51
|
Idrus NM, Breit KR, Thomas JD. Dietary choline levels modify the effects of prenatal alcohol exposure in rats. Neurotoxicol Teratol 2017; 59:43-52. [PMID: 27888055 PMCID: PMC5770193 DOI: 10.1016/j.ntt.2016.11.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 01/21/2023]
Abstract
Prenatal alcohol exposure can cause a range of physical and behavioral alterations; however, the outcome among children exposed to alcohol during pregnancy varies widely. Some of this variation may be due to nutritional factors. Indeed, higher rates of fetal alcohol spectrum disorders (FASD) are observed in countries where malnutrition is prevalent. Epidemiological studies have shown that many pregnant women throughout the world may not be consuming adequate levels of choline, an essential nutrient critical for brain development, and a methyl donor. In this study, we examined the influence of dietary choline deficiency on the severity of fetal alcohol effects. Pregnant Sprague-Dawley rats were randomly assigned to receive diets containing 40, 70, or 100% recommended choline levels. A group from each diet condition was exposed to ethanol (6.0g/kg/day) from gestational day 5 to 20 via intubation. Pair-fed and ad lib lab chow control groups were also included. Physical and behavioral development was measured in the offspring. Prenatal alcohol exposure delayed motor development, and 40% choline altered performance on the cliff avoidance task, independent of one another. However, the combination of low choline and prenatal alcohol produced the most severe impairments in development. Subjects exposed to ethanol and fed the 40% choline diet exhibited delayed eye openings, significantly fewer successes in hindlimb coordination, and were significantly overactive compared to all other groups. These data suggest that suboptimal intake of a single nutrient can exacerbate some of ethanol's teratogenic effects, a finding with important implications for the prevention of FASD.
Collapse
Affiliation(s)
- Nirelia M Idrus
- Department of Psychology, Center for Behavioral Teratology, San Diego State University, San Diego, CA 92120, USA
| | - Kristen R Breit
- Department of Psychology, Center for Behavioral Teratology, San Diego State University, San Diego, CA 92120, USA
| | - Jennifer D Thomas
- Department of Psychology, Center for Behavioral Teratology, San Diego State University, San Diego, CA 92120, USA.
| |
Collapse
|
52
|
Jaiswal SK, Sukla KK, Chauhan A, Lakhotia AR, Kumar A, Rai AK. Choline metabolic pathway gene polymorphisms and risk for Down syndrome: An association study in a population with folate-homocysteine metabolic impairment. Eur J Clin Nutr 2016; 71:45-50. [PMID: 27677362 DOI: 10.1038/ejcn.2016.190] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 08/23/2016] [Accepted: 08/26/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND/OBJECTIVES Choline is an essential nutrient involved in one-carbon metabolism, but its role in mechanisms underlying meiotic non-disjunction is poorly known. The relationship between folate-homocysteine metabolic pathway gene polymorphism and Down syndrome (DS) risk has been widely analyzed, but there are limited reports on its correlation with choline metabolism. In the present case-control association study, we investigated the relationship of three single-nucleotide polymorphisms (SNPs) (phosphatidylethanolamine N-methyltransferase (PEMT) rs12325817, choline dehydrogenase (CHDH) rs12676 and homocysteine methyltransferase (BHMT) rs3733890) of choline metabolism with risk for DS. SUBJECT/METHODS Genotyping of 228 mothers of a down syndrome child (DSM) and 200 control mothers (CMs) for all SNPs was performed by PCR coupled with restriction fragment length polymorphism method. RESULTS A significantly increased risk for BHMT +742AA genotype with an odds ratio of 4.96 (95% confidence interval (CI): 1.66-14.88, P=0.0036) was observed. For PEMT rs12325817 and CHDH rs12676, no significant difference in allelic and genotypic frequencies was observed. In genotypic combination analysis considering PEMT -744GG/CHDH +432GG/BHMT +742GG as the reference combination, PEMT -744GC/CHDH +432GG/BHMT +742GG genotypic combination was significantly higher in DSM compared with that in CMs with an odds ratio of 2.061 (95% CI: 1.10-3.86, P=0.0342). We also observed an epistatic interaction between methylenetetrahydrofolate reductase (MTHFR) rs1801133 and choline metabolic pathway gene variants. CONCLUSIONS Our findings indicate impaired choline metabolism showing a greater risk for DS, especially in a population associated with homocysteine-folate impairment. Further studies are required to confirm our findings.
Collapse
Affiliation(s)
- S K Jaiswal
- Centre for Genetic Disorders, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - K K Sukla
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - A Chauhan
- Applied Microbiology, Department of Botany, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - A R Lakhotia
- Department of Gynecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - A Kumar
- Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - A K Rai
- Centre for Genetic Disorders, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
53
|
Dietary choline and betaine intake, choline-metabolising genetic polymorphisms and breast cancer risk: a case-control study in China. Br J Nutr 2016; 116:961-8. [PMID: 27488260 DOI: 10.1017/s0007114516002956] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Choline and betaine are essential nutrients involved in one-carbon metabolism and have been hypothesised to affect breast cancer risk. Functional polymorphisms in genes encoding choline-related one-carbon metabolism enzymes, including phosphatidylethanolamine N-methyltransferase (PEMT), choline dehydrogenase (CHDH) and betaine-homocysteine methyltransferase (BHMT), have important roles in choline metabolism and may thus interact with dietary choline and betaine intake to modify breast cancer risk. This study aimed to investigate the interactive effect of polymorphisms in PEMT, BHMT and CHDH genes with choline/betaine intake on breast cancer risk among Chinese women. This hospital-based case-control study consecutively recruited 570 cases with histologically confirmed breast cancer and 576 age-matched (5-year interval) controls. Choline and betaine intakes were assessed by a validated FFQ, and genotyping was conducted for PEMT rs7946, CHDH rs9001 and BHMT rs3733890. OR and 95 % CI were estimated using unconditional logistic regression. Compared with the highest quartile of choline intake, the lowest intake quartile showed a significant increased risk of breast cancer. The SNP PEMT rs7946, CHDH rs9001 and BHMT rs3733890 had no overall association with breast cancer, but a significant risk reduction was observed among postmenopausal women with AA genotype of BHMT rs3733890 (OR 0·49; 95 % CI 0·25, 0·98). Significant interactions were observed between choline intake and SNP PEMT rs7946 (P interaction=0·029) and BHMT rs3733890 (P interaction=0·006) in relation to breast cancer risk. Our results suggest that SNP PEMT rs7946 and BHMT rs3733890 may interact with choline intake on breast cancer risk.
Collapse
|
54
|
|
55
|
Ganz AB, Shields K, Fomin VG, Lopez YS, Mohan S, Lovesky J, Chuang JC, Ganti A, Carrier B, Yan J, Taeswuan S, Cohen VV, Swersky CC, Stover JA, Vitiello GA, Malysheva OV, Mudrak E, Caudill MA. Genetic impairments in folate enzymes increase dependence on dietary choline for phosphatidylcholine production at the expense of betaine synthesis. FASEB J 2016; 30:3321-3333. [PMID: 27342765 DOI: 10.1096/fj.201500138rr] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 06/14/2016] [Indexed: 01/15/2023]
Abstract
Although single nucleotide polymorphisms (SNPs) in folate-mediated pathways predict susceptibility to choline deficiency during severe choline deprivation, it is unknown if effects persist at recommended intakes. Thus, we used stable isotope liquid chromatography-mass spectrometry (LC-MS) methodology to examine the impact of candidate SNPs on choline metabolism in a long-term, randomized, controlled feeding trial among pregnant, lactating, and nonpregnant (NP) women consuming 480 or 930 mg/d choline (22% as choline-d9, with d9 indicating a deuterated trimethyl amine group) and meeting folate-intake recommendations. Variants impairing folate metabolism, methylenetetrahydrofolate reductase (MTHFR) rs1801133, methionine synthase (MTR) rs1805087 [wild-type (WT)], MTR reductase (MTRR) rs1801394, and methylenetetrahydrofolate dehydrogenase-methenyltetrahydrofolate cyclohydrolase-formyltetrahydrofolate synthetase (MTHFD1) rs2236225, influenced choline dynamics, frequently through interactions with reproductive state and choline intake, with fewer genotypic alterations observed among pregnant women. Women with these variants partitioned more dietary choline toward phosphatidylcholine (PC) biosynthesis via the cytidine diphosphate (CDP)-choline pathway at the expense of betaine synthesis even when use of betaine as a methyl donor was increased. Choline intakes of 930 mg/d restored partitioning of dietary choline between betaine and CDP-PC among NP (MTHFR rs1801133 and MTR rs1805087 WT) and lactating (MTHFD1 rs2236225) women with risk genotypes. Overall, our findings indicate that loss-of-function variants in folate-metabolizing enzymes strain cellular PC production, possibly via impaired folate-dependent phosphatidylethanolamine-N-methyltransferase (PEMT)-PC synthesis, and suggest that women with these risk genotypes may benefit from choline intakes exceeding current recommendations.-Ganz, A. B., Shields, K., Fomin, V. G., Lopez, Y. S., Mohan, S., Lovesky, J., Chuang, J. C., Ganti, A., Carrier, B., Yan, J., Taeswuan, S., Cohen, V. V., Swersky, C. C., Stover, J. A., Vitiello, G. A., Malysheva, O. V., Mudrak, E., Caudill, M. A. Genetic impairments in folate enzymes increase dependence on dietary choline for phosphatidylcholine production at the expense of betaine synthesis.
Collapse
Affiliation(s)
- Ariel B Ganz
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Kelsey Shields
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Vlad G Fomin
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Yusnier S Lopez
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Sanjay Mohan
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Jessica Lovesky
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Jasmine C Chuang
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Anita Ganti
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Bradley Carrier
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Jian Yan
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Siraphat Taeswuan
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Vanessa V Cohen
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Camille C Swersky
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Julie A Stover
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Gerardo A Vitiello
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Olga V Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Erika Mudrak
- Statistical Consulting Unit, Cornell University, Ithaca, New York, USA
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| |
Collapse
|
56
|
Robinson JL, Bertolo RF. The Pediatric Methionine Requirement Should Incorporate Remethylation Potential and Transmethylation Demands. Adv Nutr 2016; 7:523-34. [PMID: 27184279 PMCID: PMC4863267 DOI: 10.3945/an.115.010843] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The metabolic demand for methionine is great in neonates. Indeed, methionine is the only indispensable sulfur amino acid and is required not only for protein synthesis and growth but is also partitioned to a greater extent to transsulfuration for cysteine and taurine synthesis and to >50 transmethylation reactions that serve to methylate DNA and synthesize metabolites, including creatine and phosphatidylcholine. Therefore, the pediatric methionine requirement must accommodate the demands of rapid protein turnover as well as vast nonprotein demands. Because cysteine spares the methionine requirement, it is likely that the dietary provision of transmethylation products can also feasibly spare methionine. However, understanding the requirement of methionine is further complicated because demethylated methionine can be remethylated by the dietary methyl donors folate and betaine (derived from choline). Intakes of dietary methyl donors are highly variable, which is of particular concern for newborns. It has been demonstrated that many populations have enhanced requirements for these nutrients, and nutrient fortification may exacerbate this phenomenon by selecting phenotypes that increase methyl requirements. Moreover, higher transmethylation rates can limit methyl supply and affect other transmethylation reactions as well as protein synthesis. Therefore, careful investigations are needed to determine how remethylation and transmethylation contribute to the methionine requirement. The purpose of this review is to support our hypothesis that dietary methyl donors and consumers can drive methionine availability for protein synthesis and transmethylation reactions. We argue that nutritional strategies in neonates need to ensure that methionine is available to meet requirements for growth as well as for transmethylation products.
