. All these conditions, characterized by increased membrane instability and oxidative damage, will be reviewed in chapter three. Chronically proceeding processes can be described by permanently invading polymorphonuclear neutrophils into inflammatory loci. The degranulation of the azurophilic granula in polymorphonuclear leukocytes causes the release of highly reactive substances, for instance the myeloperoxidase-generated hypochlorous acid (HOCl) acting as effective oxidant. Therefore, special attention will be paid to neutrophil-derived HOCl. The last chapter deals with currently used methods of detecting plasmalogens and their degradation products. Although chromatographic methods will be also discussed, special attention will be given to (31)P NMR spectroscopy and soft ionization techniques of mass spectrometry as electrospray ionization or matrix-assisted laser desorption and ionization time-of-flight mass spectrometry.

7.
Advances in the Biosynthetic Pathways and Application Potential of Plasmalogens in Medicine.

Zhou Y, Yu N, Zhao J, et al.

Frontiers in Cell and Developmental Biology. 2020;8:765. doi:10.3389/fcell.2020.00765.

Plasmalogens are a special class of polar glycerolipids containing a vinyl-ether bond and an ester bond at sn-1 and sn-2 positions of the glycerol backbone, respectively. In animals, impaired biosynthesis and regulation of plasmalogens may lead to certain neurological and metabolic diseases. Plasmalogens deficiency was proposed to be strongly associated with neurodegenerative and metabolic diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), and appropriate supplement of plasmalogens could help to prevent and possibly provide therapy of these diseases. Plasmalogens evolved first in anaerobic bacteria with an anaerobic biosynthetic pathway. Later, an oxygen-dependent biosynthesis of plasmalogens appeared in animal cells. This review summarizes and updates current knowledge of anaerobic and aerobic pathways of plasmalogens biosynthesis, including the enzymes involved, steps and aspects of the regulation of these processes. Strategies for increasing the expression of plasmalogen synthetic genes using synthetic biology techniques under specific conditions are discussed. Deep understanding of plasmalogens biosynthesis will provide the bases for the use of plasmalogens and their precursors as potential therapeutic regimens for age-related degenerative and metabolic diseases.

8.
Plasmalogen Homeostasis - Regulation of Plasmalogen Biosynthesis and Its Physiological Consequence in Mammals.

Honsho M, Fujiki Y.

FEBS Letters. 2017;591(18):2720-2729. doi:10.1002/1873-3468.12743.

Plasmalogens, mostly ethanolamine-containing alkenyl ether phospholipids, are a major subclass of glycerophospholipids. Plasmalogen synthesis is initiated in peroxisomes and completed in the endoplasmic reticulum. The absence of plasmalogens in several organs of peroxisome biogenesis-defective patients suggests that the de novo synthesis of plasmalogens plays a pivotal role in its homeostasis in tissues. Plasmalogen synthesis is regulated by modulating the stability of fatty acyl-CoA reductase 1 on peroxisomal membranes, a rate-limiting enzyme in plasmalogen synthesis, by sensing plasmalogens in the inner leaflet of plasma membranes. Dysregulation of plasmalogen homeostasis impairs cholesterol biosynthesis by altering the stability of squalene monooxygenase, a key enzyme in cholesterol biosynthesis, implying physiological consequences of plasmalogen homeostasis with respect to cholesterol metabolism in cells, as well as in organs such as the liver.

9.
Plasmalogens and Platelet-Activating Factor Roles in Chronic Inflammatory Diseases.

Tremblay MÈ, Almsherqi ZA, Deng Y.

BioFactors (Oxford, England). 2022;48(6):1203-1216. doi:10.1002/biof.1916.

