Main Metabolic Pathways with Respect to Alzheimer’s Disease: Comparison
Please note this is a comparison between Version 2 by Jessie Wu and Version 3 by Jessie Wu.

Alzheimer’s disease (AD) is an aging-related neurodegenerative disease, leading to the progressive loss of memory and other cognitive functions. Metabolomics allows the study of biochemical alterations in pathological processes which may be involved in AD progression and to discover new therapeutic targets. Metabolites are substrates, intermediates, and products of metabolic body processes, which typically are small molecules with a molecular weight of less than ~1.5 kDa. Since low molecular weight metabolites are intermediates or end products of cellular metabolism, metabolomics, or the study of metabolism can be considered one of the core disciplines of systems biology. It can help in improving our understanding of changes in biochemical pathways, revealing crucial information that is closely related to human disease or therapeutic status.

  • Alzheimer’s disease
  • metabolomics
  • biomarkers

1. Arginine Metabolism

L-arginine is a semi-essential amino acid that can be metabolized to form a number of bioactive molecules [1] (Figure 1). It is synthesized from proline or glutamate, with the ultimate synthetic step catalyzed by argininosuccinate lyase [2]. L-arginine can be metabolized by arginases, nitric oxide synthases (NOS), and possibly also by arginine decarboxylase (ADC), resulting ultimately in the production of agmatine, ornithine, nitric oxide (NO), or urea [2]. The expression of several of these enzymes can be regulated at transcriptional and translational levels by changes in the concentration of L-arginine itself [3].
Figure 1. Arginine metabolic pathways. L-arginine can be metabolized by phosphatidic acid (PA), nitric oxide synthase (NOS), arginase, and arginine decarboxylase (ADC) to form several bioactive molecules. (ADC, Arginine decarboxylase; ADMA, NG-dimethyl-L-arginine; DDAH, dimethylarginine dimethylaminohydrolase; GABA, γ-aminobutyric acid; ODC, ornithine decarboxylase)
L-ornithine is the arginase-mediated metabolite of L-arginine, with urea as the by-product. L-ornithine can be further metabolized to form putrescine, spermidine, and spermine polyamine, which are essential for normal cell growth and functioning, or via a separate pathway to form glutamine and cell-signaling molecule, GABA [1]. Previous research has reported decreased glutamate and GABA levels in Alzheimer’s disease (AD) brains and increased glutamine synthase (GS) levels in the lumbar cerebrospinal fluid of AD patients [4][5]. In peripheral organs and also CNS, arginine can also be metabolized by ADC to produce agmatine, a neurotransmitter that plays an important role in the learning and memory process [6].
NO is a gaseous signaling molecule produced by NOS. NO, derived from neuronal NOS (nNOS), plays an important role in synaptic plasticity and learning, and memory [7][8][9]. Moreover, L-arginine and NO affect the cardiovascular system as endogenous antiatherogenic molecules that protect the endothelium, modulate vasodilatation, and interact with the vascular wall and circulating blood cells [10][11][12][13][14].

2. Alanine, Aspartate, and Glutamate Metabolism

Glutamate is the principal excitatory neurotransmitter of the brain [15]. Most neurons and glia are likely to be influenced by glutamate since they have receptors for glutamate. Glutamate is considered the main neurotransmitter of neocortical and hippocampal pyramidal neurons and is involved in higher mental functions such as cognition and memory [16]. Disturbance of excitatory glutamatergic neurotransmission is believed to be associated with many neurological disorders, including AD [16], ischemic brain damage [17], and motor neuron disease [18].
Glutamate receptors can be divided into two classes: ionotropic (N-methyl-D-aspartate, NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/kainite subtypes and metabotropic [19]. The role of glutamate and glutamate receptors in learning and memory is widely recognized. For instance, NMDA antagonists impair learning and memory while NMDA agonists and facilitators improve memory [16]; likewise, AMPAKines (positive modulators of receptor function) facilitate learning and memory [20]. Circumstantial evidence of the involvement of glutamatergic pathways derives from the well-known role of structures such as the hippocampus in learning and memory [21]. More specifically, lesions of certain glutamatergic pathways impair learning and memory [22]. Moreover, glutamate and glutamate receptors are involved in mechanisms of synaptic plasticity, which are considered to underlie learning and memory [23][24][25].

3. Purine Metabolism

Purines and pyrimidines are components of many key molecules in living organisms. The primary purines adenine and guanosine and the pyrimidines cytosine, thymidine, and uracyl are the core of DNA, RNA, nucleosides, and nucleotides involved in energy transfer (ATP, GTP) [26][27]. Several studies indirectly suggested that purine metabolism has altered in AD. Energy metabolism, which depends on mitochondrial function and ATP production, is markedly altered in AD [28][29]. In addition, oxidative damage to DNA and RNA, as revealed by the increase in 8-hydroxyguanosine, is found in the brain samples of AD [30][31][32][33]. Direct alterations of purine metabolism in AD have been detected by metabolomics in postmortem ventricular CSF [34] and in the spinal cord CSF of living individuals [35][36][37]. Only a limited number of metabolomics studies have been carried out in AD brains [37].

