1
|
King C, Plakke B. Maternal choline supplementation in neurodevelopmental disorders: mechanistic insights from animal models and future directions. Nutr Neurosci 2025; 28:405-424. [PMID: 39046330 DOI: 10.1080/1028415x.2024.2377084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
OBJECTIVES To synthesize evidence from animal models of neurodevelopmental disorders (NDD) using maternal choline supplementation, to characterize current knowledge on the mechanisms of choline's protective effects against NDD, and to identify gaps in knowledge for future study. METHODS A literature review was conducted in PubMed to identify studies using prenatal choline supplementation interventions in rodent models of neurodevelopmental disorders. 24 studies were identified, and behavioral and biological findings were extracted from each. Studies examining both genetic and environmental risk factors were included. RESULTS Maternal choline supplementation during gestation is protective against both genetic and environmental NDD risk factors. Maternal choline supplementation improves both cognitive and affective outcomes throughout the lifespan in NDD models. Prenatal choline improved these outcomes through its participation in processes like neurogenesis, epigenetic regulation, and anti-inflammatory signaling. DISCUSSION Maternal choline supplementation improves behavioral and neurobiological outcomes in animal models of NDD, paralleling findings in humans. Animal models provide a unique opportunity to study the mechanisms by which gestational choline improves neurodevelopmental outcomes. This is especially important since nearly 90% of pregnant people in the United States are deficient in choline intake. However, much is still unknown about the mechanisms through which choline and its derivatives act. Further research into this topic, especially mechanistic studies in animal models, is critical to modernize maternal choline intake guidelines and to develop interventions to increase maternal choline intake in vulnerable populations.
Collapse
Affiliation(s)
- Cole King
- Psychological Sciences, Kansas State University, Manhattan, KS, USA
| | - Bethany Plakke
- Psychological Sciences, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
2
|
France TL, Ortega AF, Richards AT, Farricker MJ, Fontoura AB, McFadden JW. Abomasal Infusion of Deuterium-Labeled Choline Confirms that Choline is a Methyl Donor in Gestating and Lactating Holstein Dairy Cattle. J Nutr 2025; 155:523-532. [PMID: 39581267 DOI: 10.1016/j.tjnut.2024.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND In cattle, dietary choline is fed in a rumen-protected form to bypass ruminal degradation while allowing intestinal absorption of choline. We require data to help us define choline utilization in the dairy cow transitioning from gestation to lactation. OBJECTIVES The study examined choline methyl group transfer in dairy cattle transitioning from gestation to lactation. METHODS Six multiparous, rumen-cannulated Holstein cows were enrolled in a longitudinal study design where they were abomasally infused with choline chloride (CC) for a 5-d experimental period at -3 wk from the expected due date and again at 2 wk postpartum. Twenty percent of CC provided per day was replaced with the stable-isotope trimethyl-d9-CC. Plasma, milk, liver, urine, and fecal samples were analyzed for choline and choline metabolite concentrations and isotope enrichment. RESULTS Plasma betaine (P < 0.01) and liver choline and betaine concentrations were lower during lactation (P < 0.01). During lactation, cows had greater circulating enrichment of d3-choline, d3-betaine, d9-betaine, and d9-phosphatidylcholine (PC; P ≤ 0.05), and greater liver enrichment of d3-glycerophosphorylcholine and d9-sphingomyelin (P ≤ 0.02). Greater urinary and fecal losses of d3- and d9-trimethylamine (TMA), and d3- and d9-TMA N-oxide (TMAO) were observed during pregnancy (P ≤ 0.05). Total choline metabolites detected in urine from infused CC were greater during pregnancy, relative to lactation (P < 0.01). Total fecal choline metabolites from infused CC were greater during lactation than during pregnancy (P = 0.03). CONCLUSIONS Our data confirm that choline is a methyl donor (e.g., detected d3-choline), and choline is used by both the cytidine diphosphate-choline and phosphatidylethanolamine N-methyl transferase pathways in the cow during gestation and lactation. We also provide evidence for the endogenous recycling of absorbed choline via bile (i.e. presence of d3-TMA). Degradation of choline to TMA and TMAO must be considered when evaluating choline bioavailability.
Collapse
Affiliation(s)
- Tanya L France
- Department of Animal Science, Cornell University, Ithaca, NY, United States
| | - Andres F Ortega
- Department of Animal Science, Cornell University, Ithaca, NY, United States
| | - Andrew T Richards
- Department of Animal Science, Cornell University, Ithaca, NY, United States
| | | | - Ananda Bp Fontoura
- Department of Animal Science, Cornell University, Ithaca, NY, United States
| | - Joseph W McFadden
- Department of Animal Science, Cornell University, Ithaca, NY, United States.
| |
Collapse
|
3
|
Fleming JM, Rosa G, Bland V, Kauwell GPA, Malysheva OV, Wettstein A, Hausman DB, Bailey LB, Park HJ. Response of One-Carbon Biomarkers in Maternal and Cord Blood to Folic Acid Dose During Pregnancy. Nutrients 2024; 16:3703. [PMID: 39519534 PMCID: PMC11547940 DOI: 10.3390/nu16213703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES The folate Recommended Daily Allowance (RDA) for pregnant women is 600 μg/day dietary folate equivalents, which is equivalent to approximately 400 μg folic acid. Many prenatal supplements contain much higher doses of folic acid. The body's ability to reduce synthetic folic acid to the metabolically active form may be exceeded with high levels of supplementation. The objective of this double-blinded randomized controlled intervention trial was to determine changes in unmetabolized folic acid and other biomarkers of folate and one-carbon metabolism in maternal and cord blood in response to a folic acid dose commonly found in prenatal supplements (800 μg/day) compared to the dose equivalent of the RDA (400 μg/day). METHODS Healthy pregnant women were randomized and provided supplements from their first prenatal visit (<12 weeks gestation) through delivery. Maternal blood was collected at baseline and delivery. Umbilical cord blood from the mothers was collected at delivery. RESULTS A repeated measures analysis of variance revealed that there was a significant group supplemental dose effect (p = 0.0225) on serum unmetabolized folic acid concentration in mothers but no difference in cord blood unmetabolized folic acid concentrations between groups. Mixed effects analysis found a significant overall effect of pre-pregnancy BMI (p = 0.0360) and length of previous folic acid supplementation (p = 0.0281) on serum folate concentrations. No treatment effect was seen in RBC folate concentrations. Choline concentrations were higher in cord blood from the 800 μg/day group compared to the 400 μg/day group, but there was no group effect in maternal choline concentrations. CONCLUSIONS The results indicate that folic acid dose during pregnancy affects certain folate and one-carbon biomarkers, and these effects are not consistent between maternal and cord blood. Potential long-term effects of these results on both mothers and offspring are unknown and merit further investigation.