Fatty acids and phospholipid molecules are essential for determining the structure and function of cell membranes, and they hence participate in many biological processes. Platelet activating factor (PAF) and its precursor plasmalogen, which represent two subclasses of ether phospholipids, have attracted increasing research attention recently due to their association with multiple chronic inflammatory, neurodegenerative, and metabolic disorders. These pathophysiological conditions commonly involve inflammatory processes linked to an excess presence of PAF and/or decreased levels of plasmalogens. However, the molecular mechanisms underlying the roles of plasmalogens in inflammation have remained largely elusive. While anti-inflammatory responses most likely involve the plasmalogen signal pathway; pro-inflammatory responses recruit arachidonic acid, a precursor of pro-inflammatory lipid mediators which is released from membrane phospholipids, notably derived from the hydrolysis of plasmalogens. Plasmalogens per se are vital membrane phospholipids in humans. Changes in their homeostatic levels may alter cell membrane properties, thus affecting key signaling pathways that mediate inflammatory cascades and immune responses. The plasmalogen analogs of PAF are also potentially important, considering that anti-PAF activity has strong anti-inflammatory effects. Plasmalogen replacement therapy was further identified as a promising anti-inflammatory strategy allowing for the relief of pathological hallmarks in patients affected by chronic diseases with an inflammatory component. The aim of this Short Review is to highlight the emerging roles and implications of plasmalogens in chronic inflammatory disorders, along with the promising outcomes of plasmalogen replacement therapy for the treatment of various PAF-related chronic inflammatory pathologies.

10.
Plasmalogens Participate in Very-Long-Chain Fatty Acid-Induced Pathology.

Brites P, Mooyer PA, El Mrabet L, Waterham HR, Wanders RJ.

Brain : A Journal of Neurology. 2009;132(Pt 2):482-92. doi:10.1093/brain/awn295.

Leading Journal

Peroxisomes are organelles responsible for multiple metabolic pathways including, the biosynthesis of plasmalogens, a class of phospholipids, and the beta-oxidation of very-long-chain fatty acids (VLCFA). Lack of peroxisomes or dysfunction in any of their normal functions is the cellular basis for human peroxisomal disorders. Here we used mouse models to understand and define the biochemical and cellular determinants that mediate the pathophysiological consequences caused by peroxisomal dysfunctions. We investigated the role and effects of cellular plasmalogens and VLCFA accumulation in liver, testis and nervous tissue using Pex7 and Abcd1 knockout (KO) mice. In addition, we also generated a Pex7:Abcd1 double KO mouse to investigate how different peroxisomal dysfunctions modulate cellular function and pathology. We found that plasmalogens function as fundamental structural phospholipids and protect cells from damage caused by VLCFA accumulation. In testis, plasmalogens protect spermatocytes from VLCFA-induced degeneration and apoptosis. In nervous tissue, we found that gliosis, inflammatory demyelination and axonopathy caused by accumulation of VLCFA are modulated by plasmalogens. Our findings demonstrate the importance of normal peroxisomal functioning and allow the understanding of the pathological causality of peroxisomal dysfunctions. Nervous tissue deficient in plasmalogens is more prone to damage, illustrating the importance of plasmalogens in peroxisomal disorders including Zellweger syndrome and X-linked adrenoleukodystrophy.

11.
Leukodystrophy Caused by Plasmalogen Deficiency Rescued by Glyceryl 1-Myristyl Ether Treatment.

Malheiro AR, Correia B, Ferreira da Silva T, et al.

Brain Pathology (Zurich, Switzerland). 2019;29(5):622-639. doi:10.1111/bpa.12710.

Plasmalogens are the most abundant form of ether phospholipids in myelin and their deficiency causes Rhizomelic Chondrodysplasia Punctata (RCDP), a severe developmental disorder. Using the Gnpat-knockout (KO) mouse as a model of RCDP, we determined the consequences of a plasmalogen deficiency during myelination and myelin homeostasis in the central nervous system (CNS). We unraveled that the lack of plasmalogens causes a generalized hypomyelination in several CNS regions including the optic nerve, corpus callosum and spinal cord. The defect in myelin content evolved to a progressive demyelination concomitant with generalized astrocytosis and white matter-selective microgliosis. Oligodendrocyte precursor cells (OPC) and mature oligodendrocytes were abundant in the CNS of Gnpat KO mice during the active period of demyelination. Axonal loss was minimal in plasmalogen-deficient mice, although axonal damage was observed in spinal cords from aged Gnpat KO mice. Characterization of the plasmalogen-deficient myelin identified myelin basic protein and septin 7 as early markers of dysmyelination, whereas myelin-associated glycoprotein was associated with the active demyelination phase. Using in vitro myelination assays, we unraveled that the intrinsic capacity of oligodendrocytes to ensheath and initiate membrane wrapping requires plasmalogens. The defect in plasmalogens was rescued with glyceryl 1-myristyl ether [1-O-tetradecyl glycerol (1-O-TDG)], a novel alternative precursor in the plasmalogen biosynthesis pathway. 1-O-TDG treatment rescued myelination in plasmalogen-deficient oligodendrocytes and in mutant mice. Our results demonstrate the importance of plasmalogens for oligodendrocyte function and myelin assembly, and identified a novel strategy to promote myelination in nervous tissue.

