Choline is an essential nutrient involved in membrane lipid formation, neurotransmission and one carbon metabolism. Most of dietary choline is produced from phosphatidylcholine, a phospholipid abundant in eggs, meat and dairy products. Choline deficiency is implicated in neuronal development, liver disease, insulin resistance, muscle damage and choline is also vital during fetal development. The focus of this review is to shed light on the critical contributions of choline to membrane phosphatidylcholine formation and energy homeostasis. Moreover, this review will also focus on the role of choline in protecting against developmental disorders, particularly during pregnancy and fetal development while also focusing on adult onset metabolic disorders such as non-alcoholic liver disease and diabetes.
*Corresponding Author:
Dr. Marica Bakovic
Department of Human Health and Nutritional Sciences, University of Guelph, N1G 2W1, Guelph, Ontario, Canada,
E-mail: mbakovic@uoguelph.ca
Abstract
Choline is an essential nutrient involved in membrane lipid formation, neurotransmission and one carbon metabolism. Most of dietary choline is produced from phosphatidylcholine, a phospholipid abundant in eggs, meat and dairy products. Choline deficiency is implicated in neuronal development, liver disease, insulin resistance, muscle damage and choline is also vital during fetal development. The focus of this review is to shed light on the critical contributions of choline to membrane phosphatidylcholine formation and energy homeostasis. Moreover, this review will also focus on the role of choline in protecting against developmental disorders, particularly during pregnancy and fetal development while also focusing on adult onset metabolic disorders such as non-alcoholic liver disease and diabetes.
Key Words: Choline; Betaine; Neurotransmission; Phosphatidylcholine.
Introduction
Choline is an essential nutrient involved in membrane lipid formation, neurotransmission and one carbon metabolism. In addition to free choline, most dietary choline is produced from the phospholipid phosphatidylcholine (PC) [1]. Choline Adequate Intake (AI) of 550 mg/day for men, 425 mg/day for women, 450 mg/day for pregnant women and 550 mg/day for lactating women and has been considered a required dietary nutrient by the US Institute of Medicine’s Food and Nutrition Board since 1998. However, numerous studies have reported that 4 in 5 Americans are not reaching the AI for choline. The Framingham Heart Study reported that Americans consumed on average 203 mg/day of choline whereas the Nurses’ Health Study and the Atherosclerosis Risk in Communities study reported 293 and 217 mg/day choline intake respectively, far below the AI value [2-5]. Choline is most abundant in eggs, meat and dairy products [6]. To help improve choline intake, choline rich cereal based functional foods [7], milk [8] and goats [9] have been developed.
Choline deficiency is implicated in fatty liver [10], insulin resistance [11] and muscle damage in humans [12] and rodents [13]. Humans who have mutations in PCYT1A, the gene that encodes the rate limiting enzyme for PC synthesis via the CDP-choline pathway, have been shown to have severe insulin resistance and lipodystrophy [14]. In mice, choline has been shown to be particularly important during fetal development as gestational deficiency leads to memory and learning deficits later in life [15]. The focus of this review is to shed light on the importance of choline in numerous metabolic functions and suboptimal choline intake can hinder the ability for humans to carry out these functions.
Upon entry into the cell, most of choline is incorporated into phospholipid phosphatidylcholine (PC) via CDP-choline (Kennedy) pathway (Figure 1). In the first step of the pathway choline is phosphorylated by choline kinase (CK) to generate phosphocholine (PCho). PCho is then coupled with CTP by the CTP: phosphocholine cytidylyltransferase (CCT/Pcyt1) to generate CDP-choline. In the final step of the pathway, CDP-choline is condensed with diacylglycerol (DAG) by CDP-choline: DAG phosphotransferase in the endoplasmic reticulum to yield PC [16-20]. In the liver, PC is additionally generated from another phospholipid, phosphatidylethanolamine (PE), by a three-step methylation reaction catalyzed by PE methyltransferase (PEMT) [21]. This is an important backup pathway that prevents choline deficiencies when demands for choline are high, such as during embryonal development, pregnancy, and lactation [22]. Moreover, PEMT formed PC is incorporated into very-low density lipoproteins and as such transported from the liver into other tissues [23]. PEMT formed PC is enriched with n-3 PUFA, which is crucial for providing fetuses with n-3 PUFA for proper developmental programming [24] and the optimal function of neurological tissues [25- 27].
Degradation of PC by phospholipases is a critical element of choline homeostasis [28]. Choline is released from PC by phospholipase D (PLD), phospholipase A2 (PLA2) and then PLD, or by the PC base-exchange mechanisms (PSS1) as further described in Figure 1. PC is also converted into glycerophosphocholine (GPC), a prominent acetylcholine precursor and osmolyte via PLA2 [29]. PLD1 and PLD2 cleave PC between the phosphate moiety and choline head group to generate free choline and phosphatidic acid (PA). PA is an important lipid intermediate utilized for DAG formation or directly involved in signaling pathways such as the mTOR pathway [30]. Moreover, in genetic disorders with choline deficiencies such as postural tachycardia syndrome, there is an increase in PLD activity apparently in an attempt to generate more intracellular choline when choline transport in reduced [31]. Free choline is also released at the mitochondria associated ER membranes (MAM) when PC is converted into phosphatidylserine (PS) by PS synthase 1 (PSS1) but the exact utilization of the choline formed at the MAM is not known [32].
Choline oxidative demethylation is linked to energy metabolism
Aside from its role in PC synthesis, choline is oxidized in mitochondria of liver and kidney to betaine. Betaine is a methyl group donor in the one-carbon (methyonine-homocysteine) cycle (Figure 2) [33-35]. Choline and betaine supplementation to positively affect one carbon metabolism to correct perturbations in fatty acid (FA) oxidation and energy balance which are evident in obesity. In mice with excess fat accumulation, 4 weeks of choline supplementation increased plasma glycerol and glycine, which are products of increased lipolysis and choline oxidation respectively [36]. Moreover, plasma creatine and sarcosine were reduced, further indicating an increase in homocysteine/methionine cycling to breakdown choline [36]. In this model, SAM was more devoted to maintaining cellular PC balance, which is often perturbed in obesity, instead of guanidinoacetate methylation for creatine synthesis. In the skeletal muscle of mice, the main beneficial effects of supplemental choline included the reduction of energy stored as free FA, DAG and TAG and increasing energy utilization [37]. Mice with excess fat accumulation supplemented with choline or supplemented for 8 weeks with betaine showed increased oxidative demethylation of these methyl donors [38]. Oxidative demethylation of choline and betaine is beneficial as this requires energy in the form of reducing equivalents produced from succinate and α-ketoglutarate in the TCA cycle to be broken down [38]. In essence, choline and betaine breakdown increased metabolic demand, thereby decreasing the propensity for energy to be stored as fat throughout the body.
Choline and betaine protect against fatty liver and insulin resistance
Non-alcoholic fatty liver disease (NAFLD) has become the most prominent chronic disease in humans and its prevalence has risen concomitant with insulin resistance, type 2 diabetes (T2D) and obesity over the past 30 years [39]. NAFLD comprises a wide spectrum of pathologies, ranging from lipid droplets and simple steatosis to hepatitis to fibrosis and cirrhosis [40]. Dietary factors such as high caloric intake, high fat intake [41], high fructose intake, refined grain and processed meat consumption have been positively correlated with NAFLD development [42]. Though extensively studied, the mechanism responsible for the pathogenesis of NAFLD remains poorly understood.
