Lysophospholipids for Neurodegenerative Disorders: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , ,

Neurodegenerative diseases (NDs) commonly present misfolded and aggregated proteins. Considerable research has been performed to unearth the molecular processes underpinning this pathological aggregation and develop therapeutic strategies targeting NDs. Fibrillary deposits of α-synuclein (α-Syn), a highly conserved and thermostable protein, are a critical feature in the development of NDs such as Alzheimer’s disease (AD), Lewy body disease (LBD), Parkinson’s disease (PD), and multiple system atrophy (MSA). Inhibition of α-Syn aggregation can thus serve as a potential approach for therapeutic intervention. The degradation of target proteins by small molecules has emerged as a new therapeutic modality, gaining the hotspot in pharmaceutical research. Additionally, interest is growing in the use of food-derived bioactive compounds as intervention agents against NDs via functional foods and dietary supplements. According to reports, dietary bioactive phospholipids may have cognition-enhancing and neuroprotective effects, owing to their abilities to influence cognition and mental health in vivo and in vitro. 

  • lysophospholipids
  • neurodegenerative diseases

1. Overview

Neurodegeneration has been identified as the pathophysiological hallmark in most brain-related disorders. Many neurodegenerative diseases (NDs) involve the misfolding and aggregation of specific proteins into abnormal, toxic species [1][2]. Early diagnosis is essential for treatment planning and ensuring that the right support can be provided to patients and their families. However, therapeutic strategies for NDs present unique challenges for drug development. Although no curative treatment is available for NDs, the range of therapeutic and supportive options are expanding. The most common NDs are Alzheimer’s disease (AD), Lewy body disease (LBD), Parkinson’s disease (PD), and multiple system atrophy (MSA) [3]. The observation of amyloid-like protein aggregation in the brains of patients with NDs was first described in the 1960s, and thus far, three-dimensional structures of several of these aggregates have been elucidated [4][5]. These structural data have been utilized for the design of inhibitors of amyloid-like aggregation proteins [6][7][8]. Since decades, the presence of amyloids in NDs has been associated exclusively with pathologies [9][10]. Additionally, strong evidence indicates that the activation of inflammatory processes is a hallmark of NDs. Elevated levels of pro-inflammatory cytokines and chemokines, as well as activated microglia and astrocytes, are found in the brains of patients with AD, even at very early stages of the disease [11]. Ageing affects homeostatic processes that protect against protein misfolding and is associated with an increase in oxidative stress, neuroinflammation, and mitochondrial-lysosomal dysfunction, which have been shown to directly activate microglia cells and astrocytes [12][13][14][15]. It has been argued that extracellular amyloid-β peptide (Aβ) in AD activates microglia and astrocytes, which in turn release tumor necrosis factor alpha (TNF-α) and other cytokines. TNF-α signaling enhances Aβ production and Aβ-induced neuroinflammation [16]. Neuroinflammation, oxidative stress, and mitochondrial dysfunction have all been linked to the progression of NDs [11][17][18]. The neuroprotective effects of resveratrol reportedly stem from its ability to inhibit microglial activation and regulate neuroinflammation [19][20][21]. The researchers' recent study demonstrated that porcine liver decomposition product (PLDP) could improve cognitive function in elderly adults by providing a rich source of phospholipids (PLs) and lysophospholipids (LPLs) [22]. Notably, the main constituent lipids of synaptic vesicles include cholesterol and phospholipids [23]. Although the concentration of PLDP-derived lipids in LPLs is not sufficient to yield cognitive benefits, LPLs may confer anti-neuroinflammatory effects [24][25]. Previous research suggests that LPLs have served not only as structural components of biological membranes but also as biologically active molecules [26]. LPLs influence a plethora of processes, including neurogenesis in the central nervous system (CNS) [27][28][29]. Growing interest in the involvement of extracellular LPLs in the pathology of NDs increasingly indicates that these small molecules may have therapeutic potential for NDs.