12.
Regulation of Plasmalogen Metabolism and Traffic in Mammals: The Fog Begins to Lift.

Dorninger F, Werner ER, Berger J, Watschinger K.

Frontiers in Cell and Developmental Biology. 2022;10:946393. doi:10.3389/fcell.2022.946393.

Due to their unique chemical structure, plasmalogens do not only exhibit distinct biophysical and biochemical features, but require specialized pathways of biosynthesis and metabolization. Recently, major advances have been made in our understanding of these processes, for example by the attribution of the gene encoding the enzyme, which catalyzes the final desaturation step in plasmalogen biosynthesis, or by the identification of cytochrome C as plasmalogenase, which allows for the degradation of plasmalogens. Also, models have been presented that plausibly explain the maintenance of adequate cellular levels of plasmalogens. However, despite the progress, many aspects around the questions of how plasmalogen metabolism is regulated and how plasmalogens are distributed among organs and tissues in more complex organisms like mammals, remain unresolved. Here, we summarize and interpret current evidence on the regulation of the enzymes involved in plasmalogen biosynthesis and degradation as well as the turnover of plasmalogens. Finally, we focus on plasmalogen traffic across the mammalian body - a topic of major importance, when considering plasmalogen replacement therapies in human disorders, where deficiencies in these lipids have been reported. These involve not only inborn errors in plasmalogen metabolism, but also more common diseases including Alzheimer's disease and neurodevelopmental disorders.

13.
Mouse Brain Plasmalogens Are Targets for Hypochlorous Acid-Mediated Modification in Vitro and in Vivo.

Ullen A, Fauler G, Köfeler H, et al.

Free Radical Biology & Medicine. 2010;49(11):1655-65. doi:10.1016/j.freeradbiomed.2010.08.025.

Leading Journal

Plasmalogens, 1-O-alk-1'-enyl-2-acyl-sn-glycerophospholipids, are significant constituents of cellular membranes and are essential for normal brain development. Plasmalogens, which contain a vinyl ether bond at the sn-1 position, are preferential targets for hypochlorous acid (HOCl), generated by myeloperoxidase (MPO) from H(2)O(2) and chloride ions. Because MPO is implicated in neurodegeneration, this study pursued two aims: (i) to investigate the reactivity of mouse brain plasmalogens toward HOCl in vitro and (ii) to obtain in vivo evidence for MPO-mediated brain plasmalogen modification. Liquid chromatography coupled to hybrid linear ion trap-Fourier transform-ion cyclotron resonance mass spectrometry revealed plasmalogen modification in mouse brain lipid extracts at lower HOCl concentrations as observed for diacylphospholipids, resulting in the generation of 2-chloro fatty aldehydes and lysophospholipids. Lysophosphatidylethanolamine accumulation was transient, whereas lysophosphatidylcholine species containing saturated acyl residues remained stable. In vivo, a single, systemic endotoxin injection resulted in upregulation of cerebral MPO mRNA levels to a range comparable to that observed for tumor necrosis factor-α and cyclooxygenase-2. This inflammatory response was accompanied by a significant decrease in several brain plasmalogen species and concomitant in vivo generation of 2-chlorohexadecanal. The present findings demonstrate that activation of the MPO-H(2)O(2)-chloride system under neuroinflammatory conditions results in oxidative attack of the total cerebral plasmalogen pool. As this lipid class is indispensable for normal neuronal function, HOCl-mediated plasmalogen modification is likely to compromise normal synaptic transmission.