4. Taurine and Hypotaurine Metabolism

Taurine is the second most abundant endogenous amino acid in the central nervous system (CNS) and has multiple roles in our body: thermoregulation [38], stabilization in regulating protein folding [39], anti-inflammatory effects [40], antioxidation [41], osmoregulation [42], and calcium homeostasis [43]. Recently, taurine has shown therapeutic effects as a cognitive enhancer in animal models of non-AD neurological disorders [44][45][46][47]. Taurine protected mice from the memory disruption induced by alcohol, pentobarbital, sodium nitrite, and cycloheximide but had no obvious effect on other behaviors including motor coordination, exploratory activity, and locomotor activity [44]. Intravenously injected taurine significantly improves post-injury functional impairments of traumatic brain injury in rats [45]. The intracerebroventricular (ICV) administration of taurine protects mice from learning impairment induced by hypoxia. Neither beta-alanine nor saccharose was able to mimic the effects of taurine [46]. In streptozotocin-induced sporadic dementia rat models, cognitive impairment and deterioration of neurobehavioral activities are ameliorated by taurine [47].
Taurine also has multiple disease-modifying roles to cease or prevent AD neuropathology. During the development of AD, amyloid-β (Aβ) progressively misfolded into toxic aggregates, which are strongly associated with neuronal loss, synaptic damage, and brain atrophy. An electron microscopy study indicates that taurine slightly decreases β-amyloid peptide aggregation in the brain at a millimolar concentration [48]. Taurine also has anti-inflammatory and antioxidant properties; it can provide protection for neuronal cells and mitochondria from the neurotoxicity of Aβ. By activating GABA and glycine receptors, taurine inhibits excitotoxicity caused by Aβ-induced glutamatergic transmission activation [49].

5. Cholinergic System

As acetylcholine (ACh) plays a vital role in cognitive processes, the cholinergic system is considered an important factor in AD [50]. The brain regions most affected by a loss of elements of the acetylcholine system include the hippocampus, cortex, and entorhinal [51]. Cholinesterase inhibitors are one of the few drug therapies available in the clinic for the treatment of AD, and it was inspired by the fact that cholinesterase inhibitors increase the availability of acetylcholine at brain synapses [52]. The validation of the cholinergic system was seen as an important therapeutic target in the disease.

6. Fatty Acids

Fatty acids are the basic building blocks of more complex lipids and can be classified by the number of double bonds as saturated fatty acids (SFAs) and unsaturated fatty acids. SFAs do not include any double bonds, whereas unsaturated fatty acids contain at least one (monounsaturated fatty acids, MUFAs) or two or more (polyunsaturated fatty acids, PUFAs) double bonds [53][54]. Altered unsaturated fatty acids have been associated with AD in multiple studies. The brain is especially enriched with two PUFAs: docosahexaenoic acid (DHA) and arachidonic acid (AA). DHA, as one of omega-3 PUFAs, is the predominant structural fatty acid in the mammalian brain and plays an essential role in brain functioning, especially in cognitive function; DHA levels were lower in AD brains [55][56] or plasma [57], and increased intake of DHA from fish or marine oils may lower AD risk [58][59][60]. AA of the ω-6 fatty acid family appears to play critical mediator roles in amyloid (Aβ)-induced pathogenesis, leading to learning, memory, and behavioral impairments in AD [61]. The levels of free AA have been found to increase in AD patient brain samples [62], whereas the levels of AA in phospholipids are reduced in the hippocampus of AD subjects [63].

7. Glycerolipids

Glycerolipids can be categorized into triacylglycerols (TAG, also known as triglycerides, TG), monoacylglycerol (MAG), and diacylglycerol (DAG) based on the number of acyl groups in the structure. TAG, the most predominant glycerolipids, are esters composed of a glycerol backbone and three fatty acids. TAG levels are found not to be changed in the serum of AD patients when compared to control subjects. [64]. However, MAG and DAG are elevated in both the prefrontal cortex and plasma of AD and MCI subjects in comparison to controls [65][66]. Moreover, MAG and DAG are elevated in the grey matter of MCI and AD patients, suggesting that these biochemical changes may play a role in the development of MCI and in the transition from MCI to AD [67].