Dietary choline is an important nutrient for maintaining optimal hepatic function. Choline deficiency has widespread effects on the one carbon metabolic system and lipid synthesis in the liver. In choline deficient mice, SAM concentrations were decreased by 50% after the consumption of a choline deficient diet for 2 weeks [43]. Due to this decrease in SAM concentration, there is a concomitant decrease in PEMT activity, and therefore PE derived PC phospholipid content [44]. The PEMT enzyme is important for the synthesis of PC used in very low-density lipoproteins (VLDL), which is critical for TAG export from the liver [45]. PC is a critical component of VLDL particles and a diminished capacity for the liver to synthesize PC via the PEMT pathway results in decreased lipoprotein secretion and TAG accumulation in hepatocytes [46]. Hepatic steatosis resulting from choline deficiency is a hallmark feature of VLDL deficiency in species such as humans and cats [47]. Choline supplementation improves hepatic function in mice by normalizing the expression of genes involved in lipogenesis [SREBP1, FAS, SCD1] and lipolysis [ATGL, HSL, LPL], as well as FA oxidation [PPARα] and mitochondrial biogenesis [PGC-1α] [36]. These changes result in a decrease in hepatic TAG [36, 48] and plasma acylcarnitines, which is an indication of improvements in mitochondrial fatty acid metabolism [36, 49].
Betaine has been shown to be hepatoprotective in mice with respect to numerous toxic substances such as ethanol, lipopolysaccharide and dimethylnitrosamine [50 - 52]. The liver injury resulting from these toxicants is largely due to altered sulfur amino acid metabolism, and betaine has been shown to be beneficial in this regard [53, 54]. Hepatocyte cell volume in mammalian systems is often altered during oxidative stress and these changes in cell volume activate signal transduction cascades in an attempt to allow the cell to respond to stress more effectively [55, 56]. BHMT, CHDH and PEMT expression is increased in hypotonic conditions, indicating a coordinated response to increase cell volume and regenerate methionine from Hcy for eventual methyl group donation [57]. Additionally, betaine is a lipotropic compound and helps to increase the levels of SAM and GSH in the liver which are critical for maintaining proper methylation and redox states [58].
Betaine has been shown in mice to reduce body weight induced by HFD consumption as well as hepatic and visceral fat mass by increasing AMPK activation [59]. AMPK is a positive regulator of fatty acid oxidation and decreases the expression of genes such as SREBP-1c, ACC and FAS which promote lipogenesis [60]. Furthermore, AMPK can directly phosphorylate PGC-1α to stimulate mitochondrial biogenesis [61]. Betaine has also been shown to reverse increased serum insulin levels and improve glucose tolerance [62] while also reducing serum TAG and cholesterol levels. These findings are supplemented by the activation of insulin signaling in the liver as seen by increasing phosphorylation of IRS1 and activation of Akt [63].
In mice, plasma acyl-carnitine levels are lowered substantially with choline and betaine supplementation, which is beneficial as an abundance of acyl-carnitines in the plasma is indicative of inefficient mitochondrial FA oxidation, a metabolic issue that often affects obese individuals [36, 64]. As a result, acyl-carnitine levels can provide an indication of the ability of the cell to oxidize FA [65] and to maintain optimal mitochondrial function [66]. In mice, long chain acyl-CoA species are critical regulators of metabolism as they inhibit mitochondrial adenine nucleotide translocase [ANT], which regulates ATP/ADP exchange across the inner mitochondrial membrane [66]. Additionally, malonyl-CoA is an allosteric inhibitor of CPT1, the rate limiting enzyme in the oxidation of long chain fatty acyl-CoA by facilitating its entry into the mitochondrial matrix. Moreover, choline supplementation, decreased expression of genes which are critical for fatty acid synthesis [FAS and ACC] while the expression of genes involved with FA oxidation was increased [PPARα and PGC-1α] [37].
Supplementation of choline and betaine in vivo has been shown to ameliorate perturbations with lipid metabolism that arise in obesity. Choline and betaine supplementation in mice decreased liver and adipose tissue weight while also reducing lipid droplet size which is largely due to decreased FA incorporation into TAG [37]. In addition, betaine supplementation significantly decreased plasma TAG content while choline supplementation reduced collagen deposition, indicating decreased inflammation and fibrosis throughout the liver [38]. Membrane composition was also remodeled with choline supplementation as the ratio of important lipid raft components FC: SM was increased [37]. The optimal balance between FA oxidation and lipogenesis was restored with choline supplementation by activating the main regulator of skeletal muscle FA oxidation AMPK [36, 37]. Subsequently, mTORC1 activation was decreased which in turn facilitate a decrease in SREBP1c nuclear translocation for lipogenic gene transcription [37]. With choline supplementation, glycogen content, which is often depleted in insulin resistant skeletal muscle, was also restored, indicating an enhanced ability to store glucose taken up from the blood.
Therefore, choline and betaine are critical for mitigating the development of NAFLD. Choline and betaine both decrease lipid accumulation in the liver while also decreasing hepatic fibrosis. Metabolism is impacted by stimulating TAG degradation by lipolysis to generate a fasted energy deficient state. Choline is the predominant phospholipid component of VLDL which is important for exporting TAG from the liver. Improving dietary choline and betaine levels has promoted fatty acid oxidation by increasing AMPK activation and improving insulin signaling rodent models [37]. Despite considerable mechanistic research, there is no clear physiological concentration of choline or betaine that is considered adequate for ameliorating fatty liver or insulin. However, as the majority of Americans are deficient in choline [2], reaching the AI for choline should help diminish the incidence of fatty liver [10] and insulin resistance [11].
The role of choline and epigenetics in pregnancy and gestation
During pregnancy, maternal choline affects metabolic and physiological functions of the offspring through numerous inter-related mechanisms. Maternal choline is vital for optimal placental development and maintaining optimal fetal growth [67, 68]. The increased requirement for choline during gestation is largely due to an increased use of betaine as a methyl group donor and a higher demand for choline utilization for PC synthesis [69]. This could lead to substantial depletion of choline derived methyl donors such as betaine and SAM in pregnant women [70, 71]. A reduction in choline and betaine leads to elevated homocysteine and decreased SAM levels, thereby decreasing methyltransferase activity as SAM is a positive regulator of MAT [72].
Genetic variants that increase choline requirements can leave an individual susceptible to choline inadequacy [73], and this effect can be magnified during reproduction [74]. Some of these SNPs are located in genes such as PEMT (rs12325817), choline dehydrogenase (CHDH) (rs12676) and MTHFR (rs1801133) which are critical for proper functioning of the one carbon metabolic system [75-77]. In addition, estimations from the NHANES study estimated that only 1 out of 10 pregnant women in the US are consuming adequate amounts of choline [78], which is a troubling statistic as choline is a vital nutrient for fetal development.
The prenatal period is vital for the establishment and maintenance of the epigenome [79]. The DNA methylation patterns of gametes are mostly abolished after conception; therefore de novo methylation is crucial for gene silencing [80]. DNA methylation is closely linked to histone modification in order to facilitate time-sensitive alterations throughout fetal development [81]. In addition, the fetal epigenome can be affected by numerous maternal environmental factors such as nutrition [82]. During pregnancy, it has been determined that maternal choline supply has dramatic effects on the fetal epigenome [83]. The fetuses of choline deficient mothers exhibited hypermethylation of the insulin growth factor 2 (IGF2) gene [84], which is critical for embryonic development [85]. Additionally, choline deficient mothers exhibited hypomethylation of DNMT1 and this was correlated with the epigenetic and expression changes of IGF2, suggesting that maternal choline deficiency exerted its effects on IGF2 via DNMT1 hypomethylation [86].