Collapse
Affiliation(s)
| | - Robert F Bertolo
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| |
Collapse
|
57
|
Kamada Y, Ebisutani Y, Kida S, Mizutani K, Akita M, Yamamoto A, Fujii H, Sobajima T, Terao N, Takamatsu S, Yoshida Y, Takehara T, Miyoshi E. Ectopic expression of N-acetylglucosaminyltransferase V accelerates hepatic triglyceride synthesis. Hepatol Res 2016; 46:E118-E129. [PMID: 26041473 DOI: 10.1111/hepr.12541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 05/24/2015] [Accepted: 06/02/2015] [Indexed: 01/28/2023]
Abstract
AIM Glycosylation changes induce various types of biological phenomena in human diseases. N-Acetylglucosaminyltransferase V (GnT-V) is one of the most important glycosyltransferases involved in cancer biology. Recently, many researchers have challenged studies of lipid metabolism in cancer. To elucidate the relationships between cancer and lipid metabolism more precisely, we investigated the effects of GnT-V on lipid metabolism. In this study, we investigated the effects of aberrant glycosylation by GnT-V on hepatic triglyceride production. METHODS We compared lipid metabolism in GnT-V transgenic (Tg) mice with that of wild-type (WT) mice fed with normal chow or a choline-deficient amino acid-defined (CDAA) diet in vivo. HepG2 cells and GnT-V transfectants of Hep3B cells were used in an in vitro study. RESULTS Serum triglyceride levels and hepatic very low-density lipoprotein (VLDL) secretion in Tg mice were significantly elevated compared with that of WT mice. Hepatic lipogenic genes (Lxrα, Srebp1, Fas and Acc) and VLDL secretion-related gene (Mttp1) were significantly higher in Tg mice. Expression of these genes was also significantly higher in GnT-V transfectants than in mock cells. Knockdown of GnT-V decreased, while both epidermal growth factor and transforming growth factor-β1 stimulation increased LXRα gene expression in HepG2 cells. Finally, we found that the blockade of VLDL secretion by CDAA diet induced massive hepatic steatosis in Tg mice. CONCLUSION Our study demonstrates that enhancement of hepatic GnT-V activity accelerates triglyceride synthesis and VLDL secretion. Glycosylation modification by GnT-V regulation could be a novel target for a therapeutic approach to lipid metabolism.
Collapse
Affiliation(s)
- Yoshihiro Kamada
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
- Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Japan
| | - Yusuke Ebisutani
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Sachiho Kida
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Kayo Mizutani
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Maaya Akita
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Akiko Yamamoto
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Hironobu Fujii
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Tomoaki Sobajima
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Naoko Terao
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Shinji Takamatsu
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Yuichi Yoshida
- Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Japan
| | - Tetsuo Takehara
- Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Japan
| | - Eiji Miyoshi
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| |
Collapse
|
58
|
Abstract
PURPOSE OF REVIEW The review highlights recent advances in our understanding of the interactions between genetic polymorphisms in genes that metabolize choline and the dietary requirements of choline and how these interactions relate to human health and disease. RECENT FINDINGS The importance of choline as an essential nutrient has been well established, but our appreciation of the interaction between our underlying genetic architecture and dietary choline requirements is only beginning. It has been shown in both human and animal studies that choline deficiencies contribute to diseases such as nonalcoholic fatty liver disease and various neurodegenerative diseases. An adequate supply of dietary choline is important for optimum development, highlighted by the increased maternal requirements during fetal development and in breast-fed infants. We discuss recent studies investigating variants in PEMT and MTHFR1 that are associated with a variety of birth defects. In addition to genetic interactions, we discuss several recent studies that uncover changes in fetal global methylation patterns in response to maternal dietary choline intake that result in changes in gene expression in the offspring. In contrast to the developmental role of adequate choline, there is now an appreciation of the role choline has in cardiovascular disease through the gut microbiota-mediated metabolite trimethylamine N-oxide. This pathway highlights some of our understanding of how the microbiome affects nutrient processing and bioavailability. Finally, to better characterize the genetic architecture regulating choline requirements, we discuss recent results focused on identifying polymorphisms that regulate choline and its derivative products. SUMMARY Here we discuss recent studies that have advanced our understanding of how specific alleles in key choline metabolism genes are related to dietary choline requirements and human disease.
Collapse
Affiliation(s)
- Tangi Smallwood
- Department of Genetics, University of North Carolina Chapel Hill, North Carolina 27599
| | - Hooman Allayee
- Department of Preventive Medicine and Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Brian J. Bennett
- Department of Genetics, University of North Carolina Chapel Hill, North Carolina 27599
- Nutrition Research Institute, University of North Carolina Kannapolis, North Carolina 28081
- Department of Nutrition, University of North Carolina Chapel Hill, North Carolina 27599
- Corresponding author: Brian J. Bennett, 500 Laureate Way, Suite 2303, Kannapolis NC 28081, Phone: 704-250-5044, Fax: 704-250-5000,
| |
Collapse
|
59
|
Beebe K, Kennedy AD. Sharpening Precision Medicine by a Thorough Interrogation of Metabolic Individuality. Comput Struct Biotechnol J 2016; 14:97-105. [PMID: 26929792 PMCID: PMC4744241 DOI: 10.1016/j.csbj.2016.01.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/07/2016] [Accepted: 01/10/2016] [Indexed: 12/24/2022] Open
Abstract
Precision medicine is an active component of medical practice today, but aspirations are to both broaden its reach to a greater diversity of individuals and improve its “precision” by enhancing the ability to define even more disease states in combination with associated treatments. Given complexity of human phenotypes, much work is required. In this review, we deconstruct this challenge at a high level to define what is needed to move closer toward these aspirations. In the context of the variables that influence the diverse array of phenotypes across human health and disease – genetics, epigenetics, environmental influences, and the microbiome – we detail the factors behind why an individual's biochemical (metabolite) composition is increasingly regarded as a key element to precisely defining phenotypes. Although an individual's biochemical (metabolite) composition is generally regarded, and frequently shown, to be a surrogate to the phenotypic state, we review how metabolites (and therefore an individual's metabolic profile) are also functionally related to the myriad of phenotypic influencers like genetics and the microbiota. We describe how using the technology to comprehensively measure an individual's biochemical profile – metabolomics – is integrative to defining individual phenotypes and how it is currently being deployed in efforts to continue to elaborate on human health and disease in large population studies. Finally, we summarize instances where metabolomics is being used to assess individual health in instances where signatures (i.e. biomarkers) have been defined. Untargeted biochemical profiling has the potential to phenotype individuals where genetic associations do not seem to show penetrance Metabolomics can be leveraged with other ‘omics data to discern phenotype information that is driven by environmental, microbiota, or epigenetic factors. Tracking the biochemical profile of individuals may help discern effectiveness or response to treatment or disease progression.
Collapse
|
60
|
Nilsson TK, Hurtig-Wennlöf A, Sjöström M, Herrmann W, Obeid R, Owen JR, Zeisel S. Plasma 1-carbon metabolites and academic achievement in 15-yr-old adolescents. FASEB J 2016; 30:1683-8. [PMID: 26728177 DOI: 10.1096/fj.15-281097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/14/2015] [Indexed: 11/11/2022]
Abstract
Academic achievement in adolescents is correlated with 1-carbon metabolism (1-CM), as folate intake is positively related and total plasma homocysteine (tHcy) negatively related to academic success. Because another 1-CM nutrient, choline is essential for fetal neurocognitive development, we hypothesized that choline and betaine could also be positively related to academic achievement in adolescents. In a sample of 15-yr-old children (n= 324), we measured plasma concentrations of homocysteine, choline, and betaine and genotyped them for 2 polymorphisms with effects on 1-CM, methylenetetrahydrofolate reductase (MTHFR) 677C>T, rs1801133, and phosphatidylethanolamineN-methyltransferase (PEMT), rs12325817 (G>C). The sum of school grades in 17 major subjects was used as an outcome measure for academic achievement. Lifestyle and family socioeconomic status (SES) data were obtained from questionnaires. Plasma choline was significantly and positively associated with academic achievement independent of SES factors (paternal education and income, maternal education and income, smoking, school) and of folate intake (P= 0.009,R(2)= 0.285). With the addition of thePEMTrs12325817 polymorphism, the association value was only marginally changed. Plasma betaine concentration, tHcy, and theMTHFR677C>T polymorphism did not affect academic achievement in any tested model involving choline. Dietary intake of choline is marginal in many adolescents and may be a public health concern.-Nilsson, T. K., Hurtig-Wennlöf, A., Sjöström, M., Herrmann, W., Obeid, R., Owen, J. R., Zeisel, S. Plasma 1-carbon metabolites and academic achievement in 15-yr-old adolescents.
Collapse
Affiliation(s)
- Torbjörn K Nilsson
- *Department of Medical Biosciences/Clinical Chemistry, Umeå University, Umeå, Sweden; Faculty of Medicine and Health, School of Health and Medical Sciences, Örebro University, Örebro, Sweden; Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden; Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, Homburg/Saar, Germany; Nutrition Research Institute, University of North Carolina, Chapel Hill, Kannapolis, North Carolina, USA; and Department of Nutrition, University of North Carolina Gillings School of Global Public Health, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, USA
| | - Anita Hurtig-Wennlöf
- *Department of Medical Biosciences/Clinical Chemistry, Umeå University, Umeå, Sweden; Faculty of Medicine and Health, School of Health and Medical Sciences, Örebro University, Örebro, Sweden; Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden; Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, Homburg/Saar, Germany; Nutrition Research Institute, University of North Carolina, Chapel Hill, Kannapolis, North Carolina, USA; and Department of Nutrition, University of North Carolina Gillings School of Global Public Health, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael Sjöström
- *Department of Medical Biosciences/Clinical Chemistry, Umeå University, Umeå, Sweden; Faculty of Medicine and Health, School of Health and Medical Sciences, Örebro University, Örebro, Sweden; Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden; Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, Homburg/Saar, Germany; Nutrition Research Institute, University of North Carolina, Chapel Hill, Kannapolis, North Carolina, USA; and Department of Nutrition, University of North Carolina Gillings School of Global Public Health, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, USA
| | - Wolfgang Herrmann
- *Department of Medical Biosciences/Clinical Chemistry, Umeå University, Umeå, Sweden; Faculty of Medicine and Health, School of Health and Medical Sciences, Örebro University, Örebro, Sweden; Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden; Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, Homburg/Saar, Germany; Nutrition Research Institute, University of North Carolina, Chapel Hill, Kannapolis, North Carolina, USA; and Department of Nutrition, University of North Carolina Gillings School of Global Public Health, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rima Obeid
- *Department of Medical Biosciences/Clinical Chemistry, Umeå University, Umeå, Sweden; Faculty of Medicine and Health, School of Health and Medical Sciences, Örebro University, Örebro, Sweden; Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden; Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, Homburg/Saar, Germany; Nutrition Research Institute, University of North Carolina, Chapel Hill, Kannapolis, North Carolina, USA; and Department of Nutrition, University of North Carolina Gillings School of Global Public Health, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jennifer R Owen
- *Department of Medical Biosciences/Clinical Chemistry, Umeå University, Umeå, Sweden; Faculty of Medicine and Health, School of Health and Medical Sciences, Örebro University, Örebro, Sweden; Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden; Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, Homburg/Saar, Germany; Nutrition Research Institute, University of North Carolina, Chapel Hill, Kannapolis, North Carolina, USA; and Department of Nutrition, University of North Carolina Gillings School of Global Public Health, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, USA
| | - Steven Zeisel
- *Department of Medical Biosciences/Clinical Chemistry, Umeå University, Umeå, Sweden; Faculty of Medicine and Health, School of Health and Medical Sciences, Örebro University, Örebro, Sweden; Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden; Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, Homburg/Saar, Germany; Nutrition Research Institute, University of North Carolina, Chapel Hill, Kannapolis, North Carolina, USA; and Department of Nutrition, University of North Carolina Gillings School of Global Public Health, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
61
|
|
62
|
Tyler CR, Hafez AK, Solomon ER, Allan AM. Developmental exposure to 50 parts-per-billion arsenic influences histone modifications and associated epigenetic machinery in a region- and sex-specific manner in the adult mouse brain. Toxicol Appl Pharmacol 2015; 288:40-51. [PMID: 26193056 DOI: 10.1016/j.taap.2015.07.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/13/2015] [Accepted: 07/16/2015] [Indexed: 01/07/2023]
Abstract
Epidemiological studies report that arsenic exposure via drinking water adversely impacts cognitive development in children and, in adults, can lead to greater psychiatric disease susceptibility, among other conditions. While it is known that arsenic toxicity has a profound effect on the epigenetic landscape, very few studies have investigated its effects on chromatin architecture in the brain. We have previously demonstrated that exposure to a low level of arsenic (50ppb) during all three trimesters of fetal/neonatal development induces deficits in adult hippocampal neurogenesis in the dentate gyrus (DG), depressive-like symptoms, and alterations in gene expression in the adult mouse brain. As epigenetic processes control these outcomes, here we assess the impact of our developmental arsenic exposure (DAE) paradigm on global histone posttranslational modifications and associated chromatin-modifying proteins in the dentate gyrus and frontal cortex (FC) of adult male and female mice. DAE influenced histone 3K4 trimethylation with increased levels in the male DG and FC and decreased levels in the female DG (no change in female FC). The histone methyltransferase MLL exhibited a similar sex- and region-specific expression profile as H3K4me3 levels, while histone demethylase KDM5B expression trended in the opposite direction. DAE increased histone 3K9 acetylation levels in the male DG along with histone acetyltransferase (HAT) expression of GCN5 and decreased H3K9ac levels in the male FC along with decreased HAT expression of GCN5 and PCAF. DAE decreased expression of histone deacetylase enzymes HDAC1 and HDAC2, which were concurrent with increased H3K9ac levels but only in the female DG. Levels of H3 and H3K9me3 were not influenced by DAE in either brain region of either sex. These findings suggest that exposure to a low, environmentally relevant level of arsenic during development leads to long-lasting changes in histone methylation and acetylation in the adult brain due to aberrant expression of epigenetic machinery based on region and sex.