2. Effects of LPLs on Cognitive Decline

2.1. Dietary Sources of LPLs

Because of their superior emulsification properties, LPLs have numerous applications in the food, cosmetic, and pharmaceutical industries [30]. However, their properties depend strongly on the fatty acid component present and the specific polar head bound to the glycerol backbone [31]. LPLs, as promising feed additives, have been widely used to supplement farm animals diets to improve growth performance, feed efficiency, and dietary fat absorption [32]. LPL supplementation can increase the apparent total tract digestibility of animals [33]. PLs are widely available in the intestinal lumen after eating, and their hydrolysis is catalyzed by phospholipase A2 (PLA2). LPLs (particularly LPC), the digestive products of PLs, have direct roles in mediating chylomicron assembly and secretion [34]. LPC is present in the plasma circulation at relatively high levels and includes species containing both saturated and unsaturated fatty acids [35]. Interestingly, the intake of PL-enriched diets during postnatal brain development was found to increase the number of striatal circuits [36]. In PD, dopaminergic neurons are progressively degenerated, leading to striatal dopamine depletion and movement deficits. α-Syn is intimately involved in the pathogenesis of PD, and has been implicated in the regulation of dopamine synthesis, release, and reuptake [37]. It is possible that dietary PLs have several benefits in health, including improvements in cognition across the lifespan.

2.2. Cognitive Function and LPLs

Mild cognitive impairment (MCI) is characterized by impairment of certain cognitive functions, as defined by Petersen et al. [38], and a diagnosis of mild neurocognitive disorder essentially comprises MCI [39]. It can occur in a subtle form, such as MCI, in the early stages of PD in up to 25% of newly diagnosed patients [40][41][42]. MCI is a distinct stage of cognitive loss that falls between the expected cognitive decline of physiological aging and the more serious loss of mental abilities associated with dementia. It is characterized by impairments in memory, language, thinking, or judgment that are severe enough to interfere with daily life [43]. Individuals with MCI have an increased risk of developing dementia caused by AD or other neurological conditions [44]. Growing evidence supports the hypothesis that dietary factors may play a role in healthy aging, including exertion of a protective effect against age-related cognitive decline [45][46]. The concept of functional foods was first introduced in Japan, a country with a long history of using foods for their health benefits [47]. The market for functional food ingredients covers the sale of functional food ingredients containing bioactive compounds and ingredients used in manufacturing functional food products [48]. The ingredients in functional foods provide health benefits, and some of them include supplements or other additives. Functional foods and bioactive compounds can strongly intensify the therapeutic efficacy of certain drugs by influencing different pathways [49]. A new understanding of these interactions is emerging owing to intense research on nutraceuticals in diseases, including NDs [50]. For example, Yuyama et al. showed that glucosylceramides (GlcCer) from konjac extract are linked to the attenuation of amyloid-like protein in the CNS [51]. In the brains of GlcCer-treated mice, decreased levels of several inflammatory cytokines were accompanied by the recovery of impaired synaptic densities [52]. Future studies need to evaluate GlcCer and examine their relationship with α-Syn in the brain to determine the possible use of plasma GlcCer as a prognostic biomarker of disease progression. Several dietary components including PLs, plasmalogens, ω3 fatty acid, carotenoids, vitamins, and phenolic compounds have been identified to affect cognitive functions [53]. Many studies indicate that neuroinflammation plays a fundamental role in the progression of the neuropathological changes that characterize NDs [54][55][56]; microglia are the main cellular effectors of this process [57]. Indeed, neuroinflammation is initiated by microglia, which are resident immune cells of the CNS. Once activated, microglia can synthesize and release several neurotrophic factors and antioxidants to withstand the pathological progression of NDs [58].