Maternal choline intake during pregnancy is critical for optimal cognitive function [87, 88]. There is substantial evidence that adequate maternal choline levels during pregnancy are required for optimal hippocampal function, and therefore maintaining cognitive abilities [89-91]. Choline deficiency during pregnancy has been shown to decrease hippocampal methylation of the cyclin dependent kinase inhibitor 3 (CDKN3) gene and increase the expression of kinase associated phosphatase [Kap], a known inhibitor of cell proliferation [92]. Maternal PC levels during pregnancy alter hippocampal cholinergic function of the offspring, which is associated with depression and anxiety later in life [93]. This is because PC is important for ensuring adequate neuron density [94], propagation of intracellular signals [95] and optimal membrane configuration [96]. PC can also be used for acetylcholine synthesis, and therefore cholinergic transmission [97]. Choline intake is an important component of neurological development [98] as it has a large influence on structural integrity, function and cell signaling within the brain [99, 100].
Additionally, choline supplementation in pregnant women has been shown to increase placental FATP4 content, which transports DHA to the fetus for neurological development [101]. Moreover, maternal choline supplementation increases the levels of DHA-enriched PC [PC-DHA], which is mainly synthesized by the PEMT pathway [102]. The PEMT pathway is especially important for fortifying phospholipids with DHA as this enzyme prefers PE-DHA as a substrate [103]. This is in contrast to the CDP-choline pathway which predominantly synthesizes PC containing saturated medium length FA [104]. With this in mind, maternal choline supplementation can serve as a dietary approach to supply the developing fetus with DHA. Umbilical cord choline content can be up to 5-fold higher than what is observed in maternal blood, further emphasizing the importance of choline in fetal development [99]. Furthermore, the PEMT gene has an estrogen response element within its promoter region, meaning that its expression can be induced when estrogen is present [101]. Estrogen is typically abundant during pregnancy, and is therefore important for the fortification of the developing fetus with PC-DHA [101]. In fact, women with SNPs in within the estrogen response element are much more susceptible to developing choline deficiency [101]. Prenatal omega-3 fatty acid supplementation in pregnant dams that consuming a diet low in folate and vitamin B12 normalizes global DNA methylation levels in the placenta and the brain [104]. This implies that omega-3 fatty acids are involved in modifying methylation patterns [104].
Taken together, choline and PC have critical roles with respect to pregnancy and fetal development. Choline is a critical molecule which can ultimately serve as a methyl donor within the one carbon metabolic system. As a result, choline deficiency has a negative impact on fetal development due to perturbed epigenetic regulation. In addition, choline deficiency has detrimental effects on hippocampal development, and can lead to cognitive deficiencies. Lastly, PC is an important molecule for fetal DHA enrichment and hormones [i.e. estrogen] and enzymes [PEMT] help facilitate this process.
Neuroprotective roles of choline
Choline levels have been shown to be integral to maintaining optimal neurological function over time. As the number of older individuals in our population increases, the impact of choline deficiency on the health of these populations becomes more important. With aging, cerebral function becomes impaired through myelin degradation, decreased synaptic function and dysregulation of DNA methylation. Due to these pathologies, older adults experience cognitive decline and are increasingly affected by neurological disorders like Alzheimer’s disease [AD]. Studies have established a positive correlation between individuals carrying mutations which alter MTHFR activity and AD, linking choline metabolism and neurological function. Choline can act as a reserve methyl donor when MTHFR function is diminished but this will deplete choline pools that are required for various neurological functions. These functions include the integration of choline into the neurotransmitter acetylcholine and lipids like PC, which are associated with hallmark neurological perturbations of AD such as memory impairment and anxiety [105]. Moreover, the quantity of lysoPC species, which are pro- inflammatory lipid mediators, as well as sphingolipid breakdown products have been found to be markedly increased in AD patients [106, 107].
The focus on nutrition has become increasingly important with regards to neurological disorders, largely because synaptic membranes require numerous nutrients to be synthesized. Van Wijk et al. [108] report that cognitively impaired subjects had lower circulating levels of choline and folate, both of which are fundamental to membrane phospholipid synthesis. Mellott et al. [109] have shown that neonatal choline consumption can mitigate AD related cognitive decline later in life by attenuating amyloid plaque formation. As in other models, in utero choline supplementation has been shown to prevent a decline in hippocampal neurogenesis in adulthood while also rescuing cholinergic functions, which are hallmarks of AD [110, 111]. In mice, maternal choline supplementation has therapeutic potential by normalizing the expression of genes involved with synaptic plasticity in offspring. This is key for protecting basal forebrain cholinergic neurons mitigating decline in spatial cognition [112]. Cytidine 5’-Diphosphocholine [citicoline], a PC precursor, has also been shown to improve cognitive performance in AD patients as a safe and effective agent to increase PC levels in the brain [113, 114].
Interplay between fatty acids and phospholipids in modulating membrane function
It is widely conceived that fatty acid esterification in critical for efficiently incorporating n- 3 fatty acids into the brain [115, 116], and that phospholipids are important carriers of n- 3 fatty acids [26]. Esterified fatty acids are more readily absorbed into the body relative to fatty acids in TAG or unesterified fatty acids [117]. Additionally, n-3 fatty acids are more resistant to oxidation when incorporated into phospholipids relative to TAG [118]. Moreover, fatty acids esterified in phospholipids relative to TAG are more bioavailable [119], which is especially important in brain development [120-122].
An important role of n-3 fatty acids is their role in modulating biophysical properties within the cell membrane which play a role in signal transduction cascades [123, 124]. For instance, Li et al. [2007] demonstrated that plasma membrane DHA content is integral for elevating eNOS activity by altering the lipid: protein interactions in caveolar microdomains, thereby facilitating the translocation of eNOS from the plasma membrane [125]. Moreover, glucose transport has been shown to be modulated by the effects of n-3 fatty acids on its configuration within the membrane [126 - 128]. Imbalances in the activity of enzymes that maintain phospholipid homeostasis can facilitate pathological conditions resulting from membrane perturbations [129].
Mutations in the PLA2G6 gene which encodes Ca2+ dependent phospholipase A [iPLA2] have been linked to infantile neuroaxonal dystrophy which is characterized by motor and sensory impairment [130]. iPLA2 works to breakdown PC, generating free fatty acids and lysoPC and serves as an antagonist to Pcyt1, which is a key enzyme in PC production [131]. Imbalances between the activity of iPLA2 and Pcyt1 can lead to lipid imbalances which are detrimental for membrane integrity [132]. One of the lipids that is most often liberated by iPLA2 is arachidonic acid [133, 134], an eicosanoid precursor which can facilitate PLD activation leading to the activation of subsequent signaling cascades [135]. Moreover, excessive iPLA2 mediated arachidonic acid release can lead to 4-hydroxy-2-nonenal [4-HNE] production [136]. 4-HNE is a peroxidized product of arachidonic acid which can have a significant impact on cell functions by forming protein adducts and neuroaxonal dystrophy [137, 138]. Additionally, a subset of individuals with mutations in the PLA2G6 gene develops brain iron deposits and abnormal EMG readings likely resulting from axonal swelling and deterioration [139]. The mechanism for brain iron accumulation is unclear but it is likely due to many factors from the dysfunction of proteins involved with iron transport and storage [140] and the ability for iron to participate in the Fenton reaction to generate free radicals from products of mitochondrial respiration [141].