8. Glycerophospholipids

Glycerophospholipids (GPs), also referred to as phospholipids (PLs), are typically amphipathic and make up the characteristic lipid bilayer structure of biological membranes. Moreover, GPs are the major type of lipids that make up cell membranes and account for 50–60% of the total membrane mass along with cholesterol and glycolipids [68]. GPs include phosphatidylethanolamine (PE), phosphatidic acid (PA), phosphatidylserine (PS), phosphatidylglycerol (PG), phosphatiylcholine (PC), phosphatidylinositol (PI), sphingomyelin (SM), and cardiolipin (CL) [69]. Studies on GP composition indicate that levels of PC, PE, and PI are significantly decreased in neural membranes from different regions of AD patients compared to age-matched control brains [70][71][72][73][74][75].
Phosphatidylethanolamine (PE) is converted to lysophosphatidylethanolamine (lyso-PE) by phospholipase A2 (PLA2), an important inflammatory mediator that is dysregulated in AD. PLA2 level has been found to be elevated in the human cerebral cortex [76] or decreased in the human parietal and frontal cortex [77]. Moreover, PLA2 influences the processing and secretion of amyloid precursor protein, which gives rise to the β-amyloid peptide, the major component of the amyloid plaque in AD [77]. Moreover, PLA2 has been found to play an important role in memory retrieval [78].
Phosphatidylserine (PS) is the major acidic phospholipid class that accounts for 13–15% of the phospholipids in the human cerebral cortex [79]. PS is known as a “brain nutrient”, as it can not only nourish the brain, but also enhance brain functions such as improving cognition, memory, and reaction force [80]. In six double-blind trials, PS has been found effective for AD. At daily doses of 200–300 mg for up to six months, PS consistently improved clinical global impression and activities of daily living [81]. In milder cases, PS improved orientation, concentration, learning, and memory for names, locations, and recent events. In the largest trial, involving 425 elderly patients (aged between 65 and 93 years) with moderate to severe cognitive decline, PS significantly improved memory, learning motivation, and socialization, suggesting that it has a vital impact on the quality of life of such elderly patients.
Phosphatidylcholine (PC) is an essential component of cell membranes and makes up approximately 95% of the total choline compound pool in most tissues [82][83]. Its function is defined primarily by chain length since chain length differences can affect cell membrane fluidity [84]. Three PCs (PC 16:0/20:5, PC 16:0/22:6, and PC 18:0/22:6) have been found significantly diminished in AD patients [85].
Lysophosphatidic acids (LPAs) are phospholipids derivatives that can act as signaling molecules [86]. Ahmad et al. [87] investigated the association between LPAs and CSF biomarkers of AD, Aβ-42, p-tau, and total tau levels overall and with MCI to AD progression. Five LPAs (LPA C16:0, LPA C16:1, LPA C22:4, LPA C22:6, and isomer-LPA C 22:5) correlated significantly and positively with CSF biomarkers of AD, Aβ-42, p-tau, and total tau. Additionally, LPA C16:0 and LPA C16:1 showed associations with MCI to AD dementia progression.

9. Sphingolipids

Sphingolipids, a class of membrane biomolecules, include sphingosine 1-phosphates (S1P), Ceramider (Cers), SMs, and glycosphingolipids, which are vital for maintaining cell integrity and signal transduction processes [88]. Cers, the basic structural units of the sphingolipid class, have been seen as key contributors to the pathology of AD as they are able to affect both Aβ generation and tau phosphorylation [89]. Filippov et al. found elevated levels of ceramides Cer16, Cer18, Cer20, and Cer24 in the brains of AD patients. Two saturated ceramides, Cer (d18:1/18:0) and Cer (d18:1/20:0) were significantly increased in the senile plaques [90]. High ceramide levels were also found in AD serum [91] and CSF samples [92]. The greatest genetic risk factor for late-onset AD is the ε4 allele of apolipoprotein E (ApoE). ApoE regulates the secretion of the potent neuroprotective signaling lipid S1P [93]. S1P is derived by phosphorylation of sphingosine, catalyzed by sphingosine kinases 1 and 2 (SphK1 and 2). SphK1 positively regulates glutamate secretion and synaptic strength in hippocampal neurons. Reduced levels of S1P have been found in AD brains compared to controls [93][94]. All these studies mentioned above suggested that sphingolipid metabolism plays a critical role in AD pathology.

10. Cholesterol and Cholesteryl Esters

Despite the brain occupying only 2% of total body weight, it contains 25% of the body’s cholesterol. Due to the BBB, cholesterol metabolism in the CNS is largely separated from that in the periphery and cholesterol is de novo synthesized in the CNS [95]. Studies have found that brain cholesterol was significantly increased in AD patients than in controls [96][97]. Moreover, cholesterol showed abnormal accumulation in the senile plaques of the human brain, a hallmark neuropathological feature of AD [98].