Collapse
Affiliation(s)
- Christina R Tyler
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Alexander K Hafez
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Elizabeth R Solomon
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Andrea M Allan
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
63
|
Visentin CE, Masih S, Plumptre L, Malysheva O, Nielsen DE, Sohn KJ, Ly A, Lausman AY, Berger H, Croxford R, El-Sohemy A, Caudill MA, O'Connor DL, Kim YI. Maternal Choline Status, but Not Fetal Genotype, Influences Cord Plasma Choline Metabolite Concentrations. J Nutr 2015; 145:1491-7. [PMID: 25972528 DOI: 10.3945/jn.115.211136] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/16/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Choline deficiency during pregnancy can lead to adverse birth outcomes, including impaired neurodevelopment and birth defects. Genetic variants of choline and one-carbon metabolism may also influence birth outcomes by altering plasma choline concentrations. The effects of maternal ad libitum choline intake during pregnancy and fetal genetic variants on maternal and cord concentrations of choline and its metabolites are unknown. OBJECTIVES This prospective study sought to assess the effect of 1) maternal dietary choline intake on maternal and cord plasma concentrations of choline and its metabolites, and 2) fetal genetic polymorphisms on cord plasma concentrations. METHODS The dietary choline intake of 368 pregnant Canadian women was assessed in early (0-16 wk) and late (23-37 wk) pregnancy with the use of a food frequency questionnaire. Plasma concentrations of free choline and its metabolites were measured in maternal samples at recruitment and delivery, and in the cord blood. Ten fetal genetic variants in choline and one-carbon metabolism were assessed for their association with cord plasma concentrations of free choline and its metabolites. RESULTS Mean maternal plasma free choline, dimethylglycine, and trimethylamine N-oxide (TMAO) concentrations increased during pregnancy by 49%, 17%, and 13%, respectively (P < 0.005), whereas betaine concentrations decreased by 21% (P < 0.005). Cord plasma concentrations of free choline, betaine, dimethylglycine, and TMAO were 3.2, 2.0, 1.3, and 0.88 times corresponding maternal concentrations at delivery, respectively (all P < 0.005). Maternal plasma concentrations of betaine, dimethylglycine, and TMAO (r(2) = 0.19-0.51; P < 0.0001) at delivery were moderately strong, whereas maternal concentrations of free choline were not significant (r(2) = 0.12; P = 0.06), predictors of cord plasma concentrations of these metabolites. Neither maternal dietary intake nor fetal genetic variants predicted maternal or cord plasma concentrations of choline and its metabolites. CONCLUSION These data collectively indicate that maternal choline status, but not fetal genotype, influences cord plasma concentrations of choline metabolites. This trial was registered at clinicaltrials.gov as NCT02244684.
Collapse
Affiliation(s)
- Carly E Visentin
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| | - Shannon Masih
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| | - Lesley Plumptre
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| | - Olga Malysheva
- Division of Nutritional Sciences, College of Human Ecology, Cornell University, Ithaca, NY; and
| | - Daiva E Nielsen
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada
| | - Kyoung-Jin Sohn
- Departments of Medicine and Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| | - Anna Ly
- Departments of Medicine and Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| | - Andrea Y Lausman
- Obstetrics and Gynecology, St. Michael's Hospital and University of Toronto, Toronto, Canada
| | - Howard Berger
- Obstetrics and Gynecology, St. Michael's Hospital and University of Toronto, Toronto, Canada
| | | | - Ahmed El-Sohemy
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada
| | - Marie A Caudill
- Division of Nutritional Sciences, College of Human Ecology, Cornell University, Ithaca, NY; and
| | - Deborah L O'Connor
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Research Institute, the Hospital for Sick Children, Toronto, Canada
| | - Young-In Kim
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Departments of Medicine and Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada;
| |
Collapse
|
64
|
Silver MJ, Corbin KD, Hellenthal G, da Costa KA, Dominguez-Salas P, Moore SE, Owen J, Prentice AM, Hennig BJ, Zeisel SH. Evidence for negative selection of gene variants that increase dependence on dietary choline in a Gambian cohort. FASEB J 2015; 29:3426-35. [PMID: 25921832 PMCID: PMC4511208 DOI: 10.1096/fj.15-271056] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/16/2015] [Indexed: 01/26/2023]
Abstract
Choline is an essential nutrient, and the amount needed in the diet is modulated by
several factors. Given geographical differences in dietary choline intake and
disparate frequencies of single-nucleotide polymorphisms (SNPs) in choline metabolism
genes between ethnic groups, we tested the hypothesis that 3 SNPs that increase
dependence on dietary choline would be under negative selection pressure in settings
where choline intake is low: choline dehydrogenase (CHDH) rs12676,
methylenetetrahydrofolate reductase 1 (MTHFD1) rs2236225, and
phosphatidylethanolamine-N-methyltransferase
(PEMT) rs12325817. Evidence of negative selection was assessed in
2 populations: one in The Gambia, West Africa, where there is historic evidence of a
choline-poor diet, and the other in the United States, with a comparatively
choline-rich diet. We used 2 independent methods, and confirmation of our hypothesis
was sought via a comparison with SNP data from the Maasai, an East
African population with a genetic background similar to that of Gambians but with a
traditional diet that is higher in choline. Our results show that frequencies of SNPs
known to increase dependence on dietary choline are significantly reduced in the
low-choline setting of The Gambia. Our findings suggest that adequate intake levels
of choline may have to be reevaluated in different ethnic groups and highlight a
possible approach for identifying novel functional SNPs under the influence of
dietary selective pressure.—Silver, M. J., Corbin, K. D., Hellenthal, G., da
Costa, K.-A., Dominguez-Salas, P., Moore, S. E., Owen, J., Prentice, A. M., Hennig,
B. J., Zeisel, S. H. Evidence for negative selection of gene variants that increase
dependence on dietary choline in a Gambian cohort.
Collapse
Affiliation(s)
- Matt J Silver
- *Medical Research Council International Nutrition Group, London School of Hygiene and Tropical Medicine, London, United Kingdom; Medical Research Council Unit, Banjul, The Gambia; Nutrition Research Institute, North Carolina Research Campus, Kannapolis, North Carolina, USA; Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; University College London Genetics Institute, University College London, United Kingdom; Toxicology Services, Incorporated, Chapel Hill, North Carolina, USA; and Maternal and Child Nutrition Group, Medical Research Council Human Nutrition Research, Cambridge, United Kingdom
| | - Karen D Corbin
- *Medical Research Council International Nutrition Group, London School of Hygiene and Tropical Medicine, London, United Kingdom; Medical Research Council Unit, Banjul, The Gambia; Nutrition Research Institute, North Carolina Research Campus, Kannapolis, North Carolina, USA; Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; University College London Genetics Institute, University College London, United Kingdom; Toxicology Services, Incorporated, Chapel Hill, North Carolina, USA; and Maternal and Child Nutrition Group, Medical Research Council Human Nutrition Research, Cambridge, United Kingdom
| | - Garrett Hellenthal
- *Medical Research Council International Nutrition Group, London School of Hygiene and Tropical Medicine, London, United Kingdom; Medical Research Council Unit, Banjul, The Gambia; Nutrition Research Institute, North Carolina Research Campus, Kannapolis, North Carolina, USA; Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; University College London Genetics Institute, University College London, United Kingdom; Toxicology Services, Incorporated, Chapel Hill, North Carolina, USA; and Maternal and Child Nutrition Group, Medical Research Council Human Nutrition Research, Cambridge, United Kingdom
| | - Kerry-Ann da Costa
- *Medical Research Council International Nutrition Group, London School of Hygiene and Tropical Medicine, London, United Kingdom; Medical Research Council Unit, Banjul, The Gambia; Nutrition Research Institute, North Carolina Research Campus, Kannapolis, North Carolina, USA; Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; University College London Genetics Institute, University College London, United Kingdom; Toxicology Services, Incorporated, Chapel Hill, North Carolina, USA; and Maternal and Child Nutrition Group, Medical Research Council Human Nutrition Research, Cambridge, United Kingdom
| | - Paula Dominguez-Salas
- *Medical Research Council International Nutrition Group, London School of Hygiene and Tropical Medicine, London, United Kingdom; Medical Research Council Unit, Banjul, The Gambia; Nutrition Research Institute, North Carolina Research Campus, Kannapolis, North Carolina, USA; Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; University College London Genetics Institute, University College London, United Kingdom; Toxicology Services, Incorporated, Chapel Hill, North Carolina, USA; and Maternal and Child Nutrition Group, Medical Research Council Human Nutrition Research, Cambridge, United Kingdom
| | - Sophie E Moore
- *Medical Research Council International Nutrition Group, London School of Hygiene and Tropical Medicine, London, United Kingdom; Medical Research Council Unit, Banjul, The Gambia; Nutrition Research Institute, North Carolina Research Campus, Kannapolis, North Carolina, USA; Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; University College London Genetics Institute, University College London, United Kingdom; Toxicology Services, Incorporated, Chapel Hill, North Carolina, USA; and Maternal and Child Nutrition Group, Medical Research Council Human Nutrition Research, Cambridge, United Kingdom
| | - Jennifer Owen
- *Medical Research Council International Nutrition Group, London School of Hygiene and Tropical Medicine, London, United Kingdom; Medical Research Council Unit, Banjul, The Gambia; Nutrition Research Institute, North Carolina Research Campus, Kannapolis, North Carolina, USA; Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; University College London Genetics Institute, University College London, United Kingdom; Toxicology Services, Incorporated, Chapel Hill, North Carolina, USA; and Maternal and Child Nutrition Group, Medical Research Council Human Nutrition Research, Cambridge, United Kingdom
| | - Andrew M Prentice
- *Medical Research Council International Nutrition Group, London School of Hygiene and Tropical Medicine, London, United Kingdom; Medical Research Council Unit, Banjul, The Gambia; Nutrition Research Institute, North Carolina Research Campus, Kannapolis, North Carolina, USA; Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; University College London Genetics Institute, University College London, United Kingdom; Toxicology Services, Incorporated, Chapel Hill, North Carolina, USA; and Maternal and Child Nutrition Group, Medical Research Council Human Nutrition Research, Cambridge, United Kingdom
| | - Branwen J Hennig
- *Medical Research Council International Nutrition Group, London School of Hygiene and Tropical Medicine, London, United Kingdom; Medical Research Council Unit, Banjul, The Gambia; Nutrition Research Institute, North Carolina Research Campus, Kannapolis, North Carolina, USA; Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; University College London Genetics Institute, University College London, United Kingdom; Toxicology Services, Incorporated, Chapel Hill, North Carolina, USA; and Maternal and Child Nutrition Group, Medical Research Council Human Nutrition Research, Cambridge, United Kingdom
| | - Steven H Zeisel
- *Medical Research Council International Nutrition Group, London School of Hygiene and Tropical Medicine, London, United Kingdom; Medical Research Council Unit, Banjul, The Gambia; Nutrition Research Institute, North Carolina Research Campus, Kannapolis, North Carolina, USA; Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; University College London Genetics Institute, University College London, United Kingdom; Toxicology Services, Incorporated, Chapel Hill, North Carolina, USA; and Maternal and Child Nutrition Group, Medical Research Council Human Nutrition Research, Cambridge, United Kingdom
| |
Collapse
|
65
|
Biswas S, Giri S. Importance of Choline as Essential Nutrient and Its Role in Prevention of Various Toxicities. Prague Med Rep 2015; 116:5-15. [DOI: 10.14712/23362936.2015.40] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Choline is a water-soluble essential nutrient included as a member of the vitamin B12 group owing to its structural similarities with that of the other members of the group. Its roles and functions, however, extend much wider than that of the vitamins with which it is grouped. Choline is vital for maintenance of various key metabolic processes which play a role in the prevention or progression of various health impairments. The occurrence of diseases like neural tube defect (NTD) and Alzheimer’s is prevented by the metabolic role of choline. It is also indispensable for mitigation of various forms of toxic contamination. While adequate level of choline in the body is essential, an excess of choline can result in various forms of disorder. To maintain the optimal level of choline in the body can be a challenge. The vital roles played by choline together with the range of contradictions and problems that choline presents make choline an interesting area of study. This paper attempts to summarize and review some recent publications on choline that have opened up new prospect in understanding the multiple role played by choline and in throwing light on the role played by this wonder essential nutrient in mitigating various forms of toxic contamination.