Collapse
Affiliation(s)
- Jennifer M. Fleming
- Department of Nutritional Sciences, University of Georgia, Athens, GA 30602, USA
| | - Gisselle Rosa
- Department of Nutritional Sciences, University of Georgia, Athens, GA 30602, USA
| | - Victoria Bland
- Department of Nutritional Sciences, University of Georgia, Athens, GA 30602, USA
| | - Gail P. A. Kauwell
- Department of Health Sciences, University of Central Florida, Orlando, FL 32816, USA
| | - Olga V. Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA
| | - Alleigh Wettstein
- Department of Nutritional Sciences, University of Georgia, Athens, GA 30602, USA
| | - Dorothy B. Hausman
- Department of Nutritional Sciences, University of Georgia, Athens, GA 30602, USA
| | - Lynn B. Bailey
- Department of Nutritional Sciences, University of Georgia, Athens, GA 30602, USA
| | - Hea Jin Park
- Department of Nutritional Sciences, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
4
|
King C, Plakke B. Maternal choline supplementation modulates cognition and induces anti-inflammatory signaling in the prefrontal cortices of adolescent rats exposed to maternal immune activation. Brain Behav Immun Health 2024; 40:100836. [PMID: 39206430 PMCID: PMC11350509 DOI: 10.1016/j.bbih.2024.100836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Maternal infection has long been described as a risk factor for neurodevelopmental disorders, especially autism spectrum disorders (ASD) and schizophrenia. Although many pathogens do not cross the placenta and infect the developing fetus directly, the maternal immune response to them is sufficient to alter fetal neurodevelopment, a phenomenon termed maternal immune activation (MIA). Low maternal choline is also a risk factor for neurodevelopmental disorders, and most pregnant people do not receive enough of it. In addition to its role in neurodevelopment, choline is capable of inducing anti-inflammatory signaling through a nicotinic pathway. Therefore, it was hypothesized that maternal choline supplementation would blunt the neurodevelopmental impact of MIA in offspring through long-term instigation of cholinergic anti-inflammatory signaling. To model MIA in rats, the viral mimetic polyinosinic:polycytidylic acid (poly(I:C)) was used to elicit a maternal antiviral innate immune response in dams both with and without choline supplementation. Offspring were reared to both early and late adolescent stages (postnatal days 28 and 50, respectively), where anxiety-related behaviors and cognition were examined. After behavioral testing, animals were euthanized, and their prefrontal cortices (PFCs) were collected for analysis. MIA offspring demonstrated sex-specific patterns of altered cognition and repetitive behaviors, which were modulated by maternal choline supplementation. Choline supplementation also bolstered anti-inflammatory signaling in the PFCs of MIA animals at both early and late adolescent stages. These findings suggest that maternal choline supplementation may be sufficient to blunt some of the behavioral and neurobiological impacts of inflammatory exposures in utero, indicating that it may be a cheap, safe, and effective intervention for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Cole King
- Department of Psychological Sciences, Kansas State University, 1114 Mid-Campus Drive, Manhattan, KS, 66502, USA
| | - Bethany Plakke
- Department of Psychological Sciences, Kansas State University, 1114 Mid-Campus Drive, Manhattan, KS, 66502, USA
| |
Collapse
|
5
|
Corchero-Palacios S, Alegria-Lertxundi I, de Pancorbo MM, Arroyo-Izaga M. Interactions between folate metabolism-related nutrients and polymorphisms on colorectal cancer risk: a case-control study in the Basque country. Eur J Nutr 2024; 63:1681-1693. [PMID: 38652304 PMCID: PMC11329606 DOI: 10.1007/s00394-024-03371-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 03/01/2024] [Indexed: 04/25/2024]
Abstract
Folate-mediated one-carbon metabolism (FOCM) plays an important role in colorectal carcinogenesis. Previous studies have assessed the role of folate-mediated one-carbon metabolism (FOCM)-related gene-diet interaction in the aetiology of colorectal cancer (CRC), however, the results remained inconclusive. Thus, this study aimed to investigate dietary factors and genetic variants related to FOCM, as well as potential nutrient-gene and nutrient-lifestyle interactions, on CRC risk. This observational study included 229 patients diagnosed with CRC and 229 age- and sex-matched subjects as controls from a population-based bowel cancer screening program. Conditional logistic regression was used to calculate odds ratios (ORs) and 95% confidence intervals (95%CI) for CRC risk. A Bonferroni-corrected threshold of α = 0.005 was considered significant, and P values less than 0.05 were considered to be suggestive of an association. After Bonferroni correction, a high dietary intake of betaine was associated with a decreased risk of CRC in the adjusted model (OR, 95% CI: 0.21, 0.10-0.40, P < 0.001). Two SNPs, rs1476413 and rs17824591, exhibited significant gene-diet interactions with total choline ad vitamin B12 intakes, respectively, in adjusted models (total choline, tertile 3 vs. 1, OR, 95% CI: 0.25, 0.11-0.66, Pinteraction = 0.012; vitamin B12, tertile 2 vs. tertile 1, OR, 95% CI: 2.48, 1.04-5.00, Pinteraction = 0.003). These findings suggest that betaine intake and interactions between some dietary factors and variants in MTHFR and MTHFD1 genes have an influence on CRC risk in the population studied. If these results are confirmed, specific nutritional intervention strategies could be designed.
Collapse
Affiliation(s)
- Sara Corchero-Palacios
- Department of Pharmacy and Food Sciences, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz (Araba/Álava), 01006, Spain
| | - Iker Alegria-Lertxundi
- Department of Pharmacy and Food Sciences, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz (Araba/Álava), 01006, Spain
| | - Marian M de Pancorbo
- Department of Z. and Cellular Biology A., Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz (Araba/Álava), 01006, Spain
- BIOMICs Research Group, Microfluidics & BIOMICs Cluster, Lascaray Research Center, University of the Basque Country UPV/EHU, Bioaraba, BA04.03, 01006, Vitoria-Gasteiz (Araba/Álava), Spain
| | - Marta Arroyo-Izaga
- Department of Pharmacy and Food Sciences, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz (Araba/Álava), 01006, Spain.
- BIOMICs Research Group, Microfluidics & BIOMICs Cluster, Lascaray Research Center, University of the Basque Country UPV/EHU, Bioaraba, BA04.03, 01006, Vitoria-Gasteiz (Araba/Álava), Spain.
| |
Collapse
|
6
|
Dymek A, Oleksy Ł, Stolarczyk A, Bartosiewicz A. Choline-An Underappreciated Component of a Mother-to-Be's Diet. Nutrients 2024; 16:1767. [PMID: 38892700 PMCID: PMC11174651 DOI: 10.3390/nu16111767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/27/2024] [Accepted: 06/02/2024] [Indexed: 06/21/2024] Open
Abstract
The nutritional status of the mother-to-be has a key impact on the proper development of the fetus. Although all nutrients are important for the developing baby, recent research indicates the importance of adequate choline intake during the periconceptional period, pregnancy, and lactation. Choline plays a key role in the biosynthesis of cell membranes, supporting liver function, neurotransmission, brain development, and DNA and histone methylation. Choline participates in the formation of a child's nervous system, supports its cognitive development, and reduces the risk of neural tube defects. The human body is incapable of producing sufficient choline to meet its needs; therefore, it must be obtained from the diet. Current data indicate that most women in their reproductive years do not achieve the recommended daily intake of choline. The presented narrative review indicates the importance of educating mothers-to-be and thereby increasing their awareness of the effects of choline on maternal and child health, which can lead to a more aware and healthy pregnancy and proper child development.
Collapse
Affiliation(s)
- Agnieszka Dymek
- Students Scientific Club of Dietetics, Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
| | - Łukasz Oleksy
- Department of Physiotherapy, Faculty of Health Sciences, Jagiellonian University Medical College, 31-008 Kraków, Poland
| | - Artur Stolarczyk
- Department of Orthopedics and Rehabilitation, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Anna Bartosiewicz
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
| |
Collapse
|
7
|
Kadam I, Dalloul M, Hausser J, Vaday D, Gilboa E, Wang L, Hittelman J, Hoepner L, Fordjour L, Chitamanni P, Saxena A, Jiang X. Role of one-carbon nutrient intake and diabetes during pregnancy in children's growth and neurodevelopment: A 2-year follow-up study of a prospective cohort. Clin Nutr 2024; 43:1216-1223. [PMID: 38636347 DOI: 10.1016/j.clnu.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/21/2023] [Accepted: 04/09/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND & AIMS Both maternal metabolic dysregulation, e.g., gestational diabetes mellitus (GDM), and maternal supply of nutrients that participate in one-carbon (1C) metabolism, e.g., folate, choline, betaine, and vitamin B12, have been demonstrated to influence epigenetic modification such as DNA methylation, thereby exerting long-lasting impacts on growth and development of offspring. This study aimed to determine how maternal 1C nutrient intake was associated with DNA methylation and further, development of children, as well as whether maternal GDM status modified the association in a prospective cohort. METHODS In this study, women with (n = 18) and without (n = 20) GDM were recruited at 25-33 weeks gestation. Detailed dietary intake data was collected by 3-day 24-h dietary recall and nutrient levels in maternal blood were also assessed at enrollment. The maternal-child dyads were invited to participate in a 2-year follow-up during which anthropometric measurement and the Bayley Scales of Infant and Toddler Development™ Screening Test (Third Edition) were conducted on children. The association between maternal 1C nutrients and children's developmental outcomes was analyzed with a generalized linear model controlling for maternal GDM status. RESULTS We found that children born to mothers with GDM had lower scores in the language domain of the Bayley test (p = 0.049). Higher maternal food folate and choline intakes were associated with better language scores in children (p = 0.01 and 0.025, respectively). Higher maternal food folate intakes were also associated with better cognitive scores in children (p = 0.002). Higher 1C nutrient intakes during pregnancy were associated with lower body weight of children at 2 years of age (p < 0.05). However, global DNA methylation of children's buccal cells was not associated with any maternal 1C nutrients. CONCLUSIONS In conclusion, higher 1C nutrient intake during pregnancy was associated with lower body weight and better neurodevelopmental outcomes of children. This may help overcome the lower language scores seen in GDM-affected children in this cohort. Studies in larger cohorts and with a longer follow-up duration are needed to further delineate the relationship between prenatal 1C nutrient exposure, especially in GDM-affected pregnancies, and offspring health outcomes.