2.3. PLDP and LPLs

In previous studies, over 50% of patients with PD without dementia demonstrated cognitive alterations and 20% primarily exhibited memory deficits [59]. The researchers previously reported that PLDP induces a significant increase in the Hasegawa’s dementia scale-revised (HDS-R) score and the Wechsler memory scale (WMS) score in a randomized, double-blind, placebo-controlled study in healthy humans [22]. PLDP was recently approved as a food with functional claims (FFC) in Japan. FFC was introduced in April 2015 for increasing the availability of products that are clearly labeled with certain health functions [60]. Oral administration of PLDP has been shown to enhance visual memory and delayed recall in healthy adults [22]. Notably, the oral route is the most common and preferred method of drug administration for several reasons, such as non-invasiveness, patient compliance, and convenience of drug administration [61]. These results suggest that PLDP may help improve brain function. Interestingly, PLDP is a rich source of PLs, and composition analysis of PLDP revealed that the most abundant PLs and LPLs belonged to the phosphatidylcholine (PC) and phosphatidylethanolamine (PE) classes [25][62]. LPLs influence signaling, proliferation, neural activity, and inflammation to mediate various important processes, including the pathogenesis of cerebral ischemia, vascular dementia, and AD [25][63][64][65]. However, no direct evidence indicates that any PLDP component improves cognitive function, and the effect of PLDP components on cognitive function remains unclear. The researchers' recent study identified novel cooperative actions of LPLs that inhibit IL-6 expression and the accumulation of intracellular ROS in microglia after lipopolysaccharide (LPS)-induced neuroinflammation [25]. This activation was significantly inhibited by lysophosphatidylcholine (LPC) and lysophosphatidylethanolamine (LPE), suggesting that these molecules exert significant protective effects against LPS-induced inflammation (Figure 1). This finding is important considering that microglia-mediated neuroinflammation is regarded as a pathological mechanism in many NDs, such as AD and LBD, and is a pivotal event accelerating cognitive or functional decline. Furthermore, dietary LPC and docosahexaenoic acid (DHA) have been reported to efficiently increase DHA levels in the brain, improving brain function in adult mice [66]. This report describes a novel nutraceutical approach for preventing and treating neurological diseases (such as AD) associated with DHA deficiency.
Figure 1. PLDP is a rich source of phospholipids. PLDP extracted lipids (PEL) was extracted from PLDP using the Bligh and Dyer method. This PEL is a rich source of LPLs, including LPC and LPE. LPC and LPE exerted significant protective effects against LPS-induced inflammation and oxidative stress in microglial cells. Various isoforms of PLA2 enzyme hydrolyze PC and PE at the sn-2 position to form LPLs, including LPC and LPE, respectively. α-Syn is bound to LPLs, which are known to be contained in PEL, strongly inhibit α-Syn aggregation.