Expanded question: In the patient handout, elaborate on the other metals and preservatives we discussed above.

Answered on April 12, 2025
The following patient handout elaborates on the specific toxicities and their mechanisms related to plasmalogen disruption and autism, as discussed in the previous conversation. This information is grounded in current medical literature to ensure accuracy and reliability.

Plasmalogen Disruption and Autism

Plasmalogens are a special type of fat found in the brain and other parts of the body. They help protect cells from damage and are important for brain function and development.[1-2]
How Does Disruption of Plasmalogen Synthesis Lead to Autism?
When the body cannot make enough plasmalogens, it can affect brain development and function. This disruption can lead to problems with how brain cells communicate and protect themselves, which may contribute to autism spectrum disorder (ASD).[1-3]
Mechanisms of Disruption:
Oxidative Stress: Plasmalogens help protect brain cells from damage caused by harmful molecules called reactive oxygen species (ROS). Without enough plasmalogens, brain cells are more vulnerable to damage.[2-3]
Inflammation: Low levels of plasmalogens can lead to increased inflammation in the brain, which can affect brain development and function.[2][4]
Cell Signaling: Plasmalogens are involved in important signaling pathways in the brain. Disruption in their levels can interfere with these pathways, affecting brain function and development.[2-3]
Specific Toxicities and Their Mechanisms:
Propionic Acid: This substance, produced by certain gut bacteria, can alter brain and blood plasmalogen levels. High levels of propionic acid have been linked to increased activity and changes in brain lipid composition, which may contribute to ASD.[4]
Hypochlorous Acid (HOCl): Generated by myeloperoxidase (MPO) from hydrogen peroxide and chloride ions, HOCl targets plasmalogens due to their vinyl ether bond, leading to oxidative modification and degradation. This process results in the generation of 2-chloro fatty aldehydes and lysophospholipids, compromising normal neuronal function and synaptic transmission.[5]
Lipopolysaccharide (LPS): LPS, a bacterial endotoxin, can induce systemic inflammation and activate the NF-κB pathway, leading to the downregulation of plasmalogen-synthesizing enzymes such as glycerone phosphate O-acyltransferase (Gnpat). This results in reduced plasmalogen levels and exacerbates neuroinflammation, contributing to the progression of ASD.[6]
Metals:
Iron (Fe) and Copper (Cu): These redox-active metals can catalyze the formation of ROS, leading to oxidative stress and lipid peroxidation, which disrupts plasmalogen synthesis and homeostasis. Elevated levels of Fe and Cu have been associated with increased oxidative damage and neuroinflammation.[2-3][7]
Manganese (Mn): Excess Mn can lead to mitochondrial dysfunction and oxidative stress, contributing to the dysregulation of plasmalogen synthesis. Mn accumulation has been linked to neurodegenerative processes in PD.[3][7]
Aluminum (Al): Al exposure has been implicated in the disruption of metal homeostasis and increased oxidative stress, which can impair plasmalogen synthesis and contribute to neurofibrillary tangle formation in AD.[3][7]
Mercury (Hg) and Cadmium (Cd): These metals can induce rigidity in cell membranes and disrupt plasmalogen synthesis. Hg, in particular, can cleave the vinyl ether bond in plasmalogens, leading to increased membrane rigidity and potential cell damage.[8-9]
Lead (Pb) and Arsenic (As): These metals can lower glutathione levels and inhibit methionine synthase activity, which is crucial for methylation reactions in the brain. This inhibition can contribute to neurodevelopmental disorders like ASD.[10]
Conclusion:
Disruption in plasmalogen synthesis can lead to problems in brain development and function, contributing to autism spectrum disorder. Understanding these mechanisms can help in developing potential treatments and preventive strategies.[1-3][7][6][8-10]

References

This patient handout now includes a detailed explanation of specific toxicities and their mechanisms related to plasmalogen disruption and autism. This should provide a comprehensive understanding for the intended audience.