Collapse
Affiliation(s)
- Isma'il Kadam
- Departments of Health and Nutrition Sciences, Brooklyn College of City University of New York, Brooklyn, NY 11210, USA; PhD Program in Biochemistry, Graduate Center of the City University of New York, New York, NY 10016, USA
| | - Mudar Dalloul
- Department of Obstetrics and Gynecology, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Jeanette Hausser
- Departments of Health and Nutrition Sciences, Brooklyn College of City University of New York, Brooklyn, NY 11210, USA
| | - Doron Vaday
- Departments of Health and Nutrition Sciences, Brooklyn College of City University of New York, Brooklyn, NY 11210, USA
| | - Ella Gilboa
- Departments of Health and Nutrition Sciences, Brooklyn College of City University of New York, Brooklyn, NY 11210, USA
| | - Liang Wang
- Department of Public Health, Robbins College of Human Health and Sciences, Baylor University, Waco, TX 76711, USA
| | - Joan Hittelman
- Department of Psychology, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Lori Hoepner
- Department of Environmental and Occupational Health Sciences, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Lawrence Fordjour
- Department of Pediatrics, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Pavani Chitamanni
- Department of Pediatrics, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Anjana Saxena
- Department of Biology, Brooklyn College of City University of New York, Brooklyn, NY 11210, USA
| | - Xinyin Jiang
- Departments of Health and Nutrition Sciences, Brooklyn College of City University of New York, Brooklyn, NY 11210, USA; PhD Program in Biochemistry, Graduate Center of the City University of New York, New York, NY 10016, USA.
| |
Collapse
|
8
|
Kadam I, Trasino SE, Korsmo H, Lucas J, Pinkas M, Jiang X. Prenatal Choline Supplementation Improves Glucose Tolerance and Reduces Liver Fat Accumulation in Mouse Offspring Exposed to Ethanol during the Prenatal and Postnatal Periods. Nutrients 2024; 16:1264. [PMID: 38732511 PMCID: PMC11085373 DOI: 10.3390/nu16091264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/18/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
Prenatal alcohol exposure (AE) affects cognitive development. However, it is unclear whether prenatal AE influences the metabolic health of offspring and whether postnatal AE exacerbates metabolic deterioration resulting from prenatal AE. Choline is a semi-essential nutrient that has been demonstrated to mitigate the cognitive impairment of prenatal AE. This study investigated how maternal choline supplementation (CS) may modify the metabolic health of offspring with prenatal and postnatal AE (AE/AE). C57BL/6J female mice were fed either a Lieber-DeCarli diet with 1.4% ethanol between embryonic day (E) 9.5 and E17.5 or a control diet. Choline was supplemented with 4 × concentrations versus the control throughout pregnancy. At postnatal week 7, offspring mice were exposed to 1.4% ethanol for females and 3.9% ethanol for males for 4 weeks. AE/AE increased hepatic triglyceride accumulation in male offspring only, which was normalized by prenatal CS. Prenatal CS also improved glucose tolerance compared to AE/AE animals. AE/AE suppressed hepatic gene expression of peroxisome proliferator activated receptor alpha (Ppara) and low-density lipoprotein receptor (Ldlr), which regulate fatty acid catabolism and cholesterol reuptake, respectively, in male offspring. However, these changes were not rectified by prenatal CS. In conclusion, AE/AE led to an increased risk of steatosis and was partially prevented by prenatal CS in male mice.
Collapse
Affiliation(s)
- Isma’il Kadam
- PhD Program in Biochemistry, Graduate Center of the City University of New York, New York, NY 10016, USA; (I.K.); (H.K.)
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA; (J.L.); (M.P.)
| | - Steven E. Trasino
- Nutrition Program, School of Urban Public Health, Hunter College, City University of New York, New York, NY 10065, USA
| | - Hunter Korsmo
- PhD Program in Biochemistry, Graduate Center of the City University of New York, New York, NY 10016, USA; (I.K.); (H.K.)
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA; (J.L.); (M.P.)
| | - Jessica Lucas
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA; (J.L.); (M.P.)
| | - Myriam Pinkas
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA; (J.L.); (M.P.)
| | - Xinyin Jiang
- PhD Program in Biochemistry, Graduate Center of the City University of New York, New York, NY 10016, USA; (I.K.); (H.K.)
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA; (J.L.); (M.P.)
| |
Collapse
|
9
|
Obeid R, Schön C, Derbyshire E, Jiang X, Mellott TJ, Blusztajn JK, Zeisel SH. A Narrative Review on Maternal Choline Intake and Liver Function of the Fetus and the Infant; Implications for Research, Policy, and Practice. Nutrients 2024; 16:260. [PMID: 38257153 PMCID: PMC10820518 DOI: 10.3390/nu16020260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Dietary choline is needed to maintain normal health, including normal liver function in adults. Fatty liver induced by a choline-deficient diet has been consistently observed in human and animal studies. The effect of insufficient choline intake on hepatic fat accumulation is specific and reversible when choline is added to the diet. Choline requirements are higher in women during pregnancy and lactation than in young non-pregnant women. We reviewed the evidence on whether choline derived from the maternal diet is necessary for maintaining normal liver function in the fetus and breastfed infants. Studies have shown that choline from the maternal diet is actively transferred to the placenta, fetal liver, and human milk. This maternal-to-child gradient can cause depletion of maternal choline stores and increase the susceptibility of the mother to fatty liver. Removing choline from the diet of pregnant rats causes fatty liver both in the mother and the fetus. The severity of fatty liver in the offspring was found to correspond to the severity of fatty liver in the respective mothers and to the duration of feeding the choline-deficient diet to the mother. The contribution of maternal choline intake in normal liver function of the offspring can be explained by the role of phosphatidylcholine in lipid transport and as a component of cell membranes and the function of choline as a methyl donor that enables synthesis of phosphatidylcholine in the liver. Additional evidence is needed on the effect of choline intake during pregnancy and lactation on health outcomes in the fetus and infant. Most pregnant and lactating women are currently not achieving the adequate intake level of choline through the diet. Therefore, public health policies are needed to ensure sufficient choline intake through adding choline to maternal multivitamin supplements.
Collapse
Affiliation(s)
- Rima Obeid
- Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, D-66420 Homburg, Germany
| | - Christiane Schön
- BioTeSys GmbH, Nutritional CRO, Schelztorstrasse 54-56, D-73728 Esslingen, Germany
| | | | - Xinyin Jiang
- Department of Health and Nutrition Sciences, Brooklyn College, City University of New York, 4110C Ingersoll Hall, 2900 Bedford Ave., Brooklyn, NY 11210, USA
| | - Tiffany J. Mellott
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Jan Krzysztof Blusztajn
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Steven H. Zeisel
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27514, USA
| |
Collapse
|
10
|
Obeid R, Karlsson T. Choline - a scoping review for Nordic Nutrition Recommendations 2023. Food Nutr Res 2023; 67:10359. [PMID: 38187796 PMCID: PMC10770654 DOI: 10.29219/fnr.v67.10359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 03/15/2022] [Accepted: 11/10/2023] [Indexed: 01/09/2024] Open
Abstract
Choline is an essential nutrient with metabolic roles as a methyl donor in one carbon metabolism and as a precursor for membrane phospholipids and the neurotransmitter acetylcholine. Choline content is particularly high in liver, eggs, and wheat germ, although it is present in a variety of foods. The main dietary sources of choline in the Nordic and Baltic countries are meat, dairy, eggs, and grain. A diet that is devoid of choline causes liver and muscle dysfunction within 3 weeks. Choline requirements are higher during pregnancy and lactation than in non-pregnant women. Although no randomized controlled trials are available, observational studies in human, supported by coherence from interventional studies with neurodevelopmental outcomes and experimental studies in animals, strongly suggest that sufficient intake of choline during pregnancy is necessary for normal brain development and function in the child. Observational studies suggested that adequate intake of choline could have positive effects on cognitive function in older people. However, prospective data are lacking, and no intervention studies are available in the elderly.