Conclusion
Choline is implicated in many biological processes like phospholipid biogenesis, FA oxidation, pregnancy and neurological development. However, a large proportion of the North American population is lacking in dietary choline consumption which is implicated in fatty liver, insulin resistance and obesity. As a result, it is imperative to increase choline consumption to support early life development and to help diminish the prevalence of adult onset metabolic disorders. Recommended intakes of the nutrients required for optimal health typically focus on a small number of nutrients. However, in reality, all nutrients are required in adequate amounts for optimal health, including nutrients such as choline which are often overlooked in the Western diet. The most important point of this review is that dietary choline deficiency is rampant in the Western world, and this can have widespread deleterious metabolic consequences.
References
1. Awwad HM, Ohlmann CH, Stoeckle M, Aziz R, Geisel J, Obeid R, Choline-phospholipids inter-conversion is altered in elderly patients with prostate cancer. Biochimie, 2016. 126: p. 108-114.
2. Cho E, Zeisel SH, Jacques P, Selhub J, Dougherty L, Colditz GA, et al; Dietary choline and betaine assessed by food-frequency questionnaire in relation to plasma total homocysteine concentration in the Framingham Offspring Study. Am J Clin Nutr, 2006. 83(4): p. 905-911.
3. Bidulescu A, Chambliss LE, Siega-Ruiz AM, Zeisel SH, Heiss G, Usual choline and betaine dietary intake and incident coronary heart disease: the Atherosclerosis Risk in Communities (ARIC) study. BMC Cardiovasc Disord, 2007. 7(20): p. 1-8.
4. Bidulescu A, Chambliss LE, Siega-Ruiz AM, Zeisel SH, Heiss G, Repeatability and measurement error in the assessment of choline and betaine dietary intake: the Atherosclerosis Risk in Communities (ARIC) study. Nutr J, 2009. 8(14): p. 1-6.
5. Cho E, Willett WC, Colditz GA, Fuchs CS, Wu K, Chan AT, et al; Dietary choline and betaine and the risk of distal colorectal adenoma in women. J Natl Cancer Inst, 2007. 99(16): p. 1224-1231.
6. Richard C, Lewis ED, Zhao YY, Asomaning J, Jacobs RL, Field CJ, et al; Measurement of the total choline content in 48 commercial dairy products or dairy alternatives. J Food Compost Anal, 2016. 45: p. 1-8.
7. Asomaning J, Zhao YY, Lewis ED, Wu J, Jacobs RL, Field CJ, et al; The development of a choline rich cereal based functional food: Effect of processing and storage. LWT, 2017. 75: p. 447-425.
8. de Veth MJ, Artegoitia VM, Campagna SR, Lapierre H, Harte F, Girard CL, Choline absorption and evolution of bioavailability markers when supplementing choline to lactating dairy cows. J Dairy Sci, 2016. 99(12): p. 9732-9744.
9. Savoini G, Agazzi A, Invernizzi G, Cattaneo D, Pinotti L, Baldi A, Polyunsaturated fatty acids and choline in dairy goats nutrition: Production and health benefits. Small Rumin Res, 2010. 88: p. 135-144.
10. Mehta R, Birendinc A, Younossi ZM, Host Genetic Variants in Obesity-Related Nonalcoholic Fatty Liver Disease. Clin Liver Dis, 2014. 18(1): p. 249-267.
11. Gao X, Wang Y, Sun G, High dietary choline and betaine intake is associated with low insulin resistance in the Newfoundland population. Nutrition, 2017. 33: p. 28-34.
12. Michel V, Singh RK, Bakovic M, The impact of choline availability on muscle lipid metabolism. Food Funct, 2011. 2: p. 53-62.
13. Rinella ME, Green RM, The methionine-choline deficient dietary model of steatohepatitis does not exhibit insulin resistance. J Hepatol, 2004. 40(1): p. 47-51.
14. Payne F, Lim K, Girousse A, Brown RJ, Kory N, Robbins A, et al; Mutations disrupting the Kennedy phosphatidylcholine pathway in humans with congenital lipodystrophy and fatty liver disease. Proc Natl AcadSci U S A, 2014. 111(24): p. 8901-8906.
15. Velazquez, R., Ash, J.A., Powers, B.E., Kelley, C.M., Strawderman, M., Luscher, Z.I., et al; Maternal choline supplementation improves spatial learning and adult hippocampal neurogenesis in the Ts65Dn mouse model of Down syndrome. Neurobiol Dis, 2013. 58: p. 92-101.
16. Niculescu MD, Zeisel SH, Diet, Methyl Donors and DNA Methylation: Interactions between Dietary Folate, Methionine and Choline. J Nutr, 2002. 132: p. 2333S-2335S.
17. Vance JE, Vance DE, Specific Pools of Phospholipids Are Used for Lipoprotein Secretion by Cultured Rat Hepatocytes. J Biol Chem, 1986. 261(10): p. 4486-4491.
18. Machova E, O’Regan S, Newcombe J, Meunier FM, Prentice J, Dove R, et al; Detection of choline transporter-like 1 protein CTL1 in neuroblastoma x glioma cells and in the CNS, and its role in choline uptake. J Neurochem, 2009. 110(4): p. 1297-1309.
19. Sundler R, Arvidson G, Akesson B, Pathways for the incorporation of choline into rat liver phosphatidylcholines in vivo. Biochim Biophys Acta, 1972. 280(4): p. 559-568.
20. Haddon HB, Vance DE, Tetradecanoyl-phorbol acetate stimulates phosphatidylcholine biosynthesis in HeLa cells by an increase in the rate of the reaction catalyzed by CTP:phosphocholine cytidylyltransferase. Biochim Biophys Acta, 1980. 620(3): p. 636-640.
21. Ridgway ND, Vance DE, Purification of Phosphatidylethanolamine N-methyltransferase from Rat Liver. J Biol Chem, 1987. 262(35): p.17231-17239.
22. Vance DE, Phospholipid methylation in mammals: from biochemistry to physiological function. Biochim Biophys Acta, 2014. 1838(6): p. 1477-1487.
23. Nishimaki-Mogami T, Yao Z, Fujimori K, Inhibition of phosphatidylcholine synthesis via the phosphatidylethanolamine methylation pathway impairs incorporation of bulk lipids into VLDL in cultured rat hepatocytes. J Lipid Res, 2002. 43: p. 1035-1045.
24. Jiang X, West AA. Caudill M, Maternal choline supplementation: a nutritional approach for improving offspring health? Trends Endocrinol Metab, 2014. 25(5): p. 263-273.
25. Watkins SM, Zhu X, Zeisel SH, Phosphatidylethanolamine-N- methyltransferase activity and dietary choline regulate liver-plasma lipid flux and essential fatty acid metabolism in mice. J Nutr, 2003. 133(11): p. 3386-3391.
26. Lo Van A, Sakayori N, Hachem M, Belkouch M, Picq M, Lagarde M. et al.; Mechanisms of DHA transport to the brain and potential therapy to neurodegenerative diseases. Biochimie, 2016. 130: p. 163-167.
27. Thies F, Pillon C, Moliere P, Lagarde M, Lecerf, J, Preferential incorporation of sn-2 lysoPC DHA over unesterified DHA in the young rat brain. Am J Physiol, 1994. 267: p. 1273-1279.