References

  1. Wu, G.; Morris, S.M., Jr. Arginine metabolism: Nitric oxide and beyond. Biochem. J. 1998, 336 Pt 1, 1–17.
  2. Morris, S.M., Jr. Enzymes of Arginine Metabolism. J. Nutr. 2004, 134, 2743S–2747S.
  3. Aktan, F. iNOS-mediated nitric oxide production and its regulation. Life Sci. 2004, 75, 639–653.
  4. Ellison, D.W.; Beal, M.F.; Mazurek, M.F.; Bird, E.D.; Martin, J.B. A postmortem study of amino acid neurotransmitters in Alzheimer’s disease. Ann. Neurol. 1986, 20, 616–621.
  5. Tumani, H.; Shen, G.; Peter, J.B.; Brück, W. Glutamine synthetase in cerebrospinal fluid, serum, and brain: A diagnostic marker for Alzheimer disease? Arch. Neurol. 1999, 56, 1241–1246.
  6. Satriano, J. Arginine pathways and the inflammatory response: Interregulation of nitric oxide and polyamines: Review article. Amino Acids 2004, 26, 321–329.
  7. Zhou, L.; Zhu, D.Y. Neuronal nitric oxide synthase: Structure, subcellular localization, regulation, and clinical implications. Nitric Oxide 2009, 20, 223–230.
  8. Susswein, A.J.; Katzoff, A.; Miller, N.; Hurwitz, I. Nitric oxide and memory. Neuroscientist 2004, 10, 153–162.
  9. Feil, R.; Kleppisch, T. NO/cGMP-dependent modulation of synaptic transmission. In Pharmacology of Neurotransmitter Release; Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 2008; pp. 529–560.
  10. Böger, R.H.; Bode-Böger, S.M.; Frölich, J.C. The L-arginine-nitric oxide pathway: Role in atherosclerosis and therapeutic implications. Atherosclerosis 1996, 127, 1–11.
  11. Cooke, J.P.; Dzau, V.J. Nitric oxide synthase: Role in the genesis of vascular disease. Annu. Rev. Med. 1997, 48, 489–509.
  12. Cooke, J.P. The pivotal role of nitric oxide for vascular health. Can. J. Cardiol. 2004, 20 (Suppl. B), 7b–15b.
  13. Li, X.A.; Everson, W.; Smart, E.J. Nitric oxide, caveolae, and vascular pathology. Cardiovasc. Toxicol. 2006, 6, 1–13.
  14. Napoli, C.; de Nigris, F.; Williams-Ignarro, S.; Pignalosa, O.; Sica, V.; Ignarro, L.J. Nitric oxide and atherosclerosis: An update. Nitric Oxide 2006, 15, 265–279.
  15. Fonnum, F. Glutamate: A neurotransmitter in mammalian brain. J. Neurochem. 1984, 42, 1–11.
  16. Francis, P.T.; Sims, N.R.; Procter, A.W.; Bowen, D.M. Cortical pyramidal neurone loss may cause glutamatergic hypoactivity and cognitive impairment in Alzheimer’s disease: Investigative and therapeutic perspectives. J. Neurochem. 1993, 60, 1589–1604.
  17. Bruno, V.; Battaglia, G.; Copani, A.; D’Onofrio, M.; Di Iorio, P.; De Blasi, A.; Melchiorri, D.; Flor, P.J.; Nicoletti, F. Metabotropic glutamate receptor subtypes as targets for neuroprotective drugs. J. Cereb. Blood Flow Metab. 2001, 21, 1013–1033.
  18. Gadea, A.; López-Colomé, A.M. Glial transporters for glutamate, glycine and GABA I. Glutamate transporters. J. Neurosci. Res. 2001, 63, 453–460.
  19. Ozawa, S.; Kamiya, H.; Tsuzuki, K. Glutamate receptors in the mammalian central nervous system. Prog. Neurobiol. 1998, 54, 581–618.
  20. Lynch, G. Memory and the brain: Unexpected chemistries and a new pharmacology. Neurobiol. Learn. Mem. 1998, 70, 82–100.
  21. Riedel, G.; Micheau, J. Function of the hippocampus in memory formation: Desperately seeking resolution. Prog. Neuropsychopharmacol. Biol. Psychiatry 2001, 25, 835–853.
  22. Myhrer, T. Effects of selective perirhinal and postrhinal lesions on acquisition and retention of a visual discrimination task in rats. Neurobiol. Learn. Mem. 2000, 73, 68–78.
  23. Baudry, M.; Lynch, G. Remembrance of arguments past: How well is the glutamate receptor hypothesis of LTP holding up after 20 years? Neurobiol. Learn. Mem. 2001, 76, 284–297.
  24. Jay, T.M.; Zilkha, E.; Obrenovitch, T.P. Long-term potentiation in the dentate gyrus is not linked to increased extracellular glutamate concentration. J. Neurophysiol. 1999, 81, 1741–1748.
  25. Scannevin, R.H.; Huganir, R.L. Postsynaptic organization and regulation of excitatory synapses. Nat. Rev. Neurosci. 2000, 1, 133–141.