Collapse
Affiliation(s)
- Rima Obeid
- Department of Clinical Chemistry and Laboratory Medicine, University Hospital of the Saarland, Homburg, Germany
| | - Therese Karlsson
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
11
|
Martín-Fernández-de-Labastida S, Alegria-Lertxundi I, de Pancorbo MM, Arroyo-Izaga M. Association between nutrient intake related to the one-carbon metabolism and colorectal cancer risk: a case-control study in the Basque Country. Eur J Nutr 2023; 62:3181-3191. [PMID: 37543963 PMCID: PMC10611602 DOI: 10.1007/s00394-023-03229-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 07/27/2023] [Indexed: 08/08/2023]
Abstract
PURPOSE Epidemiologic evidence for the association between methyl-donor nutrient intake and colorectal cancer (CRC) risk remains inconclusive. We aimed to examine the relationship between intake of vitamins of the B group, methionine, total choline and betaine and CRC risk, in a population from the CRC screening programme in the Basque Country. DESIGN This observational study included 308 patients with CRC and 308 age- and sex-matched subjects as controls. During recruitment, dietary, anthropometric, lifestyle, socioeconomic, demographic, and health status information was collected. Conditional logistic regression was used to estimate the odds ratios (ORs) for CRC risk. RESULTS The adjusted ORs for CRC risk decreased with higher intakes of choline and betaine (p < 0.05). After further adjustment for folate, high intake of choline and betaine remained associated with a reduced CRC risk (adjusted model for choline, OR third tertile vs first tertile = 0.45, 95% CI 0.26-0.80, p = 0.006; for betaine, OR third tertile vs first tertile = 0.27, 95% CI 0.16-0.47, p < 0.001). Regarding the other nutrients, our findings indicated a non-significant decrease in CRC risk with the high level of intake. CONCLUSIONS Our data suggest that choline and betaine intake influence CRC risk in the studied population.
Collapse
Grants
- 2011111153 Osasun Saila, Eusko Jaurlaritzako
- S-PE12UN058 Ekonomiaren Garapen eta Lehiakortasun Saila, Eusko Jaurlaritza
- IT1633-22 Hezkuntza, Hizkuntza Politika Eta Kultura Saila, Eusko Jaurlaritza
- PRE_2014_1_161 Hezkuntza, Hizkuntza Politika Eta Kultura Saila, Eusko Jaurlaritza
- PRE_2015_2_0084 Hezkuntza, Hizkuntza Politika Eta Kultura Saila, Eusko Jaurlaritza
- EP_2016_1_0098 Hezkuntza, Hizkuntza Politika Eta Kultura Saila, Eusko Jaurlaritza
- PRE_2017_2_0006 Hezkuntza, Hizkuntza Politika Eta Kultura Saila, Eusko Jaurlaritza
- Universidad del País Vasco
Collapse
Affiliation(s)
- Silvia Martín-Fernández-de-Labastida
- Department of Pharmacy and Food Sciences, Faculty of Pharmacy, University of the Basque Country UPV/EHU, 01006, Vitoria-Gasteiz, Araba/Álava, Spain
| | - Iker Alegria-Lertxundi
- Department of Pharmacy and Food Sciences, Faculty of Pharmacy, University of the Basque Country UPV/EHU, 01006, Vitoria-Gasteiz, Araba/Álava, Spain
| | - Marian M de Pancorbo
- Department of Z. and Cellular Biology A., Faculty of Pharmacy, University of the Basque Country UPV/EHU, 01006, Vitoria-Gasteiz, Araba/Álava, Spain
- BIOMICs Research Group, Microfluidics & BIOMICs Cluster, Lascaray Research Center, University of the Basque Country UPV/EHU, 01006, Vitoria-Gasteiz, Araba/Álava, Spain
- Bioaraba, BA04.03, Vitoria-Gasteiz, Araba/Álava, Spain
| | - Marta Arroyo-Izaga
- Department of Pharmacy and Food Sciences, Faculty of Pharmacy, University of the Basque Country UPV/EHU, 01006, Vitoria-Gasteiz, Araba/Álava, Spain.
- BIOMICs Research Group, Microfluidics & BIOMICs Cluster, Lascaray Research Center, University of the Basque Country UPV/EHU, 01006, Vitoria-Gasteiz, Araba/Álava, Spain.
- Bioaraba, BA04.03, Vitoria-Gasteiz, Araba/Álava, Spain.
| |
Collapse
|
12
|
Minarski M, Maas C, Heinrich C, Böckmann KA, Bernhard W, Shunova A, Poets CF, Franz AR. Choline and Betaine Levels in Plasma Mirror Choline Intake in Very Preterm Infants. Nutrients 2023; 15:4758. [PMID: 38004152 PMCID: PMC10675502 DOI: 10.3390/nu15224758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Choline is essential for cell membrane formation and methyl transfer reactions, impacting parenchymal and neurological development. It is therefore enriched via placental transfer, and fetal plasma concentrations are high. In spite of the greater needs of very low birth weight infants (VLBWI), choline content of breast milk after preterm delivery is lower (median (p25-75): 158 mg/L (61-360 mg/L) compared to term delivery (258 mg/L (142-343 mg/L)). Even preterm formula or fortified breast milk currently provide insufficient choline to achieve physiological plasma concentrations. This secondary analysis of a randomized controlled trial comparing growth of VLBWI with different levels of enteral protein supply aimed to investigate whether increased enteral choline intake results in increased plasma choline, betaine and phosphatidylcholine concentrations. We measured total choline content of breast milk from 33 mothers of 34 VLBWI. Enteral choline intake from administered breast milk, formula and fortifier was related to the respective plasma choline, betaine and phosphatidylcholine concentrations. Plasma choline and betaine levels in VLBWI correlated directly with enteral choline intake, but administered choline was insufficient to achieve physiological (fetus-like) concentrations. Hence, optimizing maternal choline status, and the choline content of milk and fortifiers, is suggested to increase plasma concentrations of choline, ameliorate the choline deficit and improve growth and long-term development of VLBWI.
Collapse
Affiliation(s)
- Michaela Minarski
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
| | - Christoph Maas
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
| | - Christine Heinrich
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
| | - Katrin A. Böckmann
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
| | - Wolfgang Bernhard
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
| | - Anna Shunova
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
| | - Christian F. Poets
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
| | - Axel R. Franz
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
- Center for Pediatric Clinical Studies, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany
| |
Collapse
|
13
|
Gautier MK, Kelley CM, Lee SH, Alldred MJ, McDaid J, Mufson EJ, Stutzmann GE, Ginsberg SD. Maternal choline supplementation protects against age-associated cholinergic and GABAergic basal forebrain neuron degeneration in the Ts65Dn mouse model of Down syndrome and Alzheimer's disease. Neurobiol Dis 2023; 188:106332. [PMID: 37890559 PMCID: PMC10752300 DOI: 10.1016/j.nbd.2023.106332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/02/2023] [Accepted: 10/22/2023] [Indexed: 10/29/2023] Open
Abstract
Down syndrome (DS) is a genetic disorder caused by triplication of human chromosome 21. In addition to intellectual disability, DS is defined by a premature aging phenotype and Alzheimer's disease (AD) neuropathology, including septohippocampal circuit vulnerability and degeneration of basal forebrain cholinergic neurons (BFCNs). The Ts65Dn mouse model recapitulates key aspects of DS/AD pathology, namely age-associated atrophy of BFCNs and cognitive decline in septohippocampal-dependent behavioral tasks. We investigated whether maternal choline supplementation (MCS), a well-tolerated treatment modality, protects vulnerable BFCNs from age- and genotype-associated degeneration in trisomic offspring. We also examined the effect of trisomy, and MCS, on GABAergic basal forebrain parvalbumin neurons (BFPNs), an unexplored neuronal population in this DS model. Unbiased stereological analyses of choline acetyltransferase (ChAT)-immunoreactive BFCNs and parvalbumin-immunoreactive BFPNs were conducted using confocal z-stacks of the medial septal nucleus and the vertical limb of the diagonal band (MSN/VDB) in Ts65Dn mice and disomic (2N) littermates at 3-4 and 10-12 months of age. MCS trisomic offspring displayed significant increases in ChAT-immunoreactive neuron number and density compared to unsupplemented counterparts, as well as increases in the area of the MSN/VDB occupied by ChAT-immunoreactive neuropil. MCS also rescued BFPN number and density in Ts65Dn offspring, a novel rescue of a non-cholinergic cell population. Furthermore, MCS prevented age-associated loss of BFCNs and MSN/VDB regional area in 2N offspring, indicating genotype-independent neuroprotective benefits. These findings demonstrate MCS provides neuroprotection of vulnerable BFCNs and non-cholinergic septohippocampal BFPNs, indicating this modality has translational value as an early life therapy for DS, as well as extending benefits to the aging population at large.