28. Dorninger F, Broode A, Braverman NE, Moser AB, Just WW, Forss-Petter S. et al; Homeostasis of phospholipids – The level of phosphatidylethanolamine tightly adapts to changes in ethanolamine plasmalogens. Biochim Biophys Acta, 2015. 1851(2): p. 117-128.
29. Braverman NE, Moser AB, Functions of plasmalogen lipids in health and science. Biochim Biophys Acta, 2012. 1822(9): p. 1442-1452.
30. Foster DA, Phosphatidic acid and lipid-sensing by mTOR. Trends Endocrinol Metab, 2013. 24(6): p. 272-278.
31. Schenkel LC, Singh RK, Michel V, Zeisel SH, da Costa KA, Johnson AR. et al; Mechanism of choline deficiency and membrane alteration in postural orthostatic syndrome primary skin fibroblasts. FASEB J, 2015. 29(5): p. 1663-1675.
32. Vance JE, Tasseva, G, Formation and function of phosphatidylserine and phosphatidylethanolamine in mammalian cells. Biochim Biophys Acta, 2013. 1831(3): p. 543-554.
33. Wong ER, Thompson W, Choline oxidation and labile methyl groups in normal and choline-deficient rat liver. Biochim Biophys Acta, 1972. 260(2): p. 259-271.
34. Barak AJ, Beckenhauer HC, Tuma DJ, S-adenosylmethionine generation and prevention of alcoholic fatty liver by betaine. Alcohol, 1994. 11(6): p. 501-503.
35. Anstee QM, Day C, S-adenosylmethionine (SAMe) therapy in liver disease: A review of current evidence and clinical utility. J Hepatol, 2012. 57(5): p. 1097-1109.
36. Schenkel LC, Sivanesan S, Zhang J, Wuyts B, Taylor A, Verbrugghe A, et al; Choline supplementation restores substrate balance and alleviates complications of Pcyt2 deficiency. J Nutr Biochem, 2015. 26(11): p. 1221-1234.
37. Taylor A, Schenkel LC, Yokich M, Bakovic, M, Adaptations to excess choline in insulin resistant and deficient skeletal muscle. Biochem Cell Biol, 2016. 95(2): p. 223-231.
38. Sivanesan S, Taylor A, Zhang J, Bakovic, M, Betaine and Choline Improve Lipid Homeostasis in Obesity by Participation in Mitochondrial Oxidative Demethylation. Front Nutr, 2018.
39. Noureddin M, Mato JM, Lu SC, Nonalcoholic fatty liver disease: Update on pathogenesis, diagnosis, treatment and the role of S-adenosylmethionine. Exp Biol Med, 2015. 240(6): p. 809-820.
40. Seko Y, Sumida Y, Tanaka S, Mori K, Taketani H, Ishiba H, et al; Insulin resistance increases the risk of incident type 2 diabetes mellitus in patients with nonalcoholic fatty liver. Hepatol Res, 2018. 48(3): p. 42-51.
41. Toohey JI, Sulfur Amino Acids in Diet-induced Fatty Liver: A New Perspective Based on Recent Findings. Molecules, 2014. 19(6): p. 8334-8349.
42. Shim P, Choi D, Park Y, Association of Blood Fatty Acid Composition and Dietary Pattern with the Risk of Non-Alcoholic Fatty Liver Disease in Patients Who Underwent Cholecystectomy. Ann Nutr Metab, 2017. 70(4): p. 303-311.
43. Zeisel SH, Blusztajn JK, Choline and Human Nutrition. Annu Rev Nutr. 1994. 14: p. 269-296.
44. Obeid R, The Metabolic Burden of Methyl Donor Deficiency with Focus on the Betaine Homocysteine Methyltransferase Pathway. Nutrients, 2013. 5(9): p. 3481-3495.
45. Khaire A, Rathod R, Kale A, Joshi S, Vitamin B12 and omega-3 fatty acids together regulate lipid metabolism in Wistar rats. Prostaglandins, Leukot Essent Fatty Acids, 2015. 99: p. 7-17.
46. Begley HN, Wang Y, Campbell MS, Yu X, Lane RH, Joss-Moore LA, Maternal docosahexaenoic acid increases adiponectin and normalizes IUGR- induced changes in rat adipose deposition. J Obes, 2013.
47. Verbrugghe A, Bakovic M, Peculiarities of One-Carbon Metabolism in the Strict Carnivorous Cat and the Role in Feline Hepatic Lipidosis. Nutrients, 2013. 5(7): p. 2811-2835.
48. Pooya S, Blaise S, Garcia MM, Giudicelli J, Alberto JM, Gueant-Rodriguez RM, et al; Methyl donor deficiency impairs fatty acid oxidation through PGC-1α hypomethylation and decreased ER-α, ERR-α, and HNF-4α in the rat liver. J Hepatol, 2012. 57: p. 344-351.
49. Dahlhoff C, Worsch S, Saller M, Hummel BA, Fiamonici J, Uebel K, et al; Methyl-donor supplementation in obese mice prevents the progression of NAFLD, activates AMPK and decreases acyl-carnitine levels. Mol Metab, 2014. 3(5): p. 565-580.
50. Schofield Z, Reed MAC, Newsome PN, Adams DH, Gunther UL, Lalor PF, Changes in human hepatic metabolism in steatosis and cirrhosis. World J Gastroenterol, 2017. 23(15): p. 2685-2695.
51. Auta J, Zhang H, Pandey SC, Guidotti A, Chronic Alcohol Exposure Differentially Alters One-Carbon Metabolism in Rat Liver and Brain. Alcohol Clin Exp Res, 2017. 41(6): p. 1105-1111.
52. Liu HS, Lu P, Guo Y, Farrell E, Zhang X, Zheng M, et al; An integrative genomic analysis identifies Bhmt2 as a diet-dependent genetic factor protecting against acetaminophen-induced liver toxicity. Genome Res, 2010. 20(1): p. 28-35.
53. Robinson JL, Bertolo RF, The Pediatric Methionine Requirement Should Incorporate Remethylation Potential and Transmethylation Demands. Adv Nutr, 2016. 7(3): p. 523-534.
54. Cote-Robitaille ME, Girard CL, Guay F, Matte JJ, Oral supplementations of betaine, choline, creatine and vitamin B6 and their influence on the development of homocysteinaemia in neonatal piglets. J Nutr Sci, 2015. 4(31): p. 1-7.
55. Miettinen TP, Pessa HKJ, Caldez MJ, Fuhrer T, Diril MK, Sauer U, et al; Identification of Transcriptional and Metabolic Programs Related to Mammalian Cell Size. Curr Biol, 2014. 24(6): p. 598-608.
56. Ginzberg MB, Kafri R, Kirschner M, On being the right (cell) size. Science, 2015. 348(6236): p. 1-19.
57. Hoffmann L, Brauers G, Gehrmann T, Haussinger D, Mayatepek E, Schless F, et al; Osmotic regulation of hepatic betaine metabolism. Am J Physiol Gastrointest Liver Physiol, 2013. 304(9): p. G835-G846.
58. Ahn CW, Choi YJ, Hong SH, Jun DS, Na JD, Choi YJ, et al; Involvement of multiple pathways in the protection of liver against high-fat diet- induced steatosis by betaine. J Funct Foods, 2015. 17: p. 66-72.
59. Than NN, Newsome PN, A concise review of non-alcoholic fatty liver disease. Atherosclerosis, 2015. 239(1): p. 192-202.