  26. Ansoleaga, B.; Jové, M.; Schlüter, A.; Garcia-Esparcia, P.; Moreno, J.; Pujol, A.; Pamplona, R.; Portero-Otín, M.; Ferrer, I. Deregulation of purine metabolism in Alzheimer’s disease. Neurobiol. Aging 2015, 36, 68–80.
  27. Ipata, P.L.; Camici, M.; Micheli, V.; Tozz, M.G. Metabolic network of nucleosides in the brain. Curr. Top. Med. Chem. 2011, 11, 909–922.
  28. Ferreira, I.L.; Resende, R.; Ferreiro, E.; Rego, A.C.; Pereira, C.F. Multiple defects in energy metabolism in Alzheimer’s disease. Curr. Drug Targets 2010, 11, 1193–1206.
  29. Ferrer, I. Altered mitochondria, energy metabolism, voltage-dependent anion channel, and lipid rafts converge to exhaust neurons in Alzheimer’s disease. J. Bioenerg. Biomembr. 2009, 41, 425–431.
  30. Lovell, M.A.; Markesbery, W.R. Oxidatively modified RNA in mild cognitive impairment. Neurobiol. Dis. 2008, 29, 169–175.
  31. Lovell, M.A.; Soman, S.; Bradley, M.A. Oxidatively modified nucleic acids in preclinical Alzheimer’s disease (PCAD) brain. Mech. Ageing Dev. 2011, 132, 443–448.
  32. Markesbery, W.R.; Lovell, M.A. DNA oxidation in Alzheimer’s disease. Antioxid. Redox Signal. 2006, 8, 2039–2045.
  33. Nunomura, A.; Tamaoki, T.; Motohashi, N.; Nakamura, M.; McKeel, D.W., Jr.; Tabaton, M.; Lee, H.G.; Smith, M.A.; Perry, G.; Zhu, X. The earliest stage of cognitive impairment in transition from normal aging to Alzheimer disease is marked by prominent RNA oxidation in vulnerable neurons. J. Neuropathol. Exp. Neurol. 2012, 71, 233–241.
  34. Kaddurah-Daouk, R.; Rozen, S.; Matson, W.; Han, X.; Hulette, C.M.; Burke, J.R.; Doraiswamy, P.M.; Welsh-Bohmer, K.A. Metabolomic changes in autopsy-confirmed Alzheimer’s disease. Alzheimers Dement. 2011, 7, 309–317.
  35. Isobe, C.; Abe, T.; Terayama, Y. Levels of reduced and oxidized coenzyme Q-10 and 8-hydroxy-2’-deoxyguanosine in the CSF of patients with Alzheimer’s disease demonstrate that mitochondrial oxidative damage and/or oxidative DNA damage contributes to the neurodegenerative process. J. Neurol. 2010, 257, 399–404.
  36. Kaddurah-Daouk, R.; Zhu, H.; Sharma, S.; Bogdanov, M.; Rozen, S.G.; Matson, W.; Oki, N.O.; Motsinger-Reif, A.A.; Churchill, E.; Lei, Z.; et al. Alterations in metabolic pathways and networks in Alzheimer’s disease. Transl. Psychiatry 2013, 3, e244.
  37. Jové, M.; Portero-Otín, M.; Naudí, A.; Ferrer, I.; Pamplona, R. Metabolomics of human brain aging and age-related neurodegenerative diseases. J. Neuropathol. Exp. Neurol. 2014, 73, 640–657.
  38. Frosini, M.; Sesti, C.; Saponara, S.; Ricci, L.; Valoti, M.; Palmi, M.; Machetti, F.; Sgaragli, G. A specific taurine recognition site in the rabbit brain is responsible for taurine effects on thermoregulation. Br. J. Pharmacol. 2003, 139, 487–494.
  39. Bhat, M.A.; Ahmad, K.; Khan, M.S.A.; Bhat, M.A.; Almatroudi, A.; Rahman, S.; Jan, A.T. Expedition into Taurine Biology: Structural Insights and Therapeutic Perspective of Taurine in Neurodegenerative Diseases. Biomolecules 2020, 10, 863.
  40. Qaradakhi, T.; Gadanec, L.K.; McSweeney, K.R.; Abraham, J.R.; Apostolopoulos, V.; Zulli, A. The Anti-Inflammatory Effect of Taurine on Cardiovascular Disease. Nutrients 2020, 12, 2847.
  41. Schaffer, S.W.; Azuma, J.; Mozaffari, M. Role of antioxidant activity of taurine in diabetes. Can. J. Physiol. Pharmacol. 2009, 87, 91–99.
  42. Schaffer, S.; Takahashi, K.; Azuma, J. Role of osmoregulation in the actions of taurine. Amino Acids 2000, 19, 527–546.
  43. Foos, T.M.; Wu, J.Y. The role of taurine in the central nervous system and the modulation of intracellular calcium homeostasis. Neurochem. Res. 2002, 27, 21–26.
  44. Vohra, B.P.; Hui, X. Improvement of impaired memory in mice by taurine. Neural Plast. 2000, 7, 245–259.
  45. Su, Y.; Fan, W.; Ma, Z.; Wen, X.; Wang, W.; Wu, Q.; Huang, H. Taurine improves functional and histological outcomes and reduces inflammation in traumatic brain injury. Neuroscience 2014, 266, 56–65.