This entry is adapted from the peer-reviewed paper 10.3390/biomedicines10123126

References

  1. Bouvier-Müller, A.; Ducongé, F. Nucleic acid aptamers for neurodegenerative diseases. Biochimie 2018, 145, 73–83.
  2. Calabrese, G.; Molzahn, C.; Mayor, T. Protein interaction networks in neurodegenerative diseases: From physiological function to aggregation. J. Biol. Chem. 2022, 298, 102062.
  3. Moussaud, S.; Jones, D.R.; Moussaud-Lamodière, E.L.; Delenclos, M.; Ross, O.A.; McLean, P.J. Alpha-synuclein and tau: Teammates in neurodegeneration? Mol. Neurodegener. 2014, 9, 43.
  4. Cohen, A.S.; Calkins, E. Electron Microscopic Observations on a Fibrous Component in Amyloid of Diverse Origins. Nature 1959, 183, 1202–1203.
  5. Merker, H.-J.; Shibolet, S.; Sohar, E.; Gafni, J.; Heller, H. Periodic Cross-banding in Amyloid Filaments. Nature 1966, 211, 1401–1402.
  6. Seidler, P.M.; Boyer, D.R.; Rodriguez, J.A.; Sawaya, M.R.; Cascio, D.; Murray, K.; Gonen, T.; Eisenberg, D.S. Structure-based inhibitors of tau aggregation. Nat. Chem. 2018, 10, 170–176.
  7. Griner, S.L.; Seidler, P.; Bowler, J.; A Murray, K.; Yang, T.P.; Sahay, S.; Sawaya, M.R.; Cascio, D.; A Rodriguez, J.; Philipp, S.; et al. Structure-based inhibitors of amyloid beta core suggest a common interface with tau. eLife 2019, 8, e46924.
  8. Balasco, N.; Diaferia, C.; Morelli, G.; Vitagliano, L.; Accardo, A. Amyloid-Like Aggregation in Diseases and Biomaterials: Osmosis of Structural Information. Front. Bioeng. Biotechnol. 2021, 9, 641372.
  9. Soto, C.; Pritzkow, S. Protein misfolding, aggregation, and conformational strains in neurodegenerative diseases. Nat. Neurosci. 2018, 21, 1332–1340.
  10. Wells, C.; Brennan, S.; Keon, M.; Ooi, L. The role of amyloid oligomers in neurodegenerative pathologies. Int. J. Biol. Macromol. 2021, 181, 582–604.
  11. Currais, A.; Fischer, W.; Maher, P.; Schubert, D. Intraneuronal protein aggregation as a trigger for inflammation and neurodegeneration in the aging brain. FASEB J. 2017, 31, 5–10.
  12. Van Den Berge, N.; Ferreira, N.; Mikkelsen, T.W.; Alstrup, A.K.O.; Tamgüney, G.; Karlsson, P.; Terkelsen, A.J.; Nyengaard, J.R.; Jensen, P.H.; Borghammer, P. Ageing promotes pathological alpha-synuclein propagation and autonomic dysfunction in wild-type rats. Brain 2021, 144, 1853–1868.
  13. Su, X.; Maguire-Zeiss, K.A.; Giuliano, R.; Prifti, L.; Venkatesh, K.; Federoff, H.J. Synuclein activates microglia in a model of Parkinson’s disease. Neurobiol. Aging 2008, 29, 1690–1701.
  14. Zhang, W.; Wang, T.; Pei, Z.; Miller, D.S.; Wu, X.; Block, M.L.; Wilson, B.; Zhang, W.; Zhou, Y.; Hong, J.-S.; et al. Aggregated α-synuclein activates microglia: A process leading to disease progression in Parkinson’s disease. FASEB J. 2005, 19, 533–542.
  15. Chou, T.-W.; Chang, N.P.; Krishnagiri, M.; Patel, A.P.; Lindman, M.; Angel, J.P.; Kung, P.-L.; Atkins, C.; Daniels, B.P. Fibrillar α-synuclein induces neurotoxic astrocyte activation via RIP kinase signaling and NF-κB. Cell Death Dis. 2021, 12, 756.
  16. Minter, M.R.; Taylor, J.M.; Crack, P.J. The contribution of neuroinflammation to amyloid toxicity in Alzheimer’s disease. J. Neurochem. 2016, 136, 457–474.
  17. Tsukahara, T.; Sahara, Y.; Ribeiro, N.; Tsukahara, R.; Gotoh, M.; Sakamoto, S.; Handa, H.; Murakami-Murofushi, K. Adenine nucleotide translocase 2, a putative target protein for 2-carba cyclic phosphatidic acid in microglial cells. Cell. Signal. 2021, 82, 109951.