Collapse
|
66
|
Wang M, Kim GH, Wei F, Chen H, Altarejos J, Han X. Improved method for quantitative analysis of methylated phosphatidylethanolamine species and its application for analysis of diabetic-mouse liver samples. Anal Bioanal Chem 2015; 407:5021-32. [PMID: 25725579 DOI: 10.1007/s00216-015-8534-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/16/2015] [Accepted: 02/02/2015] [Indexed: 10/23/2022]
Abstract
N-monomethyl phosphatidylethanolamine (MMPE) and N,N-dimethyl phosphatidylethanolamine (DMPE) species are intermediates of phosphatidylcholine (PC) de-novo biosynthesis through methylation of phosphatidylethanolamine (PE). This synthesis pathway for PC is especially important in the liver when choline is deficient in the diet. Despite some efforts focused on the analysis of MMPE and DMPE species, a cost-effective and high-throughput method for determination of individual MMPE and DMPE species, including their regioisomeric structures, is still missing. Therefore we adopted and improved the "mass-tag" strategy for determining these PE-like species by methylating PE, MMPE, and DMPE molecules with deuterated methyl iodide to generate PC molecules with nine, six, and three deuterium atoms, respectively. On the basis of the principles of multidimensional mass-spectrometry-based shotgun lipidomics we could directly identify and quantify these methylated PE species, including their fatty-acyl chains and regiospecific positions. The method provided remarkable sensitivity, with a limit of detection at 0.5 fmol μL(-1), high specificity, and a broad linear-dynamics range of >2500 folds. By applying this method to liver samples from streptozotocin (STZ)-induced diabetic mice and controls, we found that the levels of PC species tended to decrease and the amounts of PE species tended to increase in the liver of STZ-induced diabetic mice compared with controls, but no significant changes in MMPE and DMPE species were determined. However, remodeling of fatty-acyl chains in the determined lipids was observed in the liver of STZ-induced diabetic mice, with reduction in 16:1 and increases in 18:2, 18:1, and 18:0 acyl chains. These results indicated the improved method to be a powerful tool to reveal the function of the PC de-novo biosynthesis pathway through methylation of PE species in biological systems.
Collapse
Affiliation(s)
- Miao Wang
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, 6400 Sanger Road, Orlando, FL, 32827, USA
| | | | | | | | | | | |
Collapse
|
67
|
Park S, Choi SG, Yoo SM, Son JH, Jung YK. Choline dehydrogenase interacts with SQSTM1/p62 to recruit LC3 and stimulate mitophagy. Autophagy 2014; 10:1906-20. [PMID: 25483962 DOI: 10.4161/auto.32177] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
CHDH (choline dehydrogenase) is an enzyme catalyzing the dehydrogenation of choline to betaine aldehyde in mitochondria. Apart from this well-known activity, we report here a pivotal role of CHDH in mitophagy. Knockdown of CHDH expression impairs CCCP-induced mitophagy and PARK2/parkin-mediated clearance of mitochondria in mammalian cells, including HeLa cells and SN4741 dopaminergic neuronal cells. Conversely, overexpression of CHDH accelerates PARK2-mediated mitophagy. CHDH is found on both the outer and inner membranes of mitochondria in resting cells. Interestingly, upon induction of mitophagy, CHDH accumulates on the outer membrane in a mitochondrial potential-dependent manner. We found that CHDH is not a substrate of PARK2 but interacts with SQSTM1 independently of PARK2 to recruit SQSTM1 into depolarized mitochondria. The FB1 domain of CHDH is exposed to the cytosol and is required for the interaction with SQSTM1, and overexpression of the FB1 domain only in cytosol reduces CCCP-induced mitochondrial degradation via competitive interaction with SQSTM1. In addition, CHDH, but not the CHDH FB1 deletion mutant, forms a ternary protein complex with SQSTM1 and MAP1LC3 (LC3), leading to loading of LC3 onto the damaged mitochondria via SQSTM1. Further, CHDH is crucial to the mitophagy induced by MPP+ in SN4741 cells. Overall, our results suggest that CHDH is required for PARK2-mediated mitophagy for the recruitment of SQSTM1 and LC3 onto the mitochondria for cargo recognition.
Collapse
Key Words
- ANT, adenine nucleotide translocator
- Baf, bafilomycin A1
- CCCP, carbonyl cyanide m-chlorophenylhydrazone
- CHX, cycloheximide
- FB1, FAD/NAD (P)-binding domain 1
- FB2, FAD/NAD (P)-binding domain 2
- IM, inner membrane
- IMS, inter-membrane space
- LC3
- MPP+, 1-methyl-4-phenylpyridinium
- MTS, mitochondrial targeting sequence
- Mat, matrix
- OM, outer membrane
- PARK2/parkin
- PB1, Phox and Bem 1 domain
- PD, Parkinson disease
- PK, proteinase K
- RD, FAD-linked reductase domain
- SQSTM1/p62
- choline dehydrogenase
- mitophagy
Collapse
Affiliation(s)
- Sungwoo Park
- a Global Research Laboratory; School of Biological Science/Bio-MAX Institute ; Seoul National University ; Seoul , Korea
| | | | | | | | | |
Collapse
|
68
|
The effects of choline on hepatic lipid metabolism, mitochondrial function and antioxidative status in human hepatic C3A cells exposed to excessive energy substrates. Nutrients 2014; 6:2552-71. [PMID: 25010553 PMCID: PMC4113756 DOI: 10.3390/nu6072552] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 06/12/2014] [Accepted: 06/18/2014] [Indexed: 01/21/2023] Open
Abstract
Choline plays a lipotropic role in lipid metabolism as an essential nutrient. In this study, we investigated the effects of choline (5, 35 and 70 μM) on DNA methylation modifications, mRNA expression of the critical genes and their enzyme activities involved in hepatic lipid metabolism, mitochondrial membrane potential (Δψm) and glutathione peroxidase (GSH-Px) in C3A cells exposed to excessive energy substrates (lactate, 10 mM; octanoate, 2 mM and pyruvate, 1 mM; lactate, octanoate and pyruvate-supplemented medium (LOP)). Thirty five micromole or 70 μM choline alone, instead of a low dose (5 μM), reduced hepatocellular triglyceride (TG) accumulation, protected Δψm from decrement and increased GSH-Px activity in C3A cells. The increment of TG accumulation, reactive oxygen species (ROS) production and Δψm disruption were observed under LOP treatment in C3A cells after 72 h of culture, which were counteracted by concomitant treatment of choline (35 μM or 70 μM) partially via reversing the methylation status of the peroxisomal proliferator-activated receptor alpha (PPARα) gene promoter, upregulating PPARα, carnitine palmitoyl transferase-I (CPT-I) and downregulating fatty acid synthase (FAS) gene expression, as well as decreasing FAS activity and increasing CPT-I and GSH-Px activities. These findings provided a novel insight into the lipotropic role of choline as a vital methyl-donor in the intervention of chronic metabolic diseases.
Collapse
|
69
|
Jiang X, West AA, Caudill MA. Maternal choline supplementation: a nutritional approach for improving offspring health? Trends Endocrinol Metab 2014; 25:263-73. [PMID: 24680198 DOI: 10.1016/j.tem.2014.02.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 01/28/2014] [Accepted: 02/05/2014] [Indexed: 12/15/2022]
Abstract
The modulatory role of choline on the fetal epigenome and the impact of in utero choline supply on fetal programming and health are of great interest. Studies in animals and/or humans suggest that maternal choline supplementation during pregnancy benefits important physiologic systems such as offspring cognitive function, response to stress, and cerebral inhibition. Because alterations in offspring phenotype frequently coincide with epigenetic modifications and changes in gene expression, maternal choline supplementation may be a nutritional strategy to improve lifelong health of the child. Future studies are warranted to elucidate further the effect of choline on the fetal epigenome and to determine the level of maternal choline intake required for optimal offspring physiologic function.
Collapse
Affiliation(s)
- Xinyin Jiang
- Department of Health and Nutrition Sciences, Brooklyn College, Brooklyn, NY 11210, USA
| | - Allyson A West
- Division of Nutritional Sciences, Cornell University, Ithaca 14853, NY, USA
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca 14853, NY, USA.
| |
Collapse
|
70
|
Zinrajh D, Hörl G, Jürgens G, Marc J, Sok M, Cerne D. Increased phosphatidylethanolamine N-methyltransferase gene expression in non-small-cell lung cancer tissue predicts shorter patient survival. Oncol Lett 2014; 7:2175-2179. [PMID: 24932311 PMCID: PMC4049682 DOI: 10.3892/ol.2014.2035] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 03/14/2014] [Indexed: 01/31/2023] Open
Abstract
Lipid mobilization is of great importance for tumor growth and studies have suggested that cancer cells exhibit abnormal choline phospholipid metabolism. In the present study, we hypothesized that phosphatidylethanolamine N-methyltransferase (PEMT) gene expression is increased in non-small-cell lung cancer (NSCLC) tissues and that increased gene expression acts as a predictor of shorter patient survival. Forty-two consecutive patients with resected NSCLC were enrolled in this study. Paired samples of lung cancer tissues and adjacent non-cancer lung tissues were collected from resected specimens for the estimation of PEMT expression. SYBR Green-based real-time polymerase chain reaction was used for quantification of PEMT mRNA in lung cancer tissues. Lipoprotein lipase (LPL) and fatty acid synthase (FASN) activities had already been measured in the same tissues. During a four-year follow-up, 21 patients succumbed to tumor progression. One patient did not survive due to non-cancer reasons and was not included in the analysis. Cox regression analysis was used to assess the prognostic value of PEMT expression. Our findings show that elevated PEMT expression in the cancer tissue, relative to that in the adjacent non-cancer lung tissue, predicts shorter patient survival independently of standard prognostic factors and also independently of increased LPL or FASN activity, the two other lipid-related predictors of shorter patient survival. These findings suggest that active phosphatidylcholine and/or choline metabolism are essential for tumor growth and progression.