60. Kathirvel E, Morgan K, Nandgiri G, Sandoval BC, Caudill MA, Bottiglieri T, et a; Betaine improves nonalcoholic fatty liver and associated hepatic insulin resistance: a potential mechanism for hepatoprotection by betaine. Am J Physiol Gastrointest Physiol, 2010. 299(5): p. 1068-1077.
61. Jager S, Handschin C, St-Pierre J, Spiegelman BM, AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1α. Proc Natl Acad Sci U S A, 2007. 104(29): p. 12017-12022.
62. Buzzetti E, Pinzani M, Tsochatzis EA, The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism, 2016. 65(8): p. 1038-1048.
63. Hirsch E, Costa C, Ciraolo E, Phosphoinositide 3-kinases as a common platform for multi-hormone signaling. J Endocrinol, 2007. 194(2): p. 243-256.
64. Longo N, Frigeni M, Pasquali M, Carnitine transport and fatty acid oxidation. Biochim Biophys Acta, 2016. 1863(10): p. 2422-2435.
65. ter Veld F, Primassin S, Hoffmann L, Mayatepek E, Spiecherkoetter U, Corresponding increase in long-chain acyl-CoA and acylcarnitine after exercise in muscle from VLCAD mice. J Lipid Res, 2009. 50(8): p. 1556-1562.
66. Neess D, Bek S, Engelsby H, Gallego SF, Faergeman NJ, Long- chain acyl-CoA esters in metabolism and signaling: Role of acyl-CoA binding proteins. Prog Lipid Res, 2015. 59: p. 1-25.
67. King JH, Kwan ST, Yan J, Klatt KC, Jiang X, Roberson MS, et al; Maternal Choline Supplementation Alters Fetal Growth Patterns in a Mouse Model of Placental Insufficiency. Nutrients, 2017. 9: p. 1-16.
68. Nam J, Greenwald E, Jack-Roberts C, Ajeeb TT, Malysheva OV, Caudill MA, et al; Choline prevents fetal overgrowth and normalizes placental fatty acid and glucose metabolism in a mouse model of maternal obesity. J Nutr Biochem, 2017. 49: p. 80-88.
69. Yan J, Jiang X, West AA, Perry CA, Malysheva OV, Devapatla S, et al; Maternal choline intake modulates maternal and fetal biomarkers of choline metabolism in humans. Am J Clin Nutr, 2012. 95(5): p. 1060-1071.
70. Yan J, Jiang X, West AA, Perry CA, Malysheva OV, Brenna JT, et al; Pregnancy alters choline dynamics: results of a randomized trial using stable isotope methodology in pregnant and nonpregnant women. Am J Clin Nutr, 2013. 98(6): p. 1459- 1467.
71. Molloy AM, Mills JL, Cox C, Daly SF, Conle, M, Brody LC, et al; Choline and homocysteine interrelations in umbilical cord and maternal plasma at delivery. Am J Clin Nutr, 2005. 82(4): p. 836-842.
72. Velzing-Aarts FV, Holm PI, Fokkema MR, Van Der Dijs FP, Ueland PM, Muskiet FA, Plasma choline and betaine and their relation to plasma homocysteine in normal pregnancy. Am J Clin Nutr, 2005. 81(6): p. 1383-1389.
73. Brosnan JT, Brosnan ME, The Sulfur-Containing Amino Acids: An Overview. J Nutr, 2006. 136: p. 1636-1640.
74. Ganz AB, Cohen VV, Swersky CC, Stover J, Vitiello GA, Lovesky J, et al; Genetic Variation in Choline-Metabolizing Enzymes Alters Choline Metabolism in Young Women Consuming Choline Intakes Meeting Current Recommendations. Int J Mol Sci, 2017. 18: p. 1-23.
75. Jadavji NM, Deng L, Malysheva O, Caudill MA, Rozen R, MTHFR deficiency or reduced intake of folate or choline in pregnant mice results in impaired short-term memory and increased apoptosis in the hippocampus of wild-type offspring. Neuroscience, 2015. 300: p. 1-9.
76. da Costa KA, Kozyreva OG, Song J, Golanko JA, Fischer LM, Zeisel SH, Common genetic polymorphisms affect the human requirement for the nutrient choline. FASEB J, 2006. 20(9): p.1336-1344.
77. Kohlmeier M, da Costa KA, Fischer LM, Zeisel SH, Genetic variation of folate-mediated one-carbon transfer pathway predicts susceptibility to choline deficiency in humans. Proc Natl Acad Sci U S A, 2005. 102(44): p. 16025-16030.
78. Jensen HH, Batres-Marquez SP, Carriquiry A, Schalinske KL, Choline in the diets of the U.S. population: NHANES, 2003-2004. FASEB J, 2007. 21: p. 219.
79. Provencal N, Binder EB, The effects of early life stress on the epigenome: From the womb to adulthood and even before. Exp Neurol, 2015. 268: p. 10-20.
80. Smith ZD, Meissner A, DNA methylation: roles in mammalian development. Nat Rev Genet, 2013. 14(3): p. 204-220.
81. Schneider E, Dittrich M, Bock J, Nanda I, Muller T, Seidmann L, et al; CpG sites with continuously increasing or decreasing methylation from early to late human fetal brain development. Gene, 2016. 592(1): p. 110-118.
82. Hala D, Huggett DB, Burggren WW, Environmental stressors and the epigenome. Drug Discov Today Technol, 2014. 12: p. 3-8.
83. Haggarty P, Nutrition and the Epigenome. Prog Mol Biol Trans Sci, 2012. 108: p. 427-446.
84. Kovacheva VP, Mellott TJ, Davison JM, Wagner N, Lopez-Coviella, I, Schnitzler AC, et al; Gestational Choline Deficiency Causes Global and Igf2 Gene DNA Hypermethylation by Up-regulation of Dnmt1 Expression. J Biol Chem, 2007. 282(43): p. 31777-31888.
85. Agaoglu OK, Agaoglu AR, Guzeloglu A, Aslan S, Kurar E, Kayis SA, et al; Gene expression profiles of some cytokines, growth factors, receptors, and enzymes (GM-CSF, IFNγ, MMP-2, IGF-II, EGF, TGF-β, IGF-IIR) during pregnancy in the cat uterus. Theriogenology, 2016. 85(4): p. 638-644.
86. Borodovitsyna O, Flamini M, Chandler D, Noradrenergic Modulation of Cognition in Health and Disease. Neural Plast, 2017. 6031478
87. da Costa KA, Rai KS, Craciunescu CN, Parikh K, Mehedint MG, Sanders LM, et al; Dietary Docosahexaenoic Acid Supplementation Modulates Hippocampal Development in the Pemt-/- Mouse. J Biol Chem, 2009. 285(2): p. 1008-1015.
88. da Costa KA, Sanders LM, Fischer LM, Zeisel SH, Docosahexaenoic acid in plasma phosphatidylcholine may be a potential marker for in vivo phosphatidylethanolamine N-methyltransferase activity in humans. Am J Clin Nutr, 2011. 93(5): p. 968-974.
89. Nyaradi A, Li J, Hickling S, Foster J, Oddy WH, The role of nutrition in children's neurocognitive development, from pregnancy through childhood. Front Hum Neurosci, 2013. 7(97): p. 1-16.
90. Niculescu MD, Craciunescu CN, Zeisel SH, Dietary choline deficiency alter global and gene-specific DNA methylation in the developing hippocampus of mouse fetal brains. FASEB J, 2006. 20(1): p. 43-49.