Collapse
Affiliation(s)
- Megan K Gautier
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Pathobiology and Translational Medicine Program, New York University Grossman School of Medicine, New York, NY, USA; NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Christy M Kelley
- Complex Adaptive Systems Initiative, Arizona State University, Tempe, AZ, USA; Institute for Future Health, Scottsdale, AZ, USA
| | - Sang Han Lee
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Melissa J Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - John McDaid
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University/The Chicago Medical School, North Chicago, IL, USA
| | - Elliott J Mufson
- Departments of Translational Neuroscience and Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Grace E Stutzmann
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University/The Chicago Medical School, North Chicago, IL, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA; Department of Neuroscience & Physiology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
14
|
Staskova L, Marx W, Dawson SL, O'Hely M, Mansell T, Saffery R, Burgner D, Collier F, Novakovic B, Vuillermin P, Field CJ, Dewey D, Ponsonby AL. The distribution of dietary choline intake and serum choline levels in Australian women during pregnancy and associated early life factors. Eur J Nutr 2023; 62:2855-2872. [PMID: 37378694 PMCID: PMC10468947 DOI: 10.1007/s00394-023-03186-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 05/31/2023] [Indexed: 06/29/2023]
Abstract
BACKGROUND Maternal dietary choline has a central role in foetal brain development and may be associated with later cognitive function. However, many countries are reporting lower than recommended intake of choline during pregnancy. METHODS Dietary choline was estimated using food frequency questionnaires in pregnant women participating in population-derived birth cohort, the Barwon Infant Study (BIS). Dietary choline is reported as the sum of all choline-containing moieties. Serum total choline-containing compounds (choline-c), phosphatidylcholine and sphingomyelin were measured using nuclear magnetic resonance metabolomics in the third trimester. The main form of analysis was multivariable linear regression. RESULTS The mean daily dietary choline during pregnancy was 372 (standard deviation (SD) 104) mg/day. A total of 236 women (23%) had adequate choline intake (440 mg/day) based on the Australian and New Zealand guidelines, and 27 women (2.6%) took supplemental choline ([Formula: see text] 50 mg/dose) daily during pregnancy. The mean serum choline-c in pregnant women was 3.27 (SD 0.44) mmol/l. Ingested choline and serum choline-c were not correlated (R2) = - 0.005, p = 0.880. Maternal age, maternal weight gain in pregnancy, and a pregnancy with more than one infant were associated with higher serum choline-c, whereas gestational diabetes and environmental tobacco smoke during preconception and pregnancy were associated with lower serum choline-c. Nutrients or dietary patterns were not associated with variation in serum choline-c. CONCLUSION In this cohort, approximately one-quarter of women met daily choline recommendations during pregnancy. Future studies are needed to understand the potential impact of low dietary choline intake during pregnancy on infant cognition and metabolic intermediaries.
Collapse
Affiliation(s)
- Lada Staskova
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Wolfgang Marx
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, VIC, 3220, Australia
| | - Samantha L Dawson
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, VIC, 3220, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, 3052, Australia
| | - Martin O'Hely
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, VIC, 3220, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, 3052, Australia
| | - Toby Mansell
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, 3010, Australia
| | - David Burgner
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Fiona Collier
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, VIC, 3220, Australia
| | - Boris Novakovic
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, 3052, Australia
| | - Peter Vuillermin
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, VIC, 3220, Australia
- Barwon Health, Geelong, VIC, 3220, Australia
| | - Catherine J Field
- Department of Agriculture, Food and Nutritional Science, University of Alberta, 4-126C Li Ka Shing Centre for Research, Edmonton, AB, T6G 2H5, Canada
| | - Deborah Dewey
- Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, Calgary, AB, T3B 6A8, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Owerko Centre, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Anne-Louise Ponsonby
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia.
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, 3052, Australia.
| |
Collapse
|
15
|
Kadam I, Dalloul M, Hausser J, Huntley M, Hoepner L, Fordjour L, Hittelman J, Saxena A, Liu J, Futterman ID, Minkoff H, Jiang X. Associations between nutrients in one-carbon metabolism and fetal DNA methylation in pregnancies with or without gestational diabetes mellitus. Clin Epigenetics 2023; 15:137. [PMID: 37633918 PMCID: PMC10464204 DOI: 10.1186/s13148-023-01554-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/14/2023] [Indexed: 08/28/2023] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM), characterized by hyperglycemia that develops during pregnancy, increases the risk of fetal macrosomia, childhood obesity and cardiometabolic disorders later in life. This process has been attributed partly to DNA methylation modifications in growth and stress-related pathways. Nutrients involved with one-carbon metabolism (OCM), such as folate, choline, betaine, and vitamin B12, provide methyl groups for DNA methylation of these pathways. Therefore, this study aimed to determine whether maternal OCM nutrient intakes and levels modified fetal DNA methylation and in turn altered fetal growth patterns in pregnancies with and without GDM. RESULTS In this prospective study at a single academic institution from September 2016 to June 2019, we recruited 76 pregnant women with and without GDM at 25-33 weeks gestational age and assessed their OCM nutrient intake by diet recalls and measured maternal blood OCM nutrient levels. We also collected placenta and cord blood samples at delivery to examine fetal tissue DNA methylation of the genes that modify fetal growth and stress response such as insulin-like growth factor 2 (IGF2) and corticotropin-releasing hormone (CRH). We analyzed the association between maternal OCM nutrients and fetal DNA methylation using a generalized linear mixed model. Our results demonstrated that maternal choline intake was positively correlated with cord blood CRH methylation levels in both GDM and non-GDM pregnancies (r = 0.13, p = 0.007). Further, the downstream stress hormone cortisol regulated by CRH was inversely associated with maternal choline intake (r = - 0.36, p = 0.021). Higher maternal betaine intake and serum folate levels were associated with lower cord blood and placental IGF2 DNA methylation (r = - 0.13, p = 0.049 and r = - 0.065, p = 0.034, respectively) in both GDM and non-GDM pregnancies. Further, there was an inverse association between maternal betaine intake and birthweight of infants (r = - 0.28, p = 0.015). CONCLUSIONS In conclusion, we observed a complex interrelationship between maternal OCM nutrients and fetal DNA methylation levels regardless of GDM status, which may, epigenetically, program molecular pathways related to fetal growth and stress response.