Collapse
Affiliation(s)
- David Zinrajh
- Chair of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Gerd Hörl
- Department of Physiological Chemistry, Center for Physiological Medicine, Medical University of Graz, A-8010 Graz, Austria
| | - Günther Jürgens
- Department of Physiological Chemistry, Center for Physiological Medicine, Medical University of Graz, A-8010 Graz, Austria
| | - Janja Marc
- Chair of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Miha Sok
- Department of Thoracic Surgery, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Darko Cerne
- Chair of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
71
|
da Costa KA, Corbin KD, Niculescu MD, Galanko JA, Zeisel SH. Identification of new genetic polymorphisms that alter the dietary requirement for choline and vary in their distribution across ethnic and racial groups. FASEB J 2014; 28:2970-8. [PMID: 24671709 DOI: 10.1096/fj.14-249557] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Effect alleles (alleles with a polymorphism that is associated with the effect being measured) in a small number of single-nucleotide polymorphisms (SNPs) are known to influence the dietary requirement for choline. In this study, we examined a much larger number of SNPs (n=200) in 10 genes related to choline metabolism for associations with development of organ dysfunction (liver or muscle) when 79 humans were fed a low-choline diet. We confirmed that effect alleles in SNPs such as the C allele of PEMT rs12325817 increase the risk of developing organ dysfunction in women when they consume a diet low in choline, and we identified novel effect alleles, such as the C allele of CHKA SNP rs7928739, that alter dietary choline requirements. When fed a low-choline diet, some people presented with muscle damage rather than liver damage; several effect alleles in SLC44A1 (rs7873937, G allele; rs2771040, G; rs6479313, G; rs16924529, A; and rs3199966, C) and one in CHKB (rs1557502, A) were more common in these individuals. This suggests that pathways related to choline metabolism are more important for normal muscle function than previously thought. In European, Mexican, and Asian Americans, and in individuals of African descent, we examined the prevalence of the effect alleles in SNPs that alter choline requirement and found that they are differentially distributed among people of different ethnic and racial backgrounds. Overall, our study has identified novel genetic variants that modulate choline requirements and suggests that the dietary requirement for choline may be different across racial and ethnic groups.-Da Costa, K.-A., Corbin, K. D., Niculescu, M. D., Galanko, J. A., Zeisel, S. H. Identification of new genetic polymorphisms that alter the dietary requirement for choline and vary in their distribution across ethnic and racial groups.
Collapse
Affiliation(s)
- Kerry-Ann da Costa
- Department of Nutrition, School of Public Health, School of Medicine, and
| | - Karen D Corbin
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina, USA
| | - Mihai D Niculescu
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina, USA
| | - Joseph A Galanko
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; and
| | - Steven H Zeisel
- Department of Nutrition, School of Public Health, School of Medicine, and Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina, USA
| |
Collapse
|
72
|
Øyen J, Nygård OK, Gjesdal CG, Ueland PM, Apalset EM, Schartum-Hansen H, Vollset SE, Meyer K, Tell GS. Plasma choline, nicotine exposure, and risk of low bone mineral density and hip fracture: the Hordaland health study. J Bone Miner Res 2014; 29:242-50. [PMID: 23794246 DOI: 10.1002/jbmr.2025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 06/04/2013] [Accepted: 06/16/2013] [Indexed: 12/16/2022]
Abstract
Choline, obtained from diet and formed by biosynthesis, is the immediate precursor of betaine. Animal studies suggest an impact of choline on bone metabolism. We examined the associations of plasma choline and betaine with bone mineral density (BMD), the risk of hip fractures, and possible effect-modification by nicotine exposure. The Hordaland Health Study (1998 to 2000) included 7074 women and men (ages 46 to 49 or 71 to 74 years). In 5315, BMD was measured. The oldest (n = 3311) were followed for hip fractures through 2009. Risk associations were studied by logistic and Cox regression by comparing the lowest and middle tertiles with the highest, as well as trends across tertiles of plasma choline and betaine. In analyses adjusted for sex and age, participants in the lowest (odds ratio [OR] = 2.00, 95% confidence interval [CI] 1.69-2.37) and middle (OR = 1.39, CI 1.17-1.66) tertiles of plasma choline had an increased risk of low BMD (lowest quintile) (p trend < 0.001). Separate analyses for sex and age groups revealed the strongest relations in elderly women (lowest tertile: OR = 2.84, CI 1.95-4.14; middle tertile: OR = 1.80, CI 1.22-2.67, p trend < 0.001), and highest OR among those in the lowest tertile who were exposed to nicotine (OR = 4.56, CI 1.87-11.11). Low plasma choline was also associated with an increased risk of hip fracture in elderly women and men (lowest tertile: hazard ratio [HR] = 1.45, CI 1.08-1.94; middle tertile: HR = 1.13, CI 0.83-1.54, p trend = 0.012). In elderly women, the HR for hip fracture was 1.90 (CI 1.32-2.73) and 1.36 (CI 0.92-1.99) (p trend < 0.001) for lowest and middle tertiles of choline, and the highest HR was found among women in the lowest tertile exposed to nicotine (HR = 2.68, CI 1.16-6.19). Plasma betaine was not related to BMD or hip fracture. Low plasma choline was associated with low BMD in both sexes and increased the risk of hip fracture in elderly women. These results should motivate further studies on choline, nicotine exposure, and bone metabolism.
Collapse
Affiliation(s)
- Jannike Øyen
- Department of Rheumatology, Haukeland University Hospital, Bergen, Norway; Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Dietary intake and plasma levels of choline and betaine in children with autism spectrum disorders. AUTISM RESEARCH AND TREATMENT 2013; 2013:578429. [PMID: 24396597 PMCID: PMC3876775 DOI: 10.1155/2013/578429] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 11/04/2013] [Accepted: 11/08/2013] [Indexed: 11/21/2022]
Abstract
Abnormalities in folate-dependent one-carbon metabolism have been reported in many children with autism. Because inadequate choline and betaine can negatively affect folate metabolism and in turn downstream methylation and antioxidant capacity, we sought to determine whether dietary intake of choline and betaine in children with autism was adequate to meet nutritional needs based on national recommendations. Three-day food records were analyzed for 288 children with autism (ASDs) who participated in the national Autism Intervention Research Network for Physical Health (AIR-P) Study on Diet and Nutrition in children with autism. Plasma concentrations of choline and betaine were measured in a subgroup of 35 children with ASDs and 32 age-matched control children. The results indicated that 60–93% of children with ASDs were consuming less than the recommended Adequate Intake (AI) for choline. Strong positive correlations were found between dietary intake and plasma concentrations of choline and betaine in autistic children as well as lower plasma concentrations compared to the control group. We conclude that choline and betaine intake is inadequate in a significant subgroup of children with ASDs and is reflected in lower plasma levels. Inadequate intake of choline and betaine may contribute to the metabolic abnormalities observed in many children with autism and warrants attention in nutritional counseling.
Collapse
|
74
|
Chen L, Chen YM, Wang LJ, Wei J, Tan YZ, Zhou JY, Yang Y, Chen YM, Ling WH, Zhu HL. Higher homocysteine and lower betaine increase the risk of microangiopathy in patients with diabetes mellitus carrying the GG genotype of PEMT G774C. Diabetes Metab Res Rev 2013; 29:607-17. [PMID: 23794489 DOI: 10.1002/dmrr.2432] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 05/04/2013] [Accepted: 06/17/2013] [Indexed: 12/17/2022]
Abstract
BACKGROUND Diabetes represents one of the greatest medical and socioeconomic threats worldwide. The pathogenesis involved is complicated. The effect of methyl donors and genetic polymorphisms in metabolic enzymes on the risk of microangiopathy in patients with diabetes is not well understood. This study investigates the association of homocysteine, choline and betaine levels and phosphatidylethanolamine N-methyltransferase (PEMT) G774C (rs12325817) genotypes with the risk of diabetes and its related microangiopathic complications. METHODS Between January 2009 and June 2010, 184 diabetic patients and 188 non-diabetic control subjects were enrolled in the hospital-based case-control study. Serum concentrations of betaine and choline were determined by high-performance liquid chromatography (HPLC)-mass spectrometry. Serum concentrations of homocysteine were assayed using HPLC. PEMT gene mutations were detected by polymerase chain reaction and restriction fragment length polymorphism. RESULTS After adjustment for potential confounders, serum total homocysteine had a significant dose-dependent positive association, and serum choline had an inverse association with the risks of diabetes and its microangiopathic complications (both p < 0.001). Although serum betaine was not associated with the risk of diabetes, it had a significant inverse association with diabetic microangiopathy. Compared with GG genotype, the CC genotype of PEMT G774C was associated with a decreased risk of diabetes (OR 0.559, 95% CI 0.338, 0.926) and its microangiopathy (OR 0.452, 95% CI 0.218, 0.937). CONCLUSION The GG genotype of the PEMT G774C polymorphism, higher levels of serum homocysteine and lower levels of serum betaine are associated with an increased risk of microangiopathy in patients with diabetes.
Collapse
Affiliation(s)
- Li Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Vance DE. Phospholipid methylation in mammals: from biochemistry to physiological function. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:1477-87. [PMID: 24184426 DOI: 10.1016/j.bbamem.2013.10.018] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 10/18/2013] [Accepted: 10/19/2013] [Indexed: 11/29/2022]
Abstract
Phosphatidylcholine is made in the liver via the CDP-choline pathway and via the conversion of phosphatidylethanolamine to phosphatidylcholine by 3 transmethylation reactions from AdoMet catalyzed by phosphatidylethanolamine N-methyltransferase (PEMT). PEMT is a 22.3kDa integral transmembrane protein of the endoplasmic reticulum and mitochondria-associated membranes. The only tissue with quantitatively significant PEMT activity is liver; however, low levels of PEMT in adipocytes have been implicated in lipid droplet formation. PEMT activity is regulated by the concentration of substrates (phosphatidylethanolamine and AdoMet) as well as the ratio of AdoMet to AdoHcy. Transcription of PEMT is enhanced by estrogen whereas the transcription factor Sp1 is a negative regulator of PEMT transcription. Studies with mice that lack PEMT have provided novel insights into the function of this enzyme. PEMT activity is required to maintain hepatic membrane integrity and for the formation of choline when dietary choline supply is limited. PEMT is required for normal secretion of very low-density lipoproteins. The lack of PEMT protects against diet-induced atherosclerosis in two mouse models. Most unexpectedly, mice that lack PEMT are protected from diet-induced obesity and insulin resistance. Moreover, mice lacking PEMT have increased susceptibility to diet-induced fatty liver and steatohepatitis. This article is part of a Special Issue entitled: Membrane Structure and Function: Relevance in the Cell's Physiology, Pathology and Therapy.
Collapse
Affiliation(s)
- Dennis E Vance
- Group on the Molecular and Cell Biology of Lipids, Department of Biochemistry, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
76
|
Changes of Molecular Glycerophospholipid Species in Plasma and Red Blood Cells During Docosahexaenoic Acid Supplementation. Lipids 2013; 48:1103-13. [DOI: 10.1007/s11745-013-3837-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 08/22/2013] [Indexed: 01/22/2023]
|
77
|
Zeisel SH. Metabolic crosstalk between choline/1-carbon metabolism and energy homeostasis. Clin Chem Lab Med 2013; 51:467-75. [PMID: 23072856 DOI: 10.1515/cclm-2012-0518] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 09/17/2012] [Indexed: 01/01/2023]
Abstract
There are multiple identified mechanisms involved in energy metabolism, insulin resistance and adiposity, but there are here-to-fore unsuspected metabolic factors that also influence these processes. Studies in animal models suggest important links between choline/1-carbon metabolism and energy homeostasis. Rodents fed choline deficient diets become hypermetabolic. Mice with deletions in one of several different genes of choline metabolism have phenotypes that include increased metabolic rate, decreased body fat/lean mass ratio, increased insulin sensitivity, decreased ATP production by mitochondria, or decreased weight gain on a high fat diet. In addition, farmers have recognized that the addition of a metabolite of choline (betaine) to cattle and swine feed reduces body fat/lean mass ratio. Choline dietary intake in humans varies over a > three-fold range, and genetic variation exists that modifies individual requirements for this nutrient. Although there are some epidemiologic studies in humans suggesting a link between choline/1-carbon metabolism and energy metabolism, there have been no controlled studies in humans that were specifically designed to examine this relationship.
Collapse
Affiliation(s)
- Steven H Zeisel
- University of North Carolina at Chapel Hill, Nutrition Research Institute, Kannapolis, NC 28081, USA.
| |
Collapse
|
78
|
Petr M, Steffl M, Kohlíková E. Effect of the MTHFR 677C/T polymorphism on homocysteinemia in response to creatine supplementation: a case study. Physiol Res 2013; 62:721-9. [PMID: 23869894 DOI: 10.33549/physiolres.932542] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Creatine (Cr) is recommended as a dietary supplement especially for athletes but its therapeutic potential is also discussed. It is assumed that human body uses Cr for the formation of phosphocreatine, which is necessary for muscular work as a source of energy. Production of Cr in a body is closely connected to methionine cycle where guanidinoacetate (GAA) is in a final step methylated from S-adenosylmethionine (SAM). Increased availability of SAM for phosphatidylcholine (PC) and sarcosine synthesis can potentially stimulate endogenous production of betaine a thus methylation of homocysteine (HCy) to form methionine. Our subject who was methylenetetrahydrofolate reductase (MTHFR) 677TT homozygote lowered plasma HCy from 33.3 micromol/l to 17.1 micromol/l following one-month Cr supplementation (5 g/day) opposite to 677CC and CT genotypes whose HCy levels tended to increase (but still in normal ranges). We suppose that Cr supplementation stimulates pathways leading to production of sarcosine which can serve to regenerate tetrahydrofolate (THF) to form 5,10-methylene-THF. This could potentially increase MTHFR enzyme activity which may later result in increased HCy methylation. Cr supplementation significantly effects metabolism of one carbon unit and potentially lower body´s demands for methyl groups. This could be beneficial as in the case of reduced enzyme activity such as MTHFR 677C/T polymorphism.