  18. Swerdlow, R.H. Mitochondria and Mitochondrial Cascades in Alzheimer’s Disease. J. Alzheimer’s Dis. 2018, 62, 1403–1416.
  19. Capiralla, H.; Vingtdeux, V.; Zhao, H.; Sankowski, R.; Al-Abed, Y.; Davies, P.; Marambaud, P. Resveratrol mitigates lipopolysaccharide- and Aβ-mediated microglial inflammation by inhibiting the TLR4/NF-κB/STAT signaling cascade. J. Neurochem. 2012, 120, 461–472.
  20. Qi, Y.; Shang, L.; Liao, Z.; Su, H.; Jing, H.; Wu, B.; Bi, K.; Jia, Y. Intracerebroventricular injection of resveratrol ameliorated Aβ-induced learning and cognitive decline in mice. Metab. Brain Dis. 2019, 34, 257–266.
  21. Huang, J.; Huang, N.; Xu, S.; Luo, Y.; Li, Y.; Jin, H.; Yu, C.; Shi, J.; Jin, F. Signaling mechanisms underlying inhibition of neuroinflammation by resveratrol in neurodegenerative diseases. J. Nutr. Biochem. 2021, 88, 108552.
  22. Matsuda, Y.; Haniu, H.; Tsukahara, T.; Uemura, T.; Inoue, T.; Sako, K.-I.; Kojima, J.; Mori, T.; Sato, K. Oral administration of porcine liver decomposition product for 4 weeks enhances visual memory and delayed recall in healthy adults over 40 years of age: A randomized, double-blind, placebo-controlled study. Exp. Gerontol. 2020, 141, 111064.
  23. Takamori, S.; Holt, M.; Stenius, K.; Lemke, E.A.; Grønborg, M.; Riedel, D.; Urlaub, H.; Schenck, S.; Brügger, B.; Ringler, P.; et al. Molecular Anatomy of a Trafficking Organelle. Cell 2006, 127, 831–846.
  24. Tsukahara, T.; Haniu, H.; Uemura, T.; Matsuda, Y. Therapeutic Potential of Porcine Liver Decomposition Product: New Insights and Perspectives for Microglia-Mediated Neuroinflammation in Neurodegenerative Diseases. Biomedicines 2020, 8, 446.
  25. Tsukahara, T.; Hara, H.; Haniu, H.; Matsuda, Y. The Combined Effects of Lysophospholipids against Lipopolysaccharide-induced Inflammation and Oxidative Stress in Microglial Cells. J. Oleo Sci. 2021, 70, 947–954.
  26. Grzelczyk, A.; Gendaszewska-Darmach, E. Novel bioactive glycerol-based lysophospholipids: New data—New insight into their function. Biochimie 2013, 95, 667–679.
  27. Hao, Y.; Guo, M.; Feng, Y.; Dong, Q.; Cui, M. Lysophospholipids and Their G-Coupled Protein Signaling in Alzheimer’s Disease: From Physiological Performance to Pathological Impairment. Front. Mol. Neurosci. 2020, 13, 58.
  28. Rosell-Valle, C.; Pedraza, C.; Manuel, I.; Moreno-Rodríguez, M.; Rodríguez-Puertas, R.; Castilla-Ortega, E.; Caramés, J.M.; Conde, A.I.G.; Zambrana-Infantes, E.; Ortega-Pinazo, J.; et al. Chronic central modulation of LPA/LPA receptors-signaling pathway in the mouse brain regulates cognition, emotion, and hippocampal neurogenesis. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2021, 108, 110156.
  29. Birgbauer, E. Lysophosphatidic Acid Signalling in Nervous System Development and Function. NeuroMolecular Med. 2021, 23, 68–85.
  30. Shah, A.K.M.A.; Nagao, T.; Kurihara, H.; Takahashi, K. Production of a Health-Beneficial Food Emulsifier by Enzymatic Partial Hydrolysis of Phospholipids Obtained from the Head of Autumn Chum Salmon. J. Oleo Sci. 2017, 66, 147–155.
  31. D’Arrigo, P.; Servi, S. Synthesis of Lysophospholipids. Molecules 2010, 15, 1354–1377.
  32. Zhang, M.; Bai, H.; Zhao, Y.; Wang, R.; Li, G.; Zhang, G.; Zhang, Y. Effects of Dietary Lysophospholipid Inclusion on the Growth Performance, Nutrient Digestibility, Nitrogen Utilization, and Blood Metabolites of Finishing Beef Cattle. Antioxidants 2022, 11, 1486.