1.
Plasmalogen in the Brain: Effects on Cognitive Functions and Behaviors Attributable to Its Properties.

Udagawa J, Hino K.

Brain Research Bulletin. 2022;188:197-202. doi:10.1016/j.brainresbull.2022.08.008.

Ether phospholipid compositions are altered in the plasma or brain of patients with brain disorders, such as Alzheimer and Parkinson's disease, including those with psychiatric disorders like schizophrenia and bipolar disorders. Notably, plasmenyl ethanolamine has a unique chemical structure, i.e., a vinyl-ether bond at the sn-1 position, which mainly links with polyunsaturated fatty acids (PUFAs) at the sn-2 position. Those characteristic moieties give plasmalogen molecules unique biophysical and chemical properties that modulate membrane trafficking, lipid rafts, intramolecular PUFA moieties, and oxidative states. Previous reports suggested that a deficiency in plasmenyl ethanolamine leads to disturbances of the myelin structure, synaptic neurotransmission and intracellular signaling, apoptosis of neurons, and neuroinflammation, accompanied by cognitive disturbances and aberrant behaviors like hyperactivity in mice. Therefore, this review summarizes the relationship between the biological functions of plasmalogen. We also proposed biophysical properties that alter brain phospholipid compositions related to aberrant behaviors and cognitive dysfunction. Finally, a brief review of possible remedial plasmalogen replacement therapies for neurological, psychiatric, and developmental disorders attributable to disturbed plasmalogen compositions in the organs and cells was conducted.

2.
Plasmalogens, Platelet-Activating Factor and Beyond - Ether Lipids in Signaling and Neurodegeneration.

Dorninger F, Forss-Petter S, Wimmer I, Berger J.

Neurobiology of Disease. 2020;145:105061. doi:10.1016/j.nbd.2020.105061.

Glycerol-based ether lipids including ether phospholipids form a specialized branch of lipids that in mammals require peroxisomes for their biosynthesis. They are major components of biological membranes and one particular subgroup, the plasmalogens, is widely regarded as a cellular antioxidant. Their vast potential to influence signal transduction pathways is less well known. Here, we summarize the literature showing associations with essential signaling cascades for a wide variety of ether lipids, including platelet-activating factor, alkylglycerols, ether-linked lysophosphatidic acid and plasmalogen-derived polyunsaturated fatty acids. The available experimental evidence demonstrates links to several common players like protein kinase C, peroxisome proliferator-activated receptors or mitogen-activated protein kinases. Furthermore, ether lipid levels have repeatedly been connected to some of the most abundant neurological diseases, particularly Alzheimer's disease and more recently also neurodevelopmental disorders like autism. Thus, we critically discuss the potential role of these compounds in the etiology and pathophysiology of these diseases with an emphasis on signaling processes. Finally, we review the emerging interest in plasmalogens as treatment target in neurological diseases, assessing available data and highlighting future perspectives. Although many aspects of ether lipid involvement in cellular signaling identified in vitro still have to be confirmed in vivo, the compiled data show many intriguing properties and contributions of these lipids to health and disease that will trigger further research.

3.
Regulation of Plasmalogen Biosynthesis in Mammalian Cells and Tissues.

Honsho M, Fujiki Y.

Brain Research Bulletin. 2023;194:118-123. doi:10.1016/j.brainresbull.2023.01.011.

Plasmalogens are a unique family of cellular glycerophospholipids that contain a vinyl-ether bond. Synthesis of plasmalogens is initiated in peroxisomes and completed in the endoplasmic reticulum. The absence of plasmalogens in several organs of patients with deficiency in peroxisome biogenesis suggests that de novo synthesis of plasmalogens contributes significantly to plasmalogen homeostasis in humans. Plasmalogen biosynthesis is spatiotemporally regulated by a feedback mechanism that senses the amount of plasmalogens in the inner leaflet of the plasma membrane and regulates the stability of fatty acyl-CoA reductase 1 (FAR1), the rate-limiting enzyme for plasmalogen biosynthesis. Dysregulation of plasmalogen synthesis impairs cholesterol synthesis in cells and brain, resulting in the reduced expression of genes such as mRNA encoding myelin basic protein, a phenotype found in the cerebellum of plasmalogen-deficient mice. In this review, we summarize the current knowledge of molecular mechanisms underlying the regulation of plasmalogen biosynthesis and the link between plasmalogen homeostasis and cholesterol biosynthesis, and address the pathogenesis of impaired plasmalogen homeostasis in rodent and humans.