Collapse
Affiliation(s)
- M Petr
- Department of Physiology and Biochemistry, Faculty of Physical Education and Sport, Charles University in Prague, Prague, Czech Republic.
| | | | | |
Collapse
|
79
|
Mehedint MG, Zeisel SH. Choline's role in maintaining liver function: new evidence for epigenetic mechanisms. Curr Opin Clin Nutr Metab Care 2013; 16:339-45. [PMID: 23493015 PMCID: PMC3729018 DOI: 10.1097/mco.0b013e3283600d46] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW Humans eating diets low in choline develop fatty liver and liver damage. Rodents fed choline-methionine-deficient diets not only develop fatty liver, but also progress to develop fibrosis and hepatocarcinoma. This review focuses on the role of choline in liver function, with special emphasis on the epigenetic mechanisms of action. RECENT FINDINGS Dietary intake of methyl donors like choline influences the methylation of DNA and histones, thereby altering the epigenetic regulation of gene expression. The liver is the major organ within which methylation reactions occur, and many of the hepatic genes involved in pathways for the development of fatty liver, hepatic fibrosis, and hepatocarcinomas are epigenetically regulated. SUMMARY Dietary intake of choline varies over a three-fold range and many humans have genetic polymorphisms that increase their demand for choline. Choline is an important methyl donor needed for the generation of S-adenosylmethionine. Dietary choline intake is an important modifier of epigenetic marks on DNA and histones, and thereby modulates the gene expression in many of the pathways involved in liver function and dysfunction.
Collapse
Affiliation(s)
- Mihai G Mehedint
- Nutrition Research Institute at Kannapolis, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | |
Collapse
|
80
|
Abstract
Women, during pregnancy and lactation, should eat foods that contain adequate amounts of choline. A mother delivers large amounts of choline across the placenta to the fetus, and after birth she delivers large amounts of choline in milk to the infant; this greatly increases the demand on the choline stores of the mother. Adequate intake of dietary choline may be important for optimal fetal outcome (birth defects, brain development) and for maternal liver and placental function. Diets in many low income countries and in approximately one-fourth of women in high income countries, like the United States, may be too low in choline content. Prenatal vitamin supplements do not contain an adequate source of choline. For women who do not eat foods containing milk, meat, eggs, or other choline-rich foods, a diet supplement should be considered.
Collapse
Affiliation(s)
- Steven H Zeisel
- Nutrition Research Institute at Kannapolis, Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC, USA
| |
Collapse
|
81
|
Corbin KD, Abdelmalek MF, Spencer MD, da Costa KA, Galanko JA, Sha W, Suzuki A, Guy CD, Cardona DM, Torquati A, Diehl AM, Zeisel SH. Genetic signatures in choline and 1-carbon metabolism are associated with the severity of hepatic steatosis. FASEB J 2013; 27:1674-89. [PMID: 23292069 DOI: 10.1096/fj.12-219097] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Choline metabolism is important for very low-density lipoprotein secretion, making this nutritional pathway an important contributor to hepatic lipid balance. The purpose of this study was to assess whether the cumulative effects of multiple single nucleotide polymorphisms (SNPs) across genes of choline/1-carbon metabolism and functionally related pathways increase susceptibility to developing hepatic steatosis. In biopsy-characterized cases of nonalcoholic fatty liver disease and controls, we assessed 260 SNPs across 21 genes in choline/1-carbon metabolism. When SNPs were examined individually, using logistic regression, we only identified a single SNP (PNPLA3 rs738409) that was significantly associated with severity of hepatic steatosis after adjusting for confounders and multiple comparisons (P=0.02). However, when groupings of SNPs in similar metabolic pathways were defined using unsupervised hierarchical clustering, we identified groups of subjects with shared SNP signatures that were significantly correlated with steatosis burden (P=0.0002). The lowest and highest steatosis clusters could also be differentiated by ethnicity. However, unique SNP patterns defined steatosis burden irrespective of ethnicity. Our results suggest that analysis of SNP patterns in genes of choline/1-carbon metabolism may be useful for prediction of severity of steatosis in specific subsets of people, and the metabolic inefficiencies caused by these SNPs should be examined further.
Collapse
Affiliation(s)
- Karen D Corbin
- University of North Carolina at Chapel Hill Nutrition Research Institute, Kannapolis, NorthCarolina 28081, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Cheatham CL, Goldman BD, Fischer LM, da Costa KA, Reznick JS, Zeisel SH. Phosphatidylcholine supplementation in pregnant women consuming moderate-choline diets does not enhance infant cognitive function: a randomized, double-blind, placebo-controlled trial. Am J Clin Nutr 2012; 96:1465-72. [PMID: 23134891 PMCID: PMC3497930 DOI: 10.3945/ajcn.112.037184] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 08/29/2012] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Choline is essential for fetal brain development, and it is not known whether a typical American diet contains enough choline to ensure optimal brain development. OBJECTIVE The study was undertaken to determine whether supplementing pregnant women with phosphatidylcholine (the main dietary source of choline) improves the cognitive abilities of their offspring. DESIGN In a double-blind, randomized controlled trial, 140 pregnant women were randomly assigned to receive supplemental phosphatidylcholine (750 mg) or a placebo (corn oil) from 18 wk gestation through 90 d postpartum. Their infants (n = 99) were tested for short-term visuospatial memory, long-term episodic memory, language development, and global development at 10 and 12 mo of age. RESULTS The women studied ate diets that delivered ∼360 mg choline/d in foods (∼80% of the recommended intake for pregnant women, 65% of the recommended intake for lactating women). The phosphatidylcholine supplements were well tolerated. Groups did not differ significantly in global development, language development, short-term visuospatial memory, or long-term episodic memory. CONCLUSIONS Phosphatidylcholine supplementation of pregnant women eating diets containing moderate amounts of choline did not enhance their infants' brain function. It is possible that a longer follow-up period would reveal late-emerging effects. Moreover, future studies should determine whether supplementing mothers eating diets much lower in choline content, such as those consumed in several low-income countries, would enhance infant brain development.
Collapse
Affiliation(s)
- Carol L Cheatham
- Department of Psychology, University of North Carolina at Chapel Hill, Chapel Hill, NC 28081, USA.
| | | | | | | | | | | |
Collapse
|
83
|
Arendt BM, Ma DWL, Simons B, Noureldin SA, Therapondos G, Guindi M, Sherman M, Allard JP. Nonalcoholic fatty liver disease is associated with lower hepatic and erythrocyte ratios of phosphatidylcholine to phosphatidylethanolamine. Appl Physiol Nutr Metab 2012; 38:334-40. [PMID: 23537027 DOI: 10.1139/apnm-2012-0261] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is associated with altered hepatic lipid composition. Animal studies suggest that the hepatic ratio of phosphatidylcholine (PC) to phosphatidylethanolamine (PE) contributes to steatogenesis and inflammation. This ratio may be influenced by dysregulation of the PE N-methyltransferase (PEMT) pathway or by a low-choline diet. Alterations in the liver may also influence lipid composition in circulation such as in erythrocytes, which therefore may have utility as a biomarker of hepatic disease. Currently, no study has assessed both liver and erythrocyte PC/PE ratios in NAFLD. The aim of this study was to compare the PC/PE ratio in the liver and erythrocytes of patients with simple steatosis (SS) or nonalcoholic steatohepatitis (NASH) with that of healthy controls. PC and PE were measured by mass spectrometry in 28 patients with biopsy-proven NAFLD (14 SS, 14 NASH) and 9 healthy living liver donors as controls. The hepatic PC/PE ratio was lower in SS patients (median [range]) (1.23 [0.27-3.40]) and NASH patients (1.29 [0.77-3.22]) compared with controls (3.14 [2.20-3.73]); both p < 0.001) but it was not different between SS and NASH. PC was lower and PE higher in the liver of SS patients compared with controls, whereas in NASH patients only PE was higher. The PC/PE ratio in erythrocytes was also lower in SS and NASH patients compared with controls because of lower PC in both patient groups. PE in erythrocytes was not different among the groups. In conclusion, NAFLD patients have a lower PC/PE ratio in the liver and erythrocytes than do healthy controls, which may play a role in the pathogenesis. The underlying mechanisms require further investigation.
Collapse
Affiliation(s)
- Bianca M Arendt
- a Department of Medicine, University Health Network, Toronto, ON M5G 2C4, Canada
| | | | | | | | | | | | | | | |
Collapse
|
84
|
Zeisel SH, Waterland RA, Ordovás JM, Muoio DM, Jia W, Fodor A. Highlights of the 2012 Research Workshop: Using nutrigenomics and metabolomics in clinical nutrition research. JPEN J Parenter Enteral Nutr 2012; 37:190-200. [PMID: 23042849 DOI: 10.1177/0148607112462401] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) Research Workshop, "Using Nutrigenomics and Metabolomics in Clinical Nutrition Research," was held on January 21, 2012, in Orlando, Florida. The conference brought together experts in human nutrition who use nutrigenomic and metabolomic methods to better understand metabolic individuality and nutrition effects on health. We are beginning to understand how genetic variation and epigenetic events alter requirements for and responses to foods in our diet (the field of nutrigenetics/nutrigenomics and epigenetics). At the same time, methods for profiling almost all of the products of metabolism in plasma, urine, and tissues (metabolomics) are being refined. The relationships between diet and nutrigenomic-metabolomic profiles, as well as between these profiles and health, are being elucidated, and this will dramatically alter clinical practice in nutrition.
Collapse
Affiliation(s)
- Steven H Zeisel
- University of North Carolina at Chapel Hill, Kannapolis, North Carolina, USA.
| | | | | | | | | | | |
Collapse
|
85
|
Al-Humadi H, Zarros A, Kyriakaki A, Al-Saigh R, Liapi C. Choline deprivation: an overview of the major hepatic metabolic response pathways. Scand J Gastroenterol 2012; 47:874-86. [PMID: 22571353 DOI: 10.3109/00365521.2012.685755] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Choline (Ch) is an important nutrient that is involved in many physiological functions. Deprivation of Ch (CD) may lead to hepatocellular modifications and/or even hepatic tumorigenesis and it can be a frequent problem in clinical settings; it can accompany various common pathological (alcoholism and malnutrition) or physiological states (pregnancy and lactation). The aim of this review is to provide an up-to-date overview of the major metabolic pathways involved in the hepatic response toward the experimentally or clinically induced CD, and to shed more light on the implicated (and probably interrelated) mechanisms responsible for the observed hepatocellular modifications and/or carcinogenesis.
Collapse
Affiliation(s)
- Hussam Al-Humadi
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | | | | |
Collapse
|
86
|
Wu BTF, Dyer RA, King DJ, Richardson KJ, Innis SM. Early second trimester maternal plasma choline and betaine are related to measures of early cognitive development in term infants. PLoS One 2012; 7:e43448. [PMID: 22916264 PMCID: PMC3423345 DOI: 10.1371/journal.pone.0043448] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 07/20/2012] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND The importance of maternal dietary choline for fetal neural development and later cognitive function has been well-documented in experimental studies. Although choline is an essential dietary nutrient for humans, evidence that low maternal choline in pregnancy impacts neurodevelopment in human infants is lacking. We determined potential associations between maternal plasma free choline and its metabolites betaine and dimethylglycine in pregnancy and infant neurodevelopment at 18 months of age. METHODOLOGY This was a prospective study of healthy pregnant women and their full-term, single birth infants. Maternal blood was collected at 16 and 36 weeks of gestation and infant neurodevelopment was assessed at 18 months of age for 154 mother-infant pairs. Maternal plasma choline, betaine, dimethylglycine, methionine, homocysteine, cysteine, total B12, holotranscobalamin and folate were quantified. Infant neurodevelopment was evaluated using the Bayley Scales of Infant Development-III. Multivariate regression, adjusting for covariates that impact development, was used to determine the associations between maternal plasma choline, betaine and dimethylglycine and infant neurodevelopment. RESULTS The maternal plasma free choline at 16 and 36 weeks gestation was median (interquartile range) 6.70 (5.78-8.03) and 9.40 (8.10-11.3) µmol/L, respectively. Estimated choline intakes were (mean ± SD) 383 ± 98.6 mg/day, and lower than the recommended 450 mg/day. Betaine intakes were 142 ± 70.2 mg/day. Significant positive associations were found between infant cognitive test scores and maternal plasma free choline (B=6.054, SE=2.283, p=0.009) and betaine (B=7.350, SE=1.933, p=0.0002) at 16 weeks of gestation. Maternal folate, total B12, or holotranscobalamin were not related to infant development. CONCLUSION We show that choline status in the first half of pregnancy is associated with cognitive development among healthy term gestation infants. More work is needed on the potential limitation of choline or betaine in the diets of pregnant women.