  33. Zhao, P.; Li, H.; Hossain, M.; Kim, I. Effect of emulsifier (lysophospholipids) on growth performance, nutrient digestibility and blood profile in weanling pigs. Anim. Feed Sci. Technol. 2015, 207, 190–195.
  34. Hui, D.Y. Intestinal phospholipid and lysophospholipid metabolism in cardiometabolic disease. Curr. Opin. Infect. Dis. 2016, 27, 507–512.
  35. Takatera, A.; Takeuchi, A.; Saiki, K.; Morisawa, T.; Yokoyama, N.; Matsuo, M. Quantification of lysophosphatidylcholines and phosphatidylcholines using liquid chromatography–tandem mass spectrometry in neonatal serum. J. Chromatogr. B 2006, 838, 31–36.
  36. Guillermo, R.B.; Yang, P.; Vickers, M.H.; McJarrow, P.; Guan, J. Supplementation with complex milk lipids during brain development promotes neuroplasticity without altering myelination or vascular density. Food Nutr. Res. 2015, 59, 25765.
  37. Senior, S.L.; Ninkina, N.; Deacon, R.; Bannerman, D.; Buchman, V.L.; Cragg, S.J.; Wade-Martins, R. Increased striatal dopamine release and hyperdopaminergic-like behaviour in mice lacking both alpha-synuclein and gamma-synuclein. Eur. J. Neurosci. 2008, 27, 947–957.
  38. Petersen, R.C.; Smith, G.E.; Waring, S.C.; Ivnik, R.J.; Tangalos, E.G.; Kokmen, E. Mild Cognitive Impairment: Clinical characterization and outcome. Arch. Neurol. 1999, 56, 303–308.
  39. Petersen, R.C. Early Diagnosis of Alzheimers Disease: Is MCI Too Late? Curr. Alzheimer Res. 2009, 6, 324–330.
  40. Foltynie, T.; Brayne, C.R.; Robbins, T.W.; Barker, R.A. The cognitive ability of an incident cohort of Parkinson’s patients in the UK. The CamPaIGN study. Brain 2004, 127, 550–560.
  41. Muslimović, D.; Post, B.; Speelman, J.D.; Schmand, B. Cognitive profile of patients with newly diagnosed Parkinson disease. Neurology 2005, 65, 1239–1245.
  42. Hassin-Baer, S.; Cohen, O.S.; Israeli-Korn, S.; Yahalom, G.; Benizri, S.; Sand, D.; Issachar, G.; Geva, A.B.; Shani-Hershkovich, R.; Peremen, Z. Identification of an early-stage Parkinson’s disease neuromarker using event-related potentials, brain network analytics and machine-learning. PLoS ONE 2022, 17, e0261947.
  43. Facal, D.; Spuch, C.; Valladares-Rodriguez, S. New Trends in Cognitive Aging and Mild Cognitive Impairment. Geriatrics 2022, 7, 80.
  44. Campbell, N.L.; Unverzagt, F.; LaMantia, M.A.; Khan, B.A.; Boustani, M.A. Risk Factors for the Progression of Mild Cognitive Impairment to Dementia. Clin. Geriatr. Med. 2013, 29, 873–893.
  45. Godos, J.; Currenti, W.; Angelino, D.; Mena, P.; Castellano, S.; Caraci, F.; Galvano, F.; Del Rio, D.; Ferri, R.; Grosso, G. Diet and Mental Health: Review of the Recent Updates on Molecular Mechanisms. Antioxidants 2020, 9, 346.
  46. Caruso, G.; Godos, J.; Privitera, A.; Lanza, G.; Castellano, S.; Chillemi, A.; Bruni, O.; Ferri, R.; Caraci, F.; Grosso, G. Phenolic Acids and Prevention of Cognitive Decline: Polyphenols with a Neuroprotective Role in Cognitive Disorders and Alzheimer’s Disease. Nutrients 2022, 14, 819.
  47. Guiné, R.P.F.; Florença, S.G.; Barroca, M.J.; Anjos, O. The Link between the Consumer and the Innovations in Food Product Development. Foods 2020, 9, 1317.
  48. Gómez-Pinilla, F. Brain foods: The effects of nutrients on brain function. Nat. Rev. Neurosci. 2008, 9, 568–578.
  49. Chianese, R.; Coccurello, R.; Viggiano, A.; Scafuro, M.; Fiore, M.; Coppola, G.; Operto, F.F.; Fasano, S.; Laye, S.; Pierantoni, R.; et al. Impact of Dietary Fats on Brain Functions. Curr. Neuropharmacol. 2018, 16, 1059–1085.
  50. Dadhania, V.P.; Trivedi, P.P.; Vikram, A.; Tripathi, D.N. Nutraceuticals against Neurodegeneration: A Mechanistic Insight. Curr. Neuropharmacol. 2016, 14, 627–640.