4.

Gastrointestinal symptoms and altered blood phospholipid profiles have been reported in patients with autism spectrum disorders (ASD). Most of the phospholipid analyses have been conducted on the fatty acid composition of isolated phospholipid classes following hydrolysis. A paucity of information exists on how the intact phospholipid molecular species are altered in ASD. We applied ESI/MS to determine how brain and blood intact phospholipid species were altered during the induction of ASD-like behaviors in rats following intraventricular infusions with the enteric bacterial metabolite propionic acid. Animals were infused daily for 8 days, locomotor activity assessed, and animals killed during the induced behaviors. Propionic acid infusions increased locomotor activity. Lipid analysis revealed treatment altered 21 brain and 30 blood phospholipid molecular species. Notable alterations were observed in the composition of brain SM, diacyl mono and polyunsaturated PC, PI, PS, PE, and plasmalogen PC and PE molecular species. These alterations suggest that the propionic acid rat model is a useful tool to study aberrations in lipid metabolism known to affect membrane fluidity, peroxisomal function, gap junction coupling capacity, signaling, and neuroinflammation, all of which may be associated with the pathogenesis of ASD.

5.
Mouse Brain Plasmalogens Are Targets for Hypochlorous Acid-Mediated Modification in Vitro and in Vivo.

Ullen A, Fauler G, Köfeler H, et al.

Free Radical Biology & Medicine. 2010;49(11):1655-65. doi:10.1016/j.freeradbiomed.2010.08.025.

Plasmalogens, 1-O-alk-1'-enyl-2-acyl-sn-glycerophospholipids, are significant constituents of cellular membranes and are essential for normal brain development. Plasmalogens, which contain a vinyl ether bond at the sn-1 position, are preferential targets for hypochlorous acid (HOCl), generated by myeloperoxidase (MPO) from H(2)O(2) and chloride ions. Because MPO is implicated in neurodegeneration, this study pursued two aims: (i) to investigate the reactivity of mouse brain plasmalogens toward HOCl in vitro and (ii) to obtain in vivo evidence for MPO-mediated brain plasmalogen modification. Liquid chromatography coupled to hybrid linear ion trap-Fourier transform-ion cyclotron resonance mass spectrometry revealed plasmalogen modification in mouse brain lipid extracts at lower HOCl concentrations as observed for diacylphospholipids, resulting in the generation of 2-chloro fatty aldehydes and lysophospholipids. Lysophosphatidylethanolamine accumulation was transient, whereas lysophosphatidylcholine species containing saturated acyl residues remained stable. In vivo, a single, systemic endotoxin injection resulted in upregulation of cerebral MPO mRNA levels to a range comparable to that observed for tumor necrosis factor-α and cyclooxygenase-2. This inflammatory response was accompanied by a significant decrease in several brain plasmalogen species and concomitant in vivo generation of 2-chlorohexadecanal. The present findings demonstrate that activation of the MPO-H(2)O(2)-chloride system under neuroinflammatory conditions results in oxidative attack of the total cerebral plasmalogen pool. As this lipid class is indispensable for normal neuronal function, HOCl-mediated plasmalogen modification is likely to compromise normal synaptic transmission.

6.