Collapse
Affiliation(s)
- Brian T. F. Wu
- Nutrition and Metabolism Research Program, Child and Family Research Institute, Department of Paediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Roger A. Dyer
- Nutrition and Metabolism Research Program, Child and Family Research Institute, Department of Paediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - D. Janette King
- Nutrition and Metabolism Research Program, Child and Family Research Institute, Department of Paediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Kelly J. Richardson
- Nutrition and Metabolism Research Program, Child and Family Research Institute, Department of Paediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Sheila M. Innis
- Nutrition and Metabolism Research Program, Child and Family Research Institute, Department of Paediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- * E-mail:
| |
Collapse
|
87
|
Vance DE. Physiological roles of phosphatidylethanolamine N-methyltransferase. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:626-32. [PMID: 22877991 DOI: 10.1016/j.bbalip.2012.07.017] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 07/20/2012] [Accepted: 07/23/2012] [Indexed: 01/12/2023]
Abstract
Phosphatidylethanolamine N-methyltransferase (PEMT) catalyzes the methylation of phosphatidylethanolamine to phosphatidylcholine (PC). This 22.3 kDa protein is localized to the endoplasmic reticulum and mitochondria associated membranes of liver. The supply of the substrates AdoMet and phosphatidylethanolamine, and the product AdoHcy, can regulate the activity of PEMT. Estrogen has been identified as a positive activator, and Sp1 as a negative regulator, of transcription of the PEMT gene. Targeted inactivation of the PEMT gene produced mice that had a mild phenotype when fed a chow diet. However, when Pemt(-/-) mice were fed a choline-deficient diet steatohepatitis and liver failure developed after 3 days. The steatohepatitis was due to a decreased ratio of PC to phosphatidylethanolamine that caused leakage from the plasma membrane of hepatocytes. Pemt(-/-) mice exhibited attenuated secretion of very low-density lipoproteins and homocysteine. Pemt(-/-) mice bred with mice that lacked the low-density lipoprotein receptor, or apolipoprotein E were protected from high fat/high cholesterol-induced atherosclerosis. Surprisingly, Pemt(-/-) mice were protected from high fat diet-induced obesity and insulin resistance compared to wildtype mice. If the diet were supplemented with additional choline, the protection against obesity/insulin resistance in Pemt(-/-) mice was eliminated. Humans with a Val-to-Met substitution in PEMT at residue 175 may have increased susceptibility to nonalcoholic liver disease. This article is part of a Special Issue entitled Phospholipids and Phospholipid Metabolism.
Collapse
Affiliation(s)
- Dennis E Vance
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
88
|
Blusztajn JK, Mellott TJ. Choline nutrition programs brain development via DNA and histone methylation. Cent Nerv Syst Agents Med Chem 2012; 12:82-94. [PMID: 22483275 PMCID: PMC5612430 DOI: 10.2174/187152412800792706] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 12/05/2011] [Accepted: 12/06/2011] [Indexed: 11/22/2022]
Abstract
Choline is an essential nutrient for humans. Metabolically choline is used for the synthesis of membrane phospholipids (e.g. phosphatidylcholine), as a precursor of the neurotransmitter acetylcholine, and, following oxidation to betaine, choline functions as a methyl group donor in a pathway that produces S-adenosylmethionine. As a methyl donor choline influences DNA and histone methylation--two central epigenomic processes that regulate gene expression. Because the fetus and neonate have high demands for choline, its dietary intake during pregnancy and lactation is particularly important for normal development of the offspring. Studies in rodents have shown that high choline intake during gestation improves cognitive function in adulthood and prevents memory decline associated with old age. These behavioral changes are accompanied by electrophysiological, neuroanatomical, and neurochemical changes and by altered patterns of expression of multiple cortical and hippocampal genes including those encoding key proteins that contribute to the biochemical mechanisms of learning and memory. These actions of choline are observed long after the exposure to the nutrient ended (months) and correlate with fetal hepatic and cerebral cortical choline-evoked changes in global- and gene-specific DNA cytosine methylation and with dramatic changes of the methylation pattern of lysine residues 4, 9 and 27 of histone H3. Moreover, gestational choline modulates the expression of DNA (Dnmt1, Dnmt3a) and histone (G9a/Ehmt2/Kmt1c, Suv39h1/Kmt1a) methyltransferases. In addition to the central role of DNA and histone methylation in brain development, these processes are highly dynamic in adult brain, modulate the expression of genes critical for synaptic plasticity, and are involved in mechanisms of learning and memory. A recent study documented that in a cohort of normal elderly people, verbal and visual memory function correlated positively with the amount of dietary choline consumption. It will be important to determine if these actions of choline on human cognition are mediated by epigenomic mechanisms or by its influence on acetylcholine or phospholipid synthesis.
Collapse
Affiliation(s)
- Jan Krzysztof Blusztajn
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, 72 East Concord Street, L808, Boston, MA 02118, USA.
| | | |
Collapse
|
89
|
Zeisel SH. Diet-gene interactions underlie metabolic individuality and influence brain development: implications for clinical practice derived from studies on choline metabolism. ANNALS OF NUTRITION AND METABOLISM 2012; 60 Suppl 3:19-25. [PMID: 22614815 DOI: 10.1159/000337310] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
One of the underlying mechanisms for metabolic individuality is genetic variation. Single nucleotide polymorphisms (SNPs) in genes of metabolic pathways can create metabolic inefficiencies that alter the dietary requirement for, and responses to, nutrients. These SNPs can be detected using genetic profiling and the metabolic inefficiencies they cause can be detected using metabolomic profiling. Studies on the human dietary requirement for choline illustrate how useful these new approaches can be, as this requirement is influenced by SNPs in genes of choline and folate metabolism. In adults, these SNPs determine whether people develop fatty liver, liver damage and muscle damage when eating diets low in choline. Because choline is very important for fetal development, these SNPs may identify women who need to eat more choline during pregnancy. Some of the actions of choline are mediated by epigenetic mechanisms that permit 'retuning' of metabolic pathways during early life.
Collapse
Affiliation(s)
- Steven H Zeisel
- Nutrition Research Institute, University of North Carolina at Chapel Hill, 500 Laureate Way, Kannapolis, N.C. 28081, USA.
| |
Collapse
|
90
|
Johnson AR, Lao S, Wang T, Galanko JA, Zeisel SH. Choline dehydrogenase polymorphism rs12676 is a functional variation and is associated with changes in human sperm cell function. PLoS One 2012; 7:e36047. [PMID: 22558321 PMCID: PMC3338626 DOI: 10.1371/journal.pone.0036047] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 03/28/2012] [Indexed: 11/27/2022] Open
Abstract
Approximately 15% of couples are affected by infertility and up to half of these cases arise from male factor infertility. Unidentified genetic aberrations such as chromosomal deletions, translocations and single nucleotide polymorphisms (SNPs) may be the underlying cause of many cases of idiopathic male infertility. Deletion of the choline dehydrogenase (Chdh) gene in mice results in decreased male fertility due to diminished sperm motility; sperm from Chdh−/− males have decreased ATP concentrations likely stemming from abnormal sperm mitochondrial morphology and function in these cells. Several SNPs have been identified in the human CHDH gene that may result in altered CHDH enzymatic activity. rs12676 (G233T), a non-synonymous SNP located in the CHDH coding region, is associated with increased susceptibility to dietary choline deficiency and risk of breast cancer. We now report evidence that this SNP is also associated with altered sperm motility patterns and dysmorphic mitochondrial structure in sperm. Sperm produced by men who are GT or TT for rs12676 have 40% and 73% lower ATP concentrations, respectively, in their sperm. rs12676 is associated with decreased CHDH protein in sperm and hepatocytes. A second SNP located in the coding region of IL17BR, rs1025689, is linked to altered sperm motility characteristics and changes in choline metabolite concentrations in sperm.
Collapse
Affiliation(s)
- Amy R. Johnson
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Sai Lao
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Tongwen Wang
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Nutrition Research Institute, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Joseph A. Galanko
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Steven H. Zeisel
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Nutrition Research Institute, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
91
|
Guerrerio AL, Colvin RM, Schwartz AK, Molleston JP, Murray KF, Diehl A, Mohan P, Schwimmer JB, Lavine JE, Torbenson MS, Scheimann AO. Choline intake in a large cohort of patients with nonalcoholic fatty liver disease. Am J Clin Nutr 2012; 95:892-900. [PMID: 22338037 PMCID: PMC3302364 DOI: 10.3945/ajcn.111.020156] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND There is significant histologic and biochemical overlap between nonalcoholic fatty liver disease (NAFLD) and steatohepatitis associated with choline deficiency. OBJECTIVE We sought to determine whether subjects with biopsy-proven NAFLD and evidence of an inadequate intake of choline had more severe histologic features. DESIGN We performed a cross-sectional analysis of 664 subjects enrolled in the multicenter, prospective Nonalcoholic Steatohepatitis Clinical Research Network (NASH CRN) with baseline data on diet composition (from a recall-based food-frequency questionnaire) within 6 mo of a liver biopsy. Food questionnaires were analyzed with proprietary software to estimate daily intakes of choline. Liver biopsies were centrally read, and consensus was scored with the NASH CRN-developed scoring system. Because choline needs vary by age, sex, and menopausal status, participants were segregated into corresponding categories (children 9-13 y old, males ≥14 y old, premenopausal women ≥19 y old, and postmenopausal women) on the basis of the Institute of Medicine's definition of adequate intake (AI) for choline. Deficient intake was defined as <50% AI. RESULTS Postmenopausal women with deficient choline intake had worse fibrosis (P = 0.002) once factors associated with NAFLD (age, race-ethnicity, obesity, elevated triglycerides, diabetes, alcohol use, and steroid use) were considered in multiple ordinal logistic regression models. Choline intake was not identified as a contributor to disease severity in children, men, or premenopausal women. CONCLUSION Decreased choline intake is significantly associated with increased fibrosis in postmenopausal women with NAFLD. The Pioglitazone vs Vitamin E vs Placebo for Treatment of Non-Diabetic Patients With Nonalcoholic Steatohepatitis trial was registered at clinicaltrials.gov as NCT00063622, and the Treatment of Nonalcoholic Fatty Liver Disease in Children trial was registered at clinicaltrials.gov as NCT00063635.