  46. Malcangio, M.; Bartolini, A.; Ghelardini, C.; Bennardini, F.; Malmberg-Aiello, P.; Franconi, F.; Giotti, A. Effect of ICV taurine on the impairment of learning, convulsions and death caused by hypoxia. Psychopharmacology 1989, 98, 316–320.
  47. Javed, H.; Khan, A.; Vaibhav, K.; Moshahid Khan, M.; Ahmad, A.; Ejaz Ahmad, M.; Ahmad, A.; Tabassum, R.; Islam, F.; Safhi, M.M.; et al. Taurine ameliorates neurobehavioral, neurochemical and immunohistochemical changes in sporadic dementia of Alzheimer’s type (SDAT) caused by intracerebroventricular streptozotocin in rats. Neurol. Sci. 2013, 34, 2181–2192.
  48. Santa-María, I.; Hernández, F.; Moreno, F.J.; Avila, J. Taurine, an inducer for tau polymerization and a weak inhibitor for amyloid-beta-peptide aggregation. Neurosci. Lett. 2007, 429, 91–94.
  49. Pan, C.; Prentice, H.; Price, A.L.; Wu, J.Y. Beneficial effect of taurine on hypoxia- and glutamate-induced endoplasmic reticulum stress pathways in primary neuronal culture. Amino Acids 2012, 43, 845–855.
  50. Ferreira-Vieira, T.H.; Guimaraes, I.M.; Silva, F.R.; Ribeiro, F.M. Alzheimer’s disease: Targeting the Cholinergic System. Curr. Neuropharmacol. 2016, 14, 101–115.
  51. Kása, P.; Rakonczay, Z.; Gulya, K. The cholinergic system in Alzheimer’s disease. Prog. Neurobiol. 1997, 52, 511–535.
  52. Hampel, H.; Mesulam, M.M.; Cuello, A.C.; Farlow, M.R.; Giacobini, E.; Grossberg, G.T.; Khachaturian, A.S.; Vergallo, A.; Cavedo, E.; Snyder, P.J.; et al. The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease. Brain 2018, 141, 1917–1933.
  53. de Carvalho, C.; Caramujo, M.J. The Various Roles of Fatty Acids. Molecules 2018, 23, 2583.
  54. Kao, Y.C.; Ho, P.C.; Tu, Y.K.; Jou, I.M.; Tsai, K.J. Lipids and Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 1505.
  55. Fonteh, A.N.; Cipolla, M.; Chiang, A.J.; Edminster, S.P.; Arakaki, X.; Harrington, M.G. Polyunsaturated Fatty Acid Composition of Cerebrospinal Fluid Fractions Shows Their Contribution to Cognitive Resilience of a Pre-symptomatic Alzheimer’s Disease Cohort. Front. Physiol. 2020, 11, 83.
  56. Cunnane, S.C.; Schneider, J.A.; Tangney, C.; Tremblay-Mercier, J.; Fortier, M.; Bennett, D.A.; Morris, M.C. Plasma and brain fatty acid profiles in mild cognitive impairment and Alzheimer’s disease. J. Alzheimers Dis. 2012, 29, 691–697.
  57. Gonzalez-Dominguez, R.; Garcia-Barrera, T.; Gomez-Ariza, J.L. Using direct infusion mass spectrometry for serum metabolomics in Alzheimer’s disease. Anal. Bioanal. Chem. 2014, 406, 7137–7148.
  58. Kalmijn, S.; Launer, L.J.; Ott, A.; Witteman, J.C.; Hofman, A.; Breteler, M.M. Dietary fat intake and the risk of incident dementia in the Rotterdam Study. Ann. Neurol. 1997, 42, 776–782.
  59. Morris, M.C.; Evans, D.A.; Bienias, J.L.; Tangney, C.C.; Bennett, D.A.; Wilson, R.S.; Aggarwal, N.; Schneider, J. Consumption of fish and n-3 fatty acids and risk of incident Alzheimer disease. Arch. Neurol. 2003, 60, 940–946.
  60. Huang, T.L.; Zandi, P.P.; Tucker, K.L.; Fitzpatrick, A.L.; Kuller, L.H.; Fried, L.P.; Burke, G.L.; Carlson, M.C. Benefits of fatty fish on dementia risk are stronger for those without APOE epsilon4. Neurology 2005, 65, 1409–1414.
  61. Sanchez-Mejia, R.O.; Mucke, L. Phospholipase A2 and arachidonic acid in Alzheimer’s disease. Biochim. Biophys. Acta 2010, 1801, 784–790.
  62. Snowden, S.G.; Ebshiana, A.A.; Hye, A.; An, Y.; Pletnikova, O.; O’Brien, R.; Troncoso, J.; Legido-Quigley, C.; Thambisetty, M. Association between fatty acid metabolism in the brain and Alzheimer disease neuropathology and cognitive performance: A nontargeted metabolomic study. PLoS Med. 2017, 14, e1002266.
  63. Prasad, M.R.; Lovell, M.A.; Yatin, M.; Dhillon, H.; Markesbery, W.R. Regional membrane phospholipid alterations in Alzheimer’s disease. Neurochem. Res. 1998, 23, 81–88.