91. Schulz KM, Pearson JN, Gasparrini ME, Brooks KF, Drake-Frazier C, Zajkowski ME, et al; Dietary choline supplementation to dams during pregnancy and lactation mitigates the effects of in utero stress exposure on adult anxiety-related behaviors. Behav Brain Res, 2014. 268: p. 104-110.
92. Hancock SE, Freidrich MG, Mitchell TW, Truscott RJW, Else PL, The phospholipid composition of the human entorhinal cortex remains relatively stable over 80 years of adult aging. GeroScience, 2017. 39(1): p. 73-82.
93. Boughter CT, Monje-Galvan V, Im W, Klauda JB, Influence of Cholesterol on Phospholipid Bilayer Structure and Dynamics. J Phys Chem, 2016. 120(45): p. 11761-11772.
94. Kolisnyk B, Al-Onaizi MA, Hirata PH, Guzman MS, Nikolova S, Barbash S, et al; Forebrain deletion of the vesicular acetylcholine transporter results in deficits in executive function, metabolic, and RNA splicing abnormalities in the prefrontal cortex. J Neurosci, 2013. 33(37): p. 14908-14920.
95. Wurtman RJ, How Anticholinergic Drugs Might Promote Alzheimer's Disease: More Amyloid-β and Less Phosphatidylcholine. J Alzheimers Dis, 2015. 46(4): p. 983-987.
96. Caudill MA, Strupp BJ, Muscalu L, Nevins JEH, Canfield RL, Maternal choline supplementation during the third trimester of pregnancy improves infant information processing speed: a randomized, double-blind, controlled feeding study. FASEB J, 2018. 32(4): p. 2172-2180.
97. Zhu C, Wu T, Jin Y, Huang B, Zhou R, Wang Y, et al; Prenatal choline supplementation attenuates spatial learning deficits of offspring rats exposed to low-protein diet during fetal period. J Nutr Biochem, 2016. 32: p. 163-170.
98. Craciunescu CN, Albright CD, Mar MH, Song J, Zeisel SH, Choline Availability During Embryonic Development Alters Progenitor Cell Mitosis in Developing Mouse Hippocampus. J Nutr, 2003. 133(11): p. 3614-3618.
99. Kwan ST, King JH, Yan J, Wang Z, Jiang X, Hutzler JS, et al; Maternal Choline Supplementation Modulates Placental Nutrient Transport and Metabolism in Late Gestation of Mouse Pregnancy. J Nutr, 2017. 147(11): p. 2083-2092.
100. DeLong CJ, Shen YJ, Thomas MJ, Cui Z, Molecular Distinction of Phosphatidylcholine Synthesis between the CDP-Choline Pathway and Phosphatidylethanolamine Methylation Pathway. J Nutr Biochem, 1999. 274(42): p. 29683-29688.
101. Resseguie M, Song J, Niculescu MD, da Costa KA, Randall TA, Zeisel SH, Phosphatidylethanolamine N-methyltransferase (PEMT) gene expression is induced by estrogen in human and mouse primary hepatocytes. FASEB J, 2007. 21(10): p. 2622-2632.
102. Fischer LM, da Costa KA, Kwock L, Stewart PW, Lu TS, Stabler SP, et al; Sex and menopausal status influence human dietary requirements for the nutrient choline. Am J Clin Nutr, 2017. 85(5): p. 1275-1285.
103. Sundrani D, Khot V, Joshi S, DNA Methylation for Prediction of Adverse Pregnancy Outcomes. In: Epigenetic Biomarkers and Diagnostics. San Diego, California, United States: Academic Press; 2016. p. 351-376.
104. Kulharni A, Dangat K, Kale A, Sable P, Chavan-Gautam P, Josh, S, Effects of Altered Maternal Folic Acid, Vitamin B12 and Docosahexaenoic Acid on Placental Global DNA Methylation Patterns in Wistar Rats. PLoS ONE, 2011. 6: e17706.
105. Bahous RH, Cosin-Tomas M, Deng L, Leclerc D, Malysheva O, Ho MK, et al; Early Manifestations of Brain Aging in Mice Due to Low Dietary Folate and Mild MTHFR Deficiency. Mol Neurobiol, 2019. 56(6): p. 4175-4191.
106. Dorninger F, Moser AB, Kou J, Weisinger C, Forss-Petter S, Gleiss A, et al; Alterations in the Plasma Levels of Specific Choline Phospholipids in Alzheimer’s Disease Mimic Accelerated Aging. J Alzheimers Dis, 2018. 62(2): p. 841-854.
107. Lin W, Zhang J, Liu Y, Wu R, Yang H, Hu X. et al; Studies on diagnostic biomarkers and therapeutic mechanism of Alzheimer’s disease through metabolomics and hippocampal proteomics. Eur J Pharm Sci, 2017. 15(105): p. 119-126.
108. van Wijk N, Slot RER, Duits FH, Strik M, Biesheuvel E, Sijben JWC, et al; Nutrients required for phospholipid synthesis are lower in blood and cerebrospinal fluid in mild cognitive impairment and Alzheimer's disease dementia. Alzheimers Dement, 2017. 16(8): p. 139-146.
109. Mellott TJ, Huleatt OM, Shade BN, Pender SM, Liu YB, Slack BE, et al; Perinatal Choline Supplementation Reduces Amyloidosis and Increases Choline Acetyltransferase Expression in the Hippocampus of the APPswePS1dE9 Alzheimer's Disease Model Mice. PLoS One, 2017. 12(1): e0170450.
110. Nickerson CA, Brown AL, Yu W, Chun Y, Glenn MJ, Prenatal choline supplementation attenuates MK-801-induced deficits in memory, motor function, and hippocampal plasticity in adult male rats. Neuroscience, 2017. 361: p. 116-128.
111. Alldred MJ, Chao HM, Lee SH, Beilin J, Powers BE, Petkova E, et al; CA1 pyramidal neuron gene expression mosaics in the Ts65Dn murine model of Down syndrome and Alzheimer's disease following maternal choline supplementation. Hippocampus, 2018. 28(4): p.251-268.
112. Powers BE, Kelley CM, Velazquez R, Ash JA, Strawderman MS, Alldred MJ, et al; Maternal choline supplementation in a mouse model of Down syndrome: Effects on attention and nucleus basalis/substantia innominata neuron morphology in adult offspring. Neuroscience, 2017. 340: p. 501-514.
113. Iulia C, Ruxandra T, Costin LB, Liliana-Mary V, Citicoline - a neuroprotector with proven effects on glaucomatous disease. Rom J Ophthalmol, 2017. 61(3): p. 152-158.
114. Roberti G, Tanga L, Michelessi M, Quaranta L, Parisi V, Manni G, et al; Cytidine 5'-Diphosphocholine (Citicoline) in Glaucoma: Rationale of Its Use, Current Evidence and Future Perspectives. Int J Mol Sc, 2015. 16(12): p. 28401-28417.
115. Thies F, Delachambre MC, Bentejac M, Lagarde M, Lecerf, J, Unsaturated Fatty Acids Esterified in 2-Acyl-1-Lysophosphatifylcholine Bound to Albumin Are More Efficiency Taken up by the Young Rat Brain than the Unesterifed Form. J Neurochem, 1992. 59(3): p. 1110-1116.