Collapse
Affiliation(s)
- Isma'il Kadam
- Departments of Health and Nutrition Sciences, Brooklyn College of City University of New York, 2900 Bedford Ave, Brooklyn, NY, 11210, USA
| | - Mudar Dalloul
- Department of Obstetrics and Gynecology, State University of New York Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Jeanette Hausser
- Departments of Health and Nutrition Sciences, Brooklyn College of City University of New York, 2900 Bedford Ave, Brooklyn, NY, 11210, USA
| | - Monique Huntley
- Departments of Health and Nutrition Sciences, Brooklyn College of City University of New York, 2900 Bedford Ave, Brooklyn, NY, 11210, USA
| | - Lori Hoepner
- Department of Environmental and Occupational Health Sciences, State University of New York Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Lawrence Fordjour
- Department of Pediatrics, State University of New York Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Joan Hittelman
- Department of Psychology, State University of New York Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Anjana Saxena
- Departments of Biology, Brooklyn College of City University of New York, Brooklyn, USA
| | - Jia Liu
- Neuroscience Initiative, Advanced Science Research Center at the Graduate Center of the CUNY, New York, NY, 10031, USA
| | - Itamar D Futterman
- Division of Maternal Fetal Medicine, Departments of Obstetrics and Gynecology, Maimonides Medical Center, Brooklyn, USA
| | - Howard Minkoff
- Department of Obstetrics and Gynecology, State University of New York Downstate Health Sciences University, Brooklyn, NY, 11203, USA
- Division of Maternal Fetal Medicine, Departments of Obstetrics and Gynecology, Maimonides Medical Center, Brooklyn, USA
| | - Xinyin Jiang
- Departments of Health and Nutrition Sciences, Brooklyn College of City University of New York, 2900 Bedford Ave, Brooklyn, NY, 11210, USA.
| |
Collapse
|
16
|
Shen L, Shen Y, Zhang Y, Cao S, Yu S, Zong X, Su Z. Effects of Anemoside B4 on Plasma Metabolites in Cows with Clinical Mastitis. Vet Sci 2023; 10:437. [PMID: 37505842 PMCID: PMC10383794 DOI: 10.3390/vetsci10070437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/29/2023] Open
Abstract
Anemoside B4 has a good curative effect on cows with CM; however, its impact on their metabolic profiles is unclear. Based on similar somatic cell counts and clinical symptoms, nine healthy dairy cows and nine cows with CM were selected, respectively. Blood samples were collected from cows with mastitis on the day of diagnosis. Cows with mastitis were injected with anemoside B4 (0.05 mL/kg, once daily) for three consecutive days, and healthy cows were injected with the same volume of normal saline. Subsequently, blood samples were collected. The plasma metabolic profiles were analyzed using untargeted mass spectrometry, and the concentrations of interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α) in serum were evaluated via ELISA. The cows with CM showed increased concentrations of IL-1β, IL-6, and TNF-α (p < 0.05). After treatment with anemoside B4, the concentrations of IL-1β, IL-6, and TNF-α were significantly decreased (p < 0.01). Untargeted metabolomics analysis showed that choline, glycocholic acid, PC (18:0/18:1), 20-HETE, PGF3α, and oleic acid were upregulated in cows with CM. After treatment with anemoside B4, the concentrations of PC (16:0/16:0), PC (18:0/18:1), linoleic acid, eicosapentaenoic acid, phosphorylcholine, and glycerophosphocholine were downregulated, while the LysoPC (14:0), LysoPC (18:0), LysoPC (18:1), and cis-9-palmitoleic acid were upregulated. This study indicated that anemoside B4 alleviated the inflammatory response in cows with CM mainly by regulating lipid metabolism.
Collapse
Affiliation(s)
- Liuhong Shen
- The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yu Shen
- The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yue Zhang
- The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Suizhong Cao
- The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Shumin Yu
- The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaolan Zong
- The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhetong Su
- Guangxi Innovates Medical Technology Co., Ltd., Lipu 546600, China
| |
Collapse
|
17
|
Shanmuganathan M, Bogert M, Kroezen Z, Britz-McKibbin P, Atkinson SA. Dynamic Metabolic Signatures of Choline and Carnitine across Healthy Pregnancy and in Cord Blood: Association with Maternal Dietary Protein. J Nutr 2023; 153:999-1007. [PMID: 36780943 DOI: 10.1016/j.tjnut.2023.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/31/2023] [Accepted: 02/08/2023] [Indexed: 02/13/2023] Open
Abstract
BACKGROUND In pregnancy, choline is deemed an essential nutrient and carnitine needs are increased, but amounts remain undefined. OBJECTIVES We aimed to measure choline and total dietary protein and dairy protein intake from food and supplements across pregnancy and the response to intake by profiling choline and carnitine metabolites across pregnancy and in cord blood. METHODS An exploratory analysis of choline and protein intake from 3-d diet records and measures of 36 serum choline and carnitine metabolites in early (12-17 wk) and late (36-38 wk) pregnancy was conducted in participants from the Be Healthy in Pregnancy study randomized to high dairy protein+walking exercise or usual care. Metabolites were measured in fasted maternal and cord serum using multisegment injection-capillary electrophoresis-mass spectrometry. Mixed ANOVA adjusted for body mass index was performed for comparison of metabolites across pregnancy and between intervention and control. RESULTS In 104 participants, the median (Q1, Q3) total choline intake was 347 (263, 427) mg/d in early and 322 (270, 437) mg/d in late pregnancy. Only ∼20% of participants achieved the recommended adequate intake (450 mg/d) and ∼10% consumed supplemental choline (8-200 mg/d). Serum-free choline (μmol/L) was higher in late compared with early pregnancy [12.9 (11.4, 15.1) compared with 9.68 (8.25, 10.61), P < 0.001], but choline downstream metabolites were similar across pregnancy. Serum carnitine [10.3 (9.01, 12.2) compared with 15.9 (14.1, 17.9) μmol/L, P < 0.001] and acetylcarnitine [2.35 (1.92, 2.68) compared with 3.0 (2.56, 3.59), P < 0.001] were significantly lower in late pregnancy. High cord:maternal serum metabolite ratios were found in most measured metabolites. CONCLUSIONS Despite inadequate choline intake, serum-free choline was elevated in late pregnancy and enriched in cord blood compared with maternal serum. Serum carnitine declined in late pregnancy despite a high protein diet. The higher cord:maternal concentrations in choline and carnitine metabolites suggest active uptake in late pregnancy, reflecting the importance of these circulating metabolites in fetal development. This trial was registered at clinicaltrials.gov as NCT01689961.
Collapse
Affiliation(s)
- Meera Shanmuganathan
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Meghan Bogert
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Zachary Kroezen
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Philip Britz-McKibbin
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | | |
Collapse
|
18
|
Wiedeman AM, Dhillon AK, Wu BT, Innis SM, Elango R, Devlin AM. Children Aged 5-6 Years in Vancouver, Canada Meet Dietary Recommendations for Folate and Vitamin B12 but not Choline. J Nutr 2023; 153:197-207. [PMID: 36913454 DOI: 10.1016/j.tjnut.2022.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/17/2022] [Accepted: 11/14/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Choline, folate, and vitamin B12 are required for growth and development, but there is limited information on the intakes and relationships to biomarkers of status in children. OBJECTIVES The objective of this study was to determine the choline and B-vitamin intakes and relationship to biomarkers of status in children. METHODS A cross-sectional study was conducted in children (n = 285, aged 5-6 y) recruited from Metro Vancouver, Canada. Dietary information was collected by using 3 24-h recalls. Nutrient intakes were estimated by using the Canadian Nutrient File and United States Department of Agriculture database for choline. Supplement information was collected by using questionnaires. Plasma biomarkers were quantified by using mass spectrometry and commercial immunoassays, and relationships to dietary and supplement intake were determined by using linear models. RESULTS Daily dietary intakes of choline, folate, and vitamin B12 were [mean (SD)] 249 (94.3) mg, 330 (120) DFE μg, and 3.60 (1.54) μg, respectively. Top food sources of choline and vitamin B12 were dairy, meats, and eggs (63%-84%) and for folate, were grains, fruits, and vegetables (67%). More than half of the children (60%) were consuming a supplement containing B-vitamins, but not choline. Only 40% of children met the choline adequate intake (AI) recommendation for North America (≥250 mg/d); 82% met the European AI (≥170 mg/d). Less than 3% of children had inadequate folate and vitamin B12 total intakes. Some children (5%) had total folic acid intakes above the North American tolerable upper intake level (UL; >400 μg/d); 10% had intakes above the European UL (>300 μg/d). Dietary choline intake was positively associated with plasma dimethylglycine, and total vitamin B12 intake was positively associated with plasma B12 (adjusted models; P < 0.001). CONCLUSIONS These findings suggest that many children are not meeting the dietary choline recommendations, and some children may have excessive folic acid intakes. The impact of imbalanced one-carbon nutrient intakes during this active period of growth and development requires further investigation.