  64. Proitsi, P.; Kim, M.; Whiley, L.; Simmons, A.; Sattlecker, M.; Velayudhan, L.; Lupton, M.K.; Soininen, H.; Kloszewska, I.; Mecocci, P.; et al. Association of blood lipids with Alzheimer’s disease: A comprehensive lipidomics analysis. Alzheimers Dement. 2017, 13, 140–151.
  65. Chan, R.B.; Oliveira, T.G.; Cortes, E.P.; Honig, L.S.; Duff, K.E.; Small, S.A.; Wenk, M.R.; Shui, G.; Di Paolo, G. Comparative lipidomic analysis of mouse and human brain with Alzheimer disease. J. Biol. Chem. 2012, 287, 2678–2688.
  66. Wood, P.L.; Medicherla, S.; Sheikh, N.; Terry, B.; Phillipps, A.; Kaye, J.A.; Quinn, J.F.; Woltjer, R.L. Targeted Lipidomics of Fontal Cortex and Plasma Diacylglycerols (DAG) in Mild Cognitive Impairment and Alzheimer’s Disease: Validation of DAG Accumulation Early in the Pathophysiology of Alzheimer’s Disease. J. Alzheimers Dis. 2015, 48, 537–546.
  67. Wood, P.L.; Barnette, B.L.; Kaye, J.A.; Quinn, J.F.; Woltjer, R.L. Non-targeted lipidomics of CSF and frontal cortex grey and white matter in control, mild cognitive impairment, and Alzheimer’s disease subjects. Acta Neuropsychiatr. 2015, 27, 270–278.
  68. Farooqui, A.A.; Horrocks, L.A.; Farooqui, T. Glycerophospholipids in brain: Their metabolism, incorporation into membranes, functions, and involvement in neurological disorders. Chem. Phys. Lipids 2000, 106, 1–29.
  69. Bargui, R.; Solgadi, A.; Prost, B.; Chester, M.; Ferreiro, A.; Piquereau, J.; Moulin, M. Phospholipids: Identification and Implication in Muscle Pathophysiology. Int. J. Mol. Sci. 2021, 22, 8176.
  70. Pettegrew, J.W.; Panchalingam, K.; Hamilton, R.L.; McClure, R.J. Brain membrane phospholipid alterations in Alzheimer’s disease. Neurochem. Res. 2001, 26, 771–782.
  71. Igarashi, M.; Ma, K.; Gao, F.; Kim, H.W.; Rapoport, S.I.; Rao, J.S. Disturbed choline plasmalogen and phospholipid fatty acid concentrations in Alzheimer’s disease prefrontal cortex. J. Alzheimers Dis. 2011, 24, 507–517.
  72. Han, X.; Holtzman, D.M.; McKeel, D.W., Jr. Plasmalogen deficiency in early Alzheimer’s disease subjects and in animal models: Molecular characterization using electrospray ionization mass spectrometry. J. Neurochem. 2001, 77, 1168–1180.
  73. Guan, Z.; Wang, Y.; Cairns, N.J.; Lantos, P.L.; Dallner, G.; Sindelar, P.J. Decrease and structural modifications of phosphatidylethanolamine plasmalogen in the brain with Alzheimer disease. J. Neuropathol. Exp. Neurol. 1999, 58, 740–747.
  74. Wells, K.; Farooqui, A.A.; Liss, L.; Horrocks, L.A. Neural membrane phospholipids in Alzheimer disease. Neurochem. Res. 1995, 20, 1329–1333.
  75. Söderberg, M.; Edlund, C.; Kristensson, K.; Dallner, G. Fatty acid composition of brain phospholipids in aging and in Alzheimer’s disease. Lipids 1991, 26, 421–425.
  76. Stephenson, D.T.; Lemere, C.A.; Selkoe, D.J.; Clemens, J.A. Cytosolic phospholipase A2 (cPLA2) immunoreactivity is elevated in Alzheimer’s disease brain. Neurobiol. Dis. 1996, 3, 51–63.
  77. Gattaz, W.F.; Maras, A.; Cairns, N.J.; Levy, R.; Förstl, H. Decreased phospholipase A2 activity in Alzheimer brains. Biol. Psychatry 1995, 37, 13–17.
  78. Schaeffer, E.L.; Gattaz, W.F. Requirement of hippocampal phospholipase A2 activity for long-term memory retrieval in rats. J. Neural Transm. 2007, 114, 379–385.
  79. Kim, H.Y.; Huang, B.X.; Spector, A.A. Phosphatidylserine in the brain: Metabolism and function. Prog. Lipid Res. 2014, 56, 1–18.
  80. Zhang, Y.Y.; Yang, L.Q.; Guo, L.M. Effect of phosphatidylserine on memory in patients and rats with Alzheimer’s disease. Genet. Mol. Res. 2015, 14, 9325–9333.
  81. Kidd, P.M. Omega-3 DHA and EPA for cognition, behavior, and mood: Clinical findings and structural-functional synergies with cell membrane phospholipids. Altern. Med. Rev. 2007, 12, 207–227.
  82. Frisardi, V.; Panza, F.; Seripa, D.; Farooqui, T.; Farooqui, A.A. Glycerophospholipids and glycerophospholipid-derived lipid mediators: A complex meshwork in Alzheimer’s disease pathology. Prog. Lipid Res. 2011, 50, 313–330.