Collapse
Affiliation(s)
- Anthony L Guerrerio
- Division of Pediatric Gastroenterology and Nutrition, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Phosphatidylethanolamine N-methyltransferase and choline dehydrogenase gene polymorphisms are associated with human sperm concentration. Asian J Androl 2012; 14:778-83. [PMID: 22387881 DOI: 10.1038/aja.2011.125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Choline is a crucial factor in the regulation of sperm membrane structure and fluidity, and this nutrient plays an important role in the maturation and fertilizing capacity of spermatozoa. Transcripts of phosphatidylethanolamine N-methyltransferase (PEMT) and choline dehydrogenase (CHDH), two basic enzymes of choline metabolism, have been observed in the human testis, demonstrating their gene expression in this tissue. In the present study, we explored the contribution of the PEMT and CHDH gene variants to sperm parameters. Two hundred oligospermic and 250 normozoospermic men were recruited. DNA was extracted from the spermatozoa, and the PEMT -774G>C and CHDH +432G>T polymorphisms were genotyped. The genotype distribution of the PEMT -774G>C polymorphism did not differ between oligospermic and normozoospermic men. In contrast, in the case of the CHDH +432G>T polymorphism, oligospermic men presented the CHDH 432G/G genotype more frequently than normozoospermic men (62% vs. 42%, P<0.001). The PEMT 774G/G genotype was associated with a higher sperm concentration compared to the PEMT 774G/C and 774C/C genotypes in oligospermic men (12.5 ± 5.6 × 10(6) spermatozoa ml(-1) vs. 8.3 ± 5.2 × 10(6) spermatozoa ml(-1), P<0.002) and normozoospermic men (81.5 ± 55.6 × 10(6) vs. 68.1 ± 44.5 × 10(6) spermatozoa ml(-1), P<0.006). In addition, the CHDH 432G/G genotype was associated with higher sperm concentration compared to CHDH 432G/T and 432T/T genotypes in oligospermic (11.8 ± 5.1 × 10(6) vs. 7.8 ± 5.3 × 10(6) spermatozoa ml(-1), P<0.003) and normozoospermic men (98.6 ± 62.2 × 10(6) vs. 58.8 ± 33.6 × 10(6) spermatozoa ml(-1), P<0.001). In our series, the PEMT -774G>C and CHDH +432G>T polymorphisms were associated with sperm concentration. This finding suggests a possible influence of these genes on sperm quality.
Collapse
|
93
|
Abstract
PURPOSE OF REVIEW Choline is an essential nutrient and the liver is a central organ responsible for choline metabolism. Hepatosteatosis and liver cell death occur when humans are deprived of choline. In the last few years, there have been significant advances in our understanding of the mechanisms that influence choline requirements in humans and in our understanding of choline's effects on liver function. These advances are useful in elucidating why nonalcoholic fatty liver disease (NAFLD) occurs and progresses sometimes to hepatocarcinogenesis. RECENT FINDINGS Humans eating low-choline diets develop fatty liver and liver damage. This dietary requirement for choline is modulated by estrogen and by single-nucleotide polymorphisms in specific genes of choline and folate metabolism. The spectrum of choline's effects on liver range from steatosis to development of hepatocarcinomas, and several mechanisms for these effects have been identified. They include abnormal phospholipid synthesis, defects in lipoprotein secretion, oxidative damage caused by mitochondrial dysfunction, and endoplasmic reticulum stress. Furthermore, the hepatic steatosis phenotype can be characterized more fully via metabolomic signatures and is influenced by the gut microbiome. Importantly, the intricate connection between liver function, one-carbon metabolism, and energy metabolism is just beginning to be elucidated. SUMMARY Choline influences liver function, and the dietary requirement for this nutrient varies depending on an individual's genotype and estrogen status. Understanding these individual differences is important for gastroenterologists seeking to understand why some individuals develop NAFLD and others do not, and why some patients tolerate total parenteral nutrition and others develop liver dysfunction.
Collapse
|
94
|
Abstract
Food intake can influence neuronal functions through different modulators expressed in the brain. The present review is a report through relevant experimental findings on the effects of choline, a nutritional component found in the diet, to identify a safe and effective dietary solution that can offer some protection against neurotoxicity and neurological disorders and that can be implemented in animals and humans in a very short period of time.
Collapse
Affiliation(s)
- Elisabetta Biasi
- Department of Pharmacology and Cancer Biology, Duke Univesity Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
95
|
Abstract
The vitamin folate functions within the cell as a carrier of one-carbon units. The requirement for one-carbon transfers is ubiquitous and all mammalian cells carry out folate dependent reactions. In recent years, low folate status has been linked to risk of numerous adverse health conditions throughout life from birth defects and complications of pregnancy to cardiovascular disease, cancer and cognitive dysfunction in the elderly. In many instances inadequate intake of folate seems to be the primary contributor but there is also evidence that an underlying genetic susceptibility can play a modest role by causing subtle alterations in the availability, metabolism or distribution of intermediates in folate related pathways. Folate linked one-carbon units are essential for DNA synthesis and repair and as a source of methyl groups for biological methylation reactions. The notion of common genetic variants being linked to risk of disease was relatively novel in 1995 when the first functional folate-related polymorphism was discovered. Numerous polymorphisms have now been identified in folate related genes and have been tested for functionality either as a modifier of folate status or as being associated with risk of disease. Moreover, there is increasing research into the importance of folate-derived one-carbon units for DNA and histone methylation reactions, which exert crucial epigenetic control over cellular protein synthesis. It is thus becoming clear that genetic aspects of folate metabolism are wide-ranging and may touch on events as disparate as prenatal imprinting to cancer susceptibility. This chapter will review the current knowledge in this area.
Collapse
Affiliation(s)
- Anne M Molloy
- School of Medicine, Trinity College Dublin, Dublin 2, Ireland,
| |
Collapse
|
96
|
Corbin KD, Zeisel SH. The nutrigenetics and nutrigenomics of the dietary requirement for choline. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 108:159-77. [PMID: 22656377 DOI: 10.1016/b978-0-12-398397-8.00007-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Advances in nutrigenetics and nutrigenomics have been instrumental in demonstrating that nutrient requirements vary among individuals. This is exemplified by studies of the nutrient choline, in which gender, single-nucleotide polymorphisms, estrogen status, and gut microbiome composition have been shown to influence its optimal intake level. Choline is an essential nutrient with a wide range of biological functions, and current studies are aimed at refining our understanding of its requirements and, importantly, on defining the molecular mechanisms that mediate its effects in instances of suboptimal dietary intake. This chapter introduces the reader to challenges in developing individual nutrition recommendations, the biological function of choline, current and future research paradigms to fully understand the consequences of inadequate choline nutrition, and some forward thinking about the potential for individualized nutrition recommendations to become a tangible application for improved health.
Collapse
Affiliation(s)
- Karen D Corbin
- University of North Carolina at Chapel Hill, Nutrition Research Institute, Kannapolis, North Carolina, USA
| | | |
Collapse
|
97
|
Zeisel SH. Dietary choline deficiency causes DNA strand breaks and alters epigenetic marks on DNA and histones. Mutat Res 2011; 733:34-8. [PMID: 22041500 DOI: 10.1016/j.mrfmmm.2011.10.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 10/03/2011] [Accepted: 10/04/2011] [Indexed: 01/30/2023]
Abstract
Dietary choline is an important modulator of gene expression (via epigenetic marks) and of DNA integrity. Choline was discovered to be an essential nutrient for some humans approximately one decade ago. This requirement is diminished in young women because estrogen drives endogenous synthesis of phosphatidylcholine, from which choline can be derived. Almost half of women have a single nucleotide polymorphism that abrogates estrogen-induction of endogenous synthesis, and these women require dietary choline just as do men. In the US, dietary intake of choline is marginal. Choline deficiency in people is associated with liver and muscle dysfunction and damage, with apoptosis, and with increased DNA strand breaks. Several mechanisms explain these modifications to DNA. Choline deficiency increases leakage of reactive oxygen species from mitochondria consequent to altered mitochondrial membrane composition and enhanced fatty acid oxidation. Choline deficiency impairs folate metabolism, resulting in decreased thymidylate synthesis and increased uracil misincorporation into DNA, with strand breaks resulting during error-prone repair attempts. Choline deficiency alters DNA methylation, which alters gene expression for critical genes involved in DNA mismatch repair, resulting in increased mutation rates. Any dietary deficiency which increases mutation rates should be associated with increased risk of cancers, and this is the case for choline deficiency. In rodent models, diets low in choline and methyl-groups result in spontaneous hepatocarcinomas. In human epidemiological studies, there are interesting data that suggest that this also may be the case for humans, especially those with SNPs that increase the dietary requirement for choline.
Collapse
Affiliation(s)
- Steven H Zeisel
- Nutrition Research Institute, School of Public Health and School of Medicine, The University of North Carolina at Chapel Hill, Kannapolis, NC 28081, United States. steven
| |
Collapse
|
98
|
Tang H, Wang XX, Zhao SG, Wang JJ, Zhang C, Lu W, Li HY, Wen QS. Association between the G175A and rs12325817 polymorphisms in the phosphatidylethanolamine N-methyltransferase gene and susceptibility to nonalcoholic fatty liver disease. Shijie Huaren Xiaohua Zazhi 2011; 19:3035-3039. [DOI: 10.11569/wcjd.v19.i29.3035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether there is an association between the G175A and rs12325817 single nucleotide polymorphisms (SNPs) in the phosphatidylethanolamine N-methyltransferase (PEMT) gene and susceptibility to non-alcoholic fatty liver disease (NAFLD).
METHODS: The G175A and rs12325817 SNPs were genotyped using polymerase chain reaction-restriction fragment length polymorphism in 156 patients with NAFLD and 188 healthy controls. Clinical parameters were compared between NAFLD patients and controls.
RESULTS: The genotype and allele frequency of G175A and rs12325817 were significantly different between NAFLD patients and controls (G175A: P = 0.03; rs12325817: P = 0.00075). The frequency of A allele in the G175A locus and C allele in the rs12325817 locus were significantly higher in the NAFLD group (G175A: P = 0.002; rs12325817: P = 0.00025) than in the control group, which indicates that these alleles are risk factors for NAFLD.
CONCLUSION: The G175A and rs12325817 polymorphisms in the PEMT gene are associated with susceptibility to NAFLD
Collapse
|
99
|
Yuan Z, Shao M, He J. Progress in hepatitis C research. Shijie Huaren Xiaohua Zazhi 2011; 19:3046-3052. [DOI: 10.11569/wcjd.v19.i29.3046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C is caused by the hepatitis C virus (HCV), which primarily has six genotypes. Hepatitis C is mainly transmitted via contact with blood contaminated by HCV. HCV infection is distributed worldwide, and China is a medium- to high-risk endemic area for HCV. Hepatitis C easily becomes chronic and even develops into liver cirrhosis and hepatoma. As a consequence, it places a major demand on early diagnosis and treatment of this disease. Currently, the diagnosis of hepatitis C mainly relies on the detection of HCV-RNA and viral genotypes. Combined therapy with polyethylene glycol interferon α (PEG-INF-α) and ribavirin (RBV) is the standard antiviral therapy regimen for HCV (SOC). What's more, the latest research has been focused on the responsible guide treat (RGT) based on the SOC. The application of newly developed small molecule drugs alone or in combination with SOC also represents a hot topic. This paper will provide a brief overview of progress in hepatitis C research.
Collapse
|
100
|
Cole LK, Vance JE, Vance DE. Phosphatidylcholine biosynthesis and lipoprotein metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:754-61. [PMID: 21979151 DOI: 10.1016/j.bbalip.2011.09.009] [Citation(s) in RCA: 273] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 09/15/2011] [Accepted: 09/19/2011] [Indexed: 12/28/2022]
Abstract
Phosphatidylcholine (PC) is the major phospholipid component of all plasma lipoprotein classes. PC is the only phospholipid which is currently known to be required for lipoprotein assembly and secretion. Impaired hepatic PC biosynthesis significantly reduces the levels of circulating very low density lipoproteins (VLDLs) and high density lipoproteins (HDLs). The reduction in plasma VLDLs is due in part to impaired hepatic secretion of VLDLs. Less PC within the hepatic secretory pathway results in nascent VLDL particles with reduced levels of PC. These particles are recognized as being defective and are degraded within the secretory system by an incompletely defined process that occurs in a post-endoplasmic reticulum compartment, consistent with degradation directed by the low-density lipoprotein receptor and/or autophagy. Moreover, VLDL particles are taken up more readily from the circulation when the PC content of the VLDLs is reduced, likely due to a preference of cell surface receptors and/or enzymes for lipoproteins that contain less PC. Impaired PC biosynthesis also reduces plasma HDLs by inhibiting hepatic HDL formation and by increasing HDL uptake from the circulation. These effects are mediated by elevated expression of ATP-binding cassette transporter A1 and hepatic scavenger receptor class B type 1, respectively. Hepatic PC availability has recently been linked to the progression of liver and heart disease. These findings demonstrate that hepatic PC biosynthesis can regulate the amount of circulating lipoproteins and suggest that hepatic PC biosynthesis may represent an important pharmaceutical target. This article is part of a Special Issue entitled Triglyceride Metabolism and Disease.
Collapse
Affiliation(s)
- Laura K Cole
- Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | | | | |
Collapse
|