Collapse
Affiliation(s)
- Alejandra M Wiedeman
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital Research Institute, Vancouver, Canada
| | - Amneet K Dhillon
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital Research Institute, Vancouver, Canada
| | - Brian T Wu
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital Research Institute, Vancouver, Canada
| | - Sheila M Innis
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital Research Institute, Vancouver, Canada
| | - Rajavel Elango
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital Research Institute, Vancouver, Canada
| | - Angela M Devlin
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital Research Institute, Vancouver, Canada.
| |
Collapse
|
19
|
Obeid R, Derbyshire E, Schön C. Association between Maternal Choline, Fetal Brain Development, and Child Neurocognition: Systematic Review and Meta-Analysis of Human Studies. Adv Nutr 2022; 13:2445-2457. [PMID: 36041182 PMCID: PMC9776654 DOI: 10.1093/advances/nmac082] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 10/01/2021] [Accepted: 07/25/2022] [Indexed: 01/29/2023] Open
Abstract
We studied associations between prenatal and early postnatal choline intake, brain development, and neurocognitive function of children. We conducted a systematic review followed by a meta-analysis and critical appraisal of human studies published from 1997 to 2021. Thirty publications were identified. The meta-analysis included 5 of 7 case-control studies studying neural tube defects (NTDs) in relation to maternal choline intakes/circulating concentrations. Low maternal choline intake/circulating concentrations were associated with a higher OR for NTDs among 1131 mothers of newborns with NTDs and 4439 control mothers (pooled estimate = 1.36; 95% CI: 1.11, 1.67). The 95% prediction intervals were 0.78, 2.36. Findings and critical evaluation of 10 publications with interventional designs showed that higher maternal choline intakes during the second half of pregnancy and early postnatal period (550 mg up to 1 g/d on top of the diet) or a child intake of 513 to 625 mg/d from supplements were safe and likely to demonstrate favorable effects on several domains of child neurocognition, such as memory, attention, and visuospatial learning versus the comparators. Findings from observational studies (n = 13) partly supported the association between maternal choline intake/serum concentrations and child neurocognition, but there was low confidence in the use of plasma choline concentrations as a choline intake marker. In conclusion, low maternal choline intakes were associated with a higher OR for NTDs. The risk could be up to 2.36-fold in some populations. Despite limitations of available trials and observational studies, higher maternal choline intake was likely to be associated with better child neurocognition/neurodevelopment. The results should be used to guide choline intake recommendations in pregnancy and lactation, especially because most young women are not achieving the reference intake of choline. This meta-analysis is registered at PROSPERO as CRD42021233790.
Collapse
|
20
|
A Study of the Metabolic Pathways Affected by Gestational Diabetes Mellitus: Comparison with Type 2 Diabetes. Diagnostics (Basel) 2022; 12:diagnostics12112881. [PMID: 36428943 PMCID: PMC9689375 DOI: 10.3390/diagnostics12112881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) remains incompletely understood and increases the risk of developing Diabetes mellitus type 2 (DM2). Metabolomics provides insights etiology and pathogenesis of disease and discovery biomarkers for accurate detection. Nuclear magnetic resonance (NMR) spectroscopy is a key platform defining metabolic signatures in intact serum/plasma. In the present study, we used NMR-based analysis of macromolecules free-serum to accurately characterize the altered metabolic pathways of GDM and assessing their similarities to DM2. Our findings could contribute to the understanding of the pathophysiology of GDM and help in the identification of metabolomic markers of the disease. METHODS Sixty-two women with GDM matched with seventy-seven women without GDM (control group). 1H NMR serum spectra were acquired on an 11.7 T Bruker Avance DRX NMR spectrometer. RESULTS We identified 55 metabolites in both groups, 25 of which were significantly altered in the GDM group. GDM group showed elevated levels of ketone bodies, 2-hydroxybutyrate and of some metabolic intermediates of branched-chain amino acids (BCAAs) and significantly lower levels of metabolites of one-carbon metabolism, energy production, purine metabolism, certain amino acids, 3-methyl-2-oxovalerate, ornithine, 2-aminobutyrate, taurine and trimethylamine N-oxide. CONCLUSION Metabolic pathways affected in GDM were beta-oxidation, ketone bodies metabolism, one-carbon metabolism, arginine and ornithine metabolism likewise in DM2, whereas BCAAs catabolism and aromatic amino acids metabolism were affected, but otherwise than in DM2.
Collapse
|
21
|
Evidence based recommendations for an optimal prenatal supplement for women in the US: vitamins and related nutrients. Matern Health Neonatol Perinatol 2022; 8:4. [PMID: 35818085 PMCID: PMC9275129 DOI: 10.1186/s40748-022-00139-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/13/2022] [Indexed: 01/21/2023] Open
Abstract
The blood levels of most vitamins decrease during pregnancy if un-supplemented, including vitamins A, C, D, K, B1, B3, B5, B6, folate, biotin, and B12. Sub-optimal intake of vitamins from preconception through pregnancy increases the risk of many pregnancy complications and infant health problems. In the U.S., dietary intake of vitamins is often below recommended intakes, especially for vitamin D, choline and DHA. Many studies suggest that insufficient vitamin intake is associated with a wide range of pregnancy complications (anemia, Cesarean section, depression, gestational diabetes, hypertension, infertility, preeclampsia, and premature rupture of membranes) and infant health problems (asthma/wheeze, autism, low birth weight, congenital heart defects, intellectual development, intrauterine growth restriction, miscarriage, neural tube defects, orofacial defects, and preterm birth). The primary goal of this paper is to review the research literature and propose evidence-based recommendations for the optimal level of prenatal supplementation for each vitamin for most women in the United States. A secondary goal was to compare these new recommendations with the levels of vitamins in over 180 commercial prenatal supplements. The analysis found that prenatal supplements vary widely in content, often contained only a subset of essential vitamins, and the levels were often below our recommendations. This suggests that increasing prenatal vitamin supplementation to the levels recommended here may reduce the incidence of many pregnancy complications and infant health problems which currently occur.
Collapse
|
22
|
Swartz T, Bradford B, Malysheva O, Caudill M, Mamedova L, Estes K. Effects of dietary rumen-protected choline supplementation on colostrum yields, quality, and choline metabolites from dairy cattle. JDS COMMUNICATIONS 2022; 3:296-300. [PMID: 36338018 PMCID: PMC9623657 DOI: 10.3168/jdsc.2021-0192] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/10/2022] [Indexed: 05/27/2023]
Abstract
Colostrum is a critical nutrient source that provides passive immunity to dairy calves. Choline is a trimethylated molecule that is frequently supplemented in the diet to periparturient dairy cows to support postpartum health and performance. Whereas choline and its metabolites have been characterized in milk, the effects of dietary rumen-protected choline (RPC) supplementation on choline metabolites in colostrum from dairy cattle have yet to be explored. Therefore, the objective of the present study was to assess the effects of dietary supplementation and dose of RPC on colostrum yields, quality, and choline metabolites. Parous Holstein cows were blocked by calving month and randomly assigned within block to receive 45 g/d (20.4 g/d of choline ions) of RPC (CHOL45, n = 22), 30 g/d (13.6 g/d of choline ions) of RPC (CHOL30, n = 20), or no RPC (control, n = 19) starting 24 d before expected calving. The effects of dietary supplementation and dose of RPC were assessed on colostrum yields, component yields, somatic cell score (SCS), quality (as assessed by Brix), and choline metabolites. Data were analyzed using a linear mixed model with the fixed effects of treatment, parity, and the 2-way interaction and the random effect of block. Regardless of dose, dietary RPC supplementation increased colostrum yields and protein yields. No effects of dietary RPC supplementation were found on colostrum component percentages, SCS, or colostrum quality. For choline metabolites, treatment interacted with parity for phosphocholine where colostrum from second-parity CHOL45 and CHOL30 cows had greater concentrations of phosphocholine than colostrum from second-parity control cows, but no treatment effect was seen in the colostrum from 3+ parity cows. Dietary choline supplementation, regardless of dose, increased trimethylamine N-oxide concentrations. Dietary choline supplementation did not affect the concentrations of choline, betaine, glycerophosphocholine, sphingomyelin, phosphatidylcholine, or total choline in colostrum. In conclusion, dietary choline supplementation increased phosphocholine concentrations in colostrum from second-parity cows, enhanced trimethylamine N-oxide concentrations, and increased colostrum yields without affecting colostrum quality.