  83. Schaeffer, E.L.; da Silva, E.R.; Novaes Bde, A.; Skaf, H.D.; Gattaz, W.F. Differential roles of phospholipases A2 in neuronal death and neurogenesis: Implications for Alzheimer disease. Prog. Neuropsychopharmacol. Biol. Psychiatry 2010, 34, 1381–1389.
  84. Perttu, E.K.; Kohli, A.G.; Szoka, F.C., Jr. Inverse-phosphocholine lipids: A remix of a common phospholipid. J. Am. Chem. Soc. 2012, 134, 4485–4488.
  85. Whiley, L.; Sen, A.; Heaton, J.; Proitsi, P.; García-Gómez, D.; Leung, R.; Smith, N.; Thambisetty, M.; Kloszewska, I.; Mecocci, P.; et al. Evidence of altered phosphatidylcholine metabolism in Alzheimer’s disease. Neurobiol. Aging 2014, 35, 271–278.
  86. Kang, S.; Han, J.; Song, S.Y.; Kim, W.S.; Shin, S.; Kim, J.H.; Ahn, H.; Jeong, J.H.; Hwang, S.J.; Sung, J.H. Lysophosphatidic acid increases the proliferation and migration of adipose-derived stem cells via the generation of reactive oxygen species. Mol. Med. Rep. 2015, 12, 5203–5210.
  87. Ahmad, S.; Orellana, A.; Kohler, I.; Frölich, L.; de Rojas, I.; Gil, S.; Boada, M.; Hernández, I.; Hausner, L.; Bakker, M.H.M.; et al. Association of lysophosphatidic acids with cerebrospinal fluid biomarkers and progression to Alzheimer’s disease. Alzheimers Res. Ther. 2020, 12, 124.
  88. Jones, E.E.; Dworski, S.; Canals, D.; Casas, J.; Fabrias, G.; Schoenling, D.; Levade, T.; Denlinger, C.; Hannun, Y.A.; Medin, J.A.; et al. On-tissue localization of ceramides and other sphingolipids by MALDI mass spectrometry imaging. Anal. Chem. 2014, 86, 8303–8311.
  89. Jazvinšćak Jembrek, M.; Hof, P.R.; Šimić, G. Ceramides in Alzheimer’s Disease: Key Mediators of Neuronal Apoptosis Induced by Oxidative Stress and Aβ Accumulation. Oxid. Med. Cell. Longev. 2015, 2015, 346783.
  90. Panchal, M.; Gaudin, M.; Lazar, A.N.; Salvati, E.; Rivals, I.; Ayciriex, S.; Dauphinot, L.; Dargère, D.; Auzeil, N.; Masserini, M.; et al. Ceramides and sphingomyelinases in senile plaques. Neurobiol. Dis. 2014, 65, 193–201.
  91. Mielke, M.M.; Bandaru, V.V.; Haughey, N.J.; Xia, J.; Fried, L.P.; Yasar, S.; Albert, M.; Varma, V.; Harris, G.; Schneider, E.B.; et al. Serum ceramides increase the risk of Alzheimer disease: The Women’s Health and Aging Study II. Neurology 2012, 79, 633–641.
  92. Satoi, H.; Tomimoto, H.; Ohtani, R.; Kitano, T.; Kondo, T.; Watanabe, M.; Oka, N.; Akiguchi, I.; Furuya, S.; Hirabayashi, Y.; et al. Astroglial expression of ceramide in Alzheimer’s disease brains: A role during neuronal apoptosis. Neuroscience 2005, 130, 657–666.
  93. Couttas, T.A.; Kain, N.; Daniels, B.; Lim, X.Y.; Shepherd, C.; Kril, J.; Pickford, R.; Li, H.; Garner, B.; Don, A.S. Loss of the neuroprotective factor Sphingosine 1-phosphate early in Alzheimer’s disease pathogenesis. Acta Neuropathol. Commun. 2014, 2, 9.
  94. He, X.; Huang, Y.; Li, B.; Gong, C.X.; Schuchman, E.H. Deregulation of sphingolipid metabolism in Alzheimer’s disease. Neurobiol. Aging 2010, 31, 398–408.
  95. Feringa, F.M.; van der Kant, R. Cholesterol and Alzheimer’s Disease; From Risk Genes to Pathological Effects. Front. Aging Neurosci. 2021, 13, 690372.
  96. Cutler, R.G.; Kelly, J.; Storie, K.; Pedersen, W.A.; Tammara, A.; Hatanpaa, K.; Troncoso, J.C.; Mattson, M.P. Involvement of oxidative stress-induced abnormalities in ceramide and cholesterol metabolism in brain aging and Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2004, 101, 2070–2075.
  97. Heverin, M.; Bogdanovic, N.; Lütjohann, D.; Bayer, T.; Pikuleva, I.; Bretillon, L.; Diczfalusy, U.; Winblad, B.; Björkhem, I. Changes in the levels of cerebral and extracerebral sterols in the brain of patients with Alzheimer’s disease. J. Lipid Res. 2004, 45, 186–193.
  98. Mori, T.; Paris, D.; Town, T.; Rojiani, A.M.; Sparks, D.L.; Delledonne, A.; Crawford, F.; Abdullah, L.I.; Humphrey, J.A.; Dickson, D.W.; et al. Cholesterol accumulates in senile plaques of Alzheimer disease patients and in transgenic APP(SW) mice. J. Neuropathol. Exp. Neurol. 2001, 60, 778–785.
More
Video Production Service