Plasmalogens are a specific glycerophospholipid class containing a vinyl ether moiety at the sn-1-position of the glycerol backbone. The high susceptibility of this vinyl ether bond to oxidative damage and traces of acids may indicate the possible function of plasmalogens in biological systems: The regarded cell-internal antioxidative defense of membranes by protecting other phospholipids or lipoprotein particles against oxidative stress is controversial. Reactive oxygen species preferably affect the vinyl ether function as well as the olefinic acyl residues at the sn-2-position of plasmalogens. This review is dedicated to the role of plasmalogens in different cells and tissues as spermatozoal cells or brain tissue. The first chapter of this review will discuss the molecular structure and chemistry of plasmalogen molecules, their distributions in cells and tissues and the species-specificity. In the second chapter their functions as lipid mediators will be considered and the controversial antioxidative function will be discussed. The supposed function of plasmalogens as "scavengers" for reactive oxygen species (ROS) in biological membranes is challenged by the finding that plasmalogen oxidation products as alpha-hydroxyaldehydes and plasmalogen epoxides accumulate in all chronic diseases as atherosclerosis and myocardial infarction, upon aging as well as in Alzheimers disease and other neuropathological conditions. All these conditions, characterized by increased membrane instability and oxidative damage, will be reviewed in chapter three. Chronically proceeding processes can be described by permanently invading polymorphonuclear neutrophils into inflammatory loci. The degranulation of the azurophilic granula in polymorphonuclear leukocytes causes the release of highly reactive substances, for instance the myeloperoxidase-generated hypochlorous acid (HOCl) acting as effective oxidant. Therefore, special attention will be paid to neutrophil-derived HOCl. The last chapter deals with currently used methods of detecting plasmalogens and their degradation products. Although chromatographic methods will be also discussed, special attention will be given to (31)P NMR spectroscopy and soft ionization techniques of mass spectrometry as electrospray ionization or matrix-assisted laser desorption and ionization time-of-flight mass spectrometry.

7.
Interactions Between the Lipidome and Genetic and Environmental Factors in Autism.

Yap CX, Henders AK, Alvares GA, et al.

Nature Medicine. 2023;29(4):936-949. doi:10.1038/s41591-023-02271-1.

Autism omics research has historically been reductionist and diagnosis centric, with little attention paid to common co-occurring conditions (for example, sleep and feeding disorders) and the complex interplay between molecular profiles and neurodevelopment, genetics, environmental factors and health. Here we explored the plasma lipidome (783 lipid species) in 765 children (485 diagnosed with autism spectrum disorder (ASD)) within the Australian Autism Biobank. We identified lipids associated with ASD diagnosis (n = 8), sleep disturbances (n = 20) and cognitive function (n = 8) and found that long-chain polyunsaturated fatty acids may causally contribute to sleep disturbances mediated by the FADS gene cluster. We explored the interplay of environmental factors with neurodevelopment and the lipidome, finding that sleep disturbances and unhealthy diet have a convergent lipidome profile (with potential mediation by the microbiome) that is also independently associated with poorer adaptive function. In contrast, ASD lipidome differences were accounted for by dietary differences and sleep disturbances. We identified a large chr19p13.2 copy number variant genetic deletion spanning the LDLR gene and two high-confidence ASD genes (ELAVL3 and SMARCA4) in one child with an ASD diagnosis and widespread low-density lipoprotein-related lipidome derangements. Lipidomics captures the complexity of neurodevelopment, as well as the biological effects of conditions that commonly affect quality of life among autistic people.

8.
Inorganic Mercury and Cadmium Induce Rigidity in Eukaryotic Lipid Extracts While Mercury Also Ruptures Red Blood Cells.

Kerek E, Hassanin M, Prenner EJ.

Biochimica Et Biophysica Acta. Biomembranes. 2018;1860(3):710-717. doi:10.1016/j.bbamem.2017.12.014.