  51. Yuyama, K.; Igarashi, Y. Linking glycosphingolipids to Alzheimer’s amyloid-ß: Extracellular vesicles and functional plant materials. Glycoconj. J. 2022, 39, 613–618.
  52. Yuyama, K.; Takahashi, K.; Usuki, S.; Mikami, D.; Sun, H.; Hanamatsu, H.; Furukawa, J.; Mukai, K.; Igarashi, Y. Plant sphingolipids promote extracellular vesicle release and alleviate amyloid-β pathologies in a mouse model of Alzheimer’s disease. Sci. Rep. 2019, 9, 16827.
  53. Ozawa, H.; Miyazawa, T.; Miyazawa, T. Effects of Dietary Food Components on Cognitive Functions in Older Adults. Nutrients 2021, 13, 2804.
  54. Kinney, J.W.; BeMiller, S.M.; Murtishaw, A.S.; Leisgang, A.M.; Salazar, A.M.; Lamb, B.T. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimer’s Dement. 2018, 4, 575–590.
  55. Garcia, P.; Jürgens-Wemheuer, W.; Huarte, O.U.; Michelucci, A.; Masuch, A.; Brioschi, S.; Weihofen, A.; Koncina, E.; Coowar, D.; Heurtaux, T.; et al. Neurodegeneration and neuroinflammation are linked, but independent of alpha-synuclein inclusions, in a seeding/spreading mouse model of Parkinson’s disease. Glia 2022, 70, 935–960.
  56. Hammond, T.R.; Marsh, S.E.; Stevens, B. Immune Signaling in Neurodegeneration. Immunity 2019, 50, 955–974.
  57. Crotti, A.; Ransohoff, R.M. Microglial Physiology and Pathophysiology: Insights from Genome-wide Transcriptional Profiling. Immunity 2016, 44, 505–515.
  58. Guzman-Martinez, L.; Maccioni, R.B.; Andrade, V.; Navarrete, L.P.; Pastor, M.G.; Ramos-Escobar, N. Neuroinflammation as a Common Feature of Neurodegenerative Disorders. Front. Pharmacol. 2019, 10, 1008.
  59. Janvin, C.C.; Larsen, J.P.; Aarsland, D.; Hugdahl, K. Subtypes of mild cognitive impairment in parkinson’s disease: Progression to dementia. Mov. Disord. 2006, 21, 1343–1349.
  60. Kamioka, H.; Tsutani, K.; Origasa, H.; Yoshizaki, T.; Kitayuguchi, J.; Shimada, M.; Wada, Y.; Takano-Ohmuro, H. Quality of Systematic Reviews of the Foods with Function Claims in Japan: Comparative Before- and After-Evaluation of Verification Reports by the Consumer Affairs Agency. Nutrients 2019, 11, 1583.
  61. Alqahtani, M.S.; Kazi, M.; Alsenaidy, M.A.; Ahmad, M.Z. Advances in Oral Drug Delivery. Front. Pharmacol. 2021, 12, 618411.
  62. Tsukahara, T.; Haniu, H.; Uemura, T.; Matsuda, Y. Porcine liver decomposition product-derived lysophospholipids promote microglial activation in vitro. Sci. Rep. 2020, 10, 3748.
  63. Gotoh, M.; Sano-Maeda, K.; Murofushi, H.; Murakami-Murofushi, K. Protection of Neuroblastoma Neuro2A Cells from Hypoxia-Induced Apoptosis by Cyclic Phosphatidic Acid (cPA). PLoS ONE 2012, 7, e51093.
  64. Sheikh, A.M.; Michikawa, M.; Kim, S.; Nagai, A. Lysophosphatidylcholine increases the neurotoxicity of Alzheimer’s amyloid β1-42 peptide: Role of oligomer formation. Neuroscience 2015, 292, 159–169.
  65. Mulder, C.; Wahlund, L.-O.; Teerlink, T.; Blomberg, M.; Veerhuis, R.; Van Kamp, G.J.; Scheltens, P.; Scheffer, P.G. Decreased lysophosphatidylcholine/phosphatidylcholine ratio in cerebrospinal fluid in Alzheimer’s disease. J. Neural Transm. 2003, 110, 949–955.
  66. Sugasini, D.; Thomas, R.; Yalagala, P.C.R.; Tai, L.M.; Subbaiah, P.V. Dietary docosahexaenoic acid (DHA) as lysophosphatidylcholine, but not as free acid, enriches brain DHA and improves memory in adult mice. Sci. Rep. 2017, 7, 11263.
More
This entry is offline, you can click here to edit this entry!
Video Production Service