116. Lagarde M, Hachem M, Bernoud-Hubac N, Picq M, Vericel E, Guichardant M, Biological properties of a DHA-containing structured phospholipid (AceDoPC) to target the brain. Prostaglandins Leukot Essent Fatty Acids, 2015. 92: p. 63-65.
117. Tamura A, Tanaka T, Yamane T, Nasu R, Fujii T, Quantitative studies on translocation and metabolic conversion of lysophosphatidylcholine incorporated into the membrane of intact human erythrocytes from the medium. J Biochem, 1985. 97(1): p. 353-359.
118. Maligan JM, Estiasih T, Kusnadi J, Structured Phospholipids from Commercial Soybean Lecithin Containing Omega-3 Fatty Acids Reduces Atheroslcerosis Risk in Male Sprague Dawley Rats Which Fed with an Atherogenic Diet. WASET, 2012. 6(9): p. 731-737.
119. Murru E, Banni S, Carta G, Nutritional properties of dietary omega-3- enriched phospholipids. Biomed Res Int, 2013.
120. Fernandes MF, Mutch DM, Leri F, The Relationship between Fatty Acids and Different Depression-Related Brain Regions, and Their Potential Role as Biomarkers of Response to Antidepressants. Nutrients, 2017. 9(3).
121. Schonfeld P, Reiser G, Brain energy metabolism spurns fatty acids as fuel due to their inherent mitotoxicity and potential capacity to unleash neurodegeneration. Neurochem Int, 2017. 109: p. 68-77.
122. Schuschardt JP, Huss M, Stauss-Grabo M, Hahn A, Significance of long-chain polyunsaturated fatty acids (PUFAs) for the development and behaviour of children. Eur J Pediatr, 2010. 169(2): p. 149-164.
123. Calder PC, Polyunsaturated fatty acids and inflammatory processes: New twists in an old tale. Biochimie, 2009. 91(6): p. 791-795.
124. Bazan NG, Molina MF, Gordon WC, Docosahexaenoic acid signalolipidomics in nutrition: significance in aging, neuroinflammation, macular degeneration, Alzheimer’s, and other neurodegenerative diseases. Annu Rev Nutr, 2011. 31: p. 321-351.
125. Li Q, Zhang Q, Wang M, Liu F, Zhao S, Ma J. et al; Docosahexaenoic acid affects endothelial nitric oxide synthase in caveolae. Arch Biochem Biophys, 2007. 466(2): p. 250-259.
126. Ma DW, Lipid mediators in membrane rafts are important determinants of human health and disease. Appl Physiol Nutr Metab, 2007. 32(3): p. 341-350.
127. Pifferi F, Jouin M, Alessandri JM, Haedke U, Roux F, Perriere, N, et al; n-3 Fatty acids modulate brain glucose transport in endothelial cells of the blood-brain barrier. Prostaglandins Leukot Essent Fatty Acids, 2007. 77(5-6): p. 279-286.
128. Pifferi F, Jouin M, Alessandri JM, Roux F, Perriere N, Langelier B, et al; n-3 long-chain fatty acids and regulation of glucose transport in two models of rat brain endothelial cells. Neurochem Int, 2010. 56(5): p. 703-710.
129. Murakami M, Taketomi Y, Miki Y, Sato H, Hirabayashi T, Yamamoto K, Recent progress in phospholipase A2 research: from cells to animals to humans. Prog Lipid Res, 2011. 50(2): p. 152-192.
130. Strokin M, Reiser G, Mitochondria from a mouse model of the human infantile neuroaxonal dystrophy (INAD) with genetic defects in VIA iPLA2 have disturbed Ca (2+) regulation with reduction in Ca(2+) capacity. Neurochem Int, 2016. 99: p. 187-193.
131. Morgan NV, Westaway SK, Morton JE, Gregory A, Gissen P, Sonek, S. et al; PLA2G6, encoding a phospholipase A2, mutated in neurodegenerative disorders with high brain iron. Nat Genet, 2006. 38(7): p.752-754.
132. Baburina I, Jackowski S, Cellular responses to excess phospholipid. J Biol Chem, 1999. 274(14): p. 9400-9408.
133. Farooqui AA, Ong WY, Horrocks LA, Farooqui T, Brain Cytosolic Phospholipase A2: Localization, Role, and Involvement in Neurological Diseases. Neuroscientist, 2000. 6(3): p. 169-180.
134. Klein J, Chalifa V, Liscovitsch M, Loffelholz K, Role of phospholipase D activation in nervous system physiology and pathophysiology. J Neurochem, 1995. 65(4): p. 1445-1455.
135. Tamango E, Robino G, Obbili A, Bardini P, Aragno M, Parola M. et al; H2O2 and 4-hydroxynonenal mediate amyloid beta-induced neuronal apoptosis by activating JNKs and p38MAPK. Exp Neurol, 2003. 180(2): p. 144-155.
136. Gegotek A, Skrzydlewska E, Biological effect of protein modifications by lipid peroxidation products. Chem Phys Lipids, 2019. 221: p. 46-52.
137. Beck G, Sugiura Y, Shinzawa K, Kato S, Setou M, Tsujimoto Y. et al; Neuroaxonal dystrophy in calcium-independent phospholipase A2β deficiency results from insufficient remodeling and degeneration of mitochondrial and presynaptic membranes. J Neurosci, 2011. 31(31): p. 11411-11420.
138. Carrilho I, Santos M, Guimaraes A, Teixeira J, Chorao R, Martins M. et al; Infantile neuroaxonal dystrophy: what’s most important for the diagnosis? Eur J Paediatr Neurol, 2008. 12(6): p. 491-500.
139. Colombelli C, Aoun M, Tiranti V, Defective lipid metabolism in neurodegeneration with brain iron accumulation (NBIA) syndromes: not only a matter of iron. J Inherit Metab Dis, 2015. 38(1): p. 123-136.
140. Dusek P, Schneider SA, Aaseth J, Iron chelation in the treatment of neurodegenerative diseases. J Trace Elem Med Biol, 2016. 38: p. 81-92.
141. Lv H, Shang P, The significance, trafficking and determination of labile iron in cytosol, mitochondria and lysosomes. Metallomics, 2018. 10(7): p. 899-916.
PEER REVIEW
Not commissioned. Externally peer reviewed.
References
The Roles of Choline in Maintaining Optimal Health
The Effects of Intake of Bread with Treated Corn Bran Inclusion on Postprandial Glyceamic Response
Food Waste throughout the Food Production Continuum – Water Food and Energy Nexus
Use of Lightly Potassium-Enriched Soy Sauce at Home Reduced Urinary Sodium-to-Potassium Ratio
A Different Type of Critical Migration
PRECEDE: A Conceptual Model to Assess Immigrant Health
Walking Together: Supporting Indigenous Student Success in University
Cosmetic Surgery and Body Image in Race/Ethnic Minorities
Our articles most useful
Adrian Taylor and Marica Bakovic*
Published : June 21, 2019
Journal of Food & Nutritional Sciences
Zhimin Cui, Lynne Kennedy, Weili Li*
Published : September 30, 2019
Journal of Food & Nutritional Sciences
Elena Castell-Perez*, Rosana G. Moreira, Hal S. Knowles, III
Published : October 01, 2019
Journal of Food & Nutritional Sciences
Nagako Okuda, Makoto Miura, Kazuyoshi Itai, Takuya Morikawa, Junko Sasaki, Tamami Asanuma, Mikako Fujii, Akira Okayama
Published : March 27, 2019
Journal of Food & Nutritional Sciences