Hg and Cd are non-essential toxic heavy metals that bioaccumulate in the tissues of living systems but less is known about their interactions with Eukaryotic lipid bilayers. Microscopy experiments showed that Hg and Cd changed the cell morphology of rabbit erythrocytes while Hg also induced cell rupture. As membranes are one of the first available targets, our study aimed to better understand metal-lipid interactions that could lead to toxic effects. Fluorescence spectroscopy (Laurdan Generalized Polarization) and dynamic light scattering were used to analyze metal-induced changes in membrane fluidity and the size of liposomes composed of Brain (Porcine), Liver (Bovine), Heart (Bovine) and Yeast (S. cerevisiae) lipid extracts. Under physiological chloride and pH levels, Hg irreversibly cleaves plasmalogens resulting in an increase in membrane rigidity. These lipids are enriched in Brain, Heart and Erythrocyte membranes and are important in signalling and the protection against oxidative stress. Interestingly, Hg had a heavily reduced effect on the plasmalogen-free Yeast extract membrane. In contrast, Cd induced rigidity by targeting negatively charged phosphatidic acid, phosphatidylserine, phosphatidylinositol, phosphatidylglycerol and cardiolipin in these extracts. Metal-induced liposome aggregation depended on the proportion of negatively charged lipids/plasmalogen and even the order of metal addition. Our results show that data from model systems correlate with trends observed in complex biological extracts and red blood cells and serve as a predictive tool for analyzing metal-lipid interactions. The determination of the specific lipid targets for Hg and Cd provides new insights how these metals exert toxic effects on cell membranes.

9.
Preferential Binding of Inorganic Mercury to Specific Lipid Classes and Its Competition With Cadmium.

Kerek E, Hassanin M, Zhang W, Prenner EJ.

Biochimica Et Biophysica Acta. Biomembranes. 2017;1859(7):1211-1221. doi:10.1016/j.bbamem.2017.03.022.

Upon uptake of Hg and Cd into living systems, possible targets for metal induced toxicity include the membranes surrounding nervous, cardiovascular and renal cells. To further our understanding of the interactions of Hg and Cd with different lipid structures under physiologically relevant chloride and pH conditions (100 mM NaCl pH 7.4), we used fluorescence spectroscopy and dynamic light scattering to monitor changes in membrane fluidity and phase transition and liposome size. The metal effects were studied on zwitterionic, cationic and anionic lipids to elucidate electrostatically driven metal-lipid interactions. The effect of Hg-catalyzed cleavage of the vinyl ether bond in plasmalogens on these aforementioned properties was studied in addition to a thermodynamic characterization of this interaction by Isothermal Titration Calorimetry. The negatively charged Hg-chloride complexes formed under our experimental conditions induce membrane rigidity in membranes containing cationic lipids and plasmalogens while this effect is heavily reduced and entirely absent with zwitterionic and anionic lipids respectively. The K for the interaction of Hg with plasmalogen containing liposomes was between 4-30 μM. Furthermore, the presence of Cd affected the interaction of Hg with plasmalogen when negatively charged PS was also present. In this case, even the order of the metal addition was important.

10.
Alternatively Spliced Methionine Synthase in SH-SY5Y Neuroblastoma Cells: Cobalamin and GSH Dependence and Inhibitory Effects of Neurotoxic Metals and Thimerosal.

Waly M, Power-Charnitsky VA, Hodgson N, et al.

Oxidative Medicine and Cellular Longevity. 2016;2016:6143753. doi:10.1155/2016/6143753.

Leading Journal

The folate and cobalamin (Cbl-) dependent enzyme methionine synthase (MS) is highly sensitive to oxidation and its activity affects all methylation reactions. Recent studies have revealed alternative splicing of MS mRNA in human brain and patient-derived fibroblasts. Here we show that MS mRNA in SH-SY5Y human neuroblastoma cells is alternatively spliced, resulting in three primary protein species, thus providing a useful model to examine cofactor dependence of these variant enzymes. MS activity was dependent upon methylcobalamin (MeCbl) or the combination of hydroxocobalamin (OHCbl) and S-adenosylmethionine (SAM). OHCbl-based activity was eliminated by depletion of the antioxidant glutathione (GSH) but could be rescued by provision of either glutathionylcobalamin (GSCbl) or MeCbl. Pretreatment of cells with lead, arsenic, aluminum, mercury, or the ethylmercury-containing preservative thimerosal lowered GSH levels and inhibited MS activity in association with decreased uptake of cysteine, which is rate-limiting for GSH synthesis. Thimerosal treatment decreased cellular levels of GSCbl and MeCbl. These findings indicate that the alternatively spliced form of MS expressed in SH-SY5Y human neuronal cells is sensitive to inhibition by thimerosal and neurotoxic metals, and lower GSH levels contribute to their inhibitory action.