Collapse
Affiliation(s)
- T.H. Swartz
- Department of Animal Science, Michigan State University, East Lansing 48824
| | - B.J. Bradford
- Department of Animal Science, Michigan State University, East Lansing 48824
| | - O. Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853
| | - M.A. Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853
| | - L.K. Mamedova
- Department of Animal Science, Michigan State University, East Lansing 48824
| | - K.A. Estes
- Balchem Corporation, New Hampton, NY 10958
| |
Collapse
|
23
|
Hasina Z, Wang N, Wang CC. Developmental Neuropathology and Neurodegeneration of Down Syndrome: Current Knowledge in Humans. Front Cell Dev Biol 2022; 10:877711. [PMID: 35676933 PMCID: PMC9168127 DOI: 10.3389/fcell.2022.877711] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/18/2022] [Indexed: 12/25/2022] Open
Abstract
Individuals with Down syndrome (DS) suffer from developmental delay, intellectual disability, and an early-onset of neurodegeneration, Alzheimer’s-like disease, or precocious dementia due to an extra chromosome 21. Studying the changes in anatomical, cellular, and molecular levels involved may help to understand the pathogenesis and develop target treatments, not just medical, but also surgical, cell and gene therapy, etc., for individuals with DS. Here we aim to identify key neurodevelopmental manifestations, locate knowledge gaps, and try to build molecular networks to better understand the mechanisms and clinical importance. We summarize current information about the neuropathology and neurodegeneration of the brain from conception to adulthood of foetuses and individuals with DS at anatomical, cellular, and molecular levels in humans. Understanding the alterations and characteristics of developing Down syndrome will help target treatment to improve the clinical outcomes. Early targeted intervention/therapy for the manifestations associated with DS in either the prenatal or postnatal period may be useful to rescue the neuropathology and neurodegeneration in DS.
Collapse
Affiliation(s)
- Zinnat Hasina
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Nicole Wang
- School of Veterinary Medicine, Glasgow University, Glasgow, United Kingdom
| | - Chi Chiu Wang
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong -Sichuan University Joint Laboratory in Reproductive Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- *Correspondence: Chi Chiu Wang,
| |
Collapse
|
24
|
Thomas MS, Puglisi M, Malysheva O, Caudill MA, Sholola M, Cooperstone JL, Fernandez ML. Eggs Improve Plasma Biomarkers in Patients with Metabolic Syndrome Following a Plant-Based Diet-A Randomized Crossover Study. Nutrients 2022; 14:nu14102138. [PMID: 35631279 PMCID: PMC9147178 DOI: 10.3390/nu14102138] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 12/12/2022] Open
Abstract
Plant-based (PB) diets are considered a healthy dietary pattern; however, eggs are not always included in this dietary regime. We hypothesized that the addition of two eggs per day would increase HDL cholesterol as well as plasma lutein, zeaxanthin and choline in individuals with metabolic syndrome (MetS). In this randomized controlled crossover intervention, we recruited 30 participants (49.3 ± 8 y) with MetS who followed a PB diet for 13 weeks. A registered dietitian advised all subjects on food selection and followed them through the intervention to ensure compliance. Participants underwent a 2-week washout with no eggs or spinach (a source of dietary lutein and zeaxanthin) and were randomly allocated to consume spinach (70 g) with either two eggs (EGG) or the equivalent amount of egg substitute (SUB) for breakfast for 4 weeks. After a 3-week washout, they were allocated the alternate breakfast. A total of 24 participants (13 women/11 men) finished the intervention. Plasma lipids, glucose, insulin, anthropometrics, plasma lutein, zeaxanthin, choline and trimethylamine oxide (TMAO) were assessed at baseline and the end of each intervention. When we compared individuals consuming the EGG versus the SUB breakfast, we observed a lower body weight (p < 0.02) and a higher HDL cholesterol (p < 0.025) after the EGG diet. There were no differences in plasma LDL cholesterol, triglycerides, glucose, insulin, or blood pressure. The number of large HDL particles measured by NMR was higher after EGG (p < 0.01) as compared to SUB. Plasma choline was higher in both treatments (p < 0.01) compared to baseline (8.3 ± 2.1 μmol/L). However, plasma choline values were higher in EGG (10.54 ± 2.8 μmol/L) compared to SUB (9.47 ± 2.7 μmol/L) p < 0.025. Both breakfasts increased plasma lutein compared to baseline (p < 0.01), while plasma zeaxanthin was only increased in the egg intervention (p < 0.01). These results indicate that consuming a plant-based diet in combination with whole eggs increases plasma HDL cholesterol, choline and zeaxanthin, important biomarkers in subjects with MetS.
Collapse
Affiliation(s)
- Minu S. Thomas
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA; (M.S.T.); (M.P.)
| | - Michael Puglisi
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA; (M.S.T.); (M.P.)
| | - Olga Malysheva
- Department of Human Nutrition, Division of Nutritional Science, Cornell University, Ithaca, NY 14860, USA; (O.M.); (M.A.C.)
| | - Marie A. Caudill
- Department of Human Nutrition, Division of Nutritional Science, Cornell University, Ithaca, NY 14860, USA; (O.M.); (M.A.C.)
| | - Maria Sholola
- Department of Food Science and Technology, The Ohio State University, Columbus, OH 43210, USA; (M.S.); (J.L.C.)
| | - Jessica L. Cooperstone
- Department of Food Science and Technology, The Ohio State University, Columbus, OH 43210, USA; (M.S.); (J.L.C.)
- Department of Horticulture and Crop Science, The Ohio State University, Columbus, OH 43210, USA
| | - Maria Luz Fernandez
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA; (M.S.T.); (M.P.)
- Correspondence:
| |
Collapse
|
25
|
Klatt KC, McDougall MQ, Malysheva OV, Taesuwan S, Loinard-González A(AP, Nevins JEH, Beckman K, Bhawal R, Anderson E, Zhang S, Bender E, Jackson KH, King DJ, Dyer RA, Devapatla S, Vidavalur R, Brenna JT, Caudill MA. Prenatal choline supplementation improves biomarkers of maternal docosahexaenoic acid (DHA) status among pregnant participants consuming supplemental DHA: a randomized controlled trial. Am J Clin Nutr 2022; 116:820-832. [PMID: 35575618 PMCID: PMC9437984 DOI: 10.1093/ajcn/nqac147] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/01/2022] [Accepted: 06/10/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Dietary methyl donors (e.g., choline) support the activity of the phosphatidylethanolamine N-methyltransferase (PEMT) pathway, which generates phosphatidylcholine (PC) molecules enriched in DHA that are exported from the liver and made available to extrahepatic tissues. OBJECTIVES This study investigated the effect of prenatal choline supplementation on biomarkers of DHA status among pregnant participants consuming supplemental DHA. METHODS Pregnant participants (n = 30) were randomly assigned to receive supplemental choline intakes of 550 mg/d [500 mg/d d0-choline + 50 mg/d deuterium-labeled choline (d9-choline); intervention] or 25 mg/d (25 mg/d d9-choline; control) from gestational week (GW) 12-16 until delivery. All participants received a daily 200-mg DHA supplement and consumed self-selected diets. Fasting blood samples were obtained at baseline, GW 20-24, and GW 28-32; maternal/cord blood was obtained at delivery. Mixed-effects linear models were used to assess the impact of prenatal choline supplementation on maternal and newborn DHA status. RESULTS Choline supplementation (550 vs. 25 mg/d) did not achieve a statistically significant intervention × time interaction for RBC PC-DHA (P = 0.11); a significant interaction was observed for plasma PC-DHA and RBC total DHA, with choline supplementation yielding higher levels (+32-38% and +8-11%, respectively) at GW 28-32 (P < 0.05) and delivery (P < 0.005). A main effect of choline supplementation on plasma total DHA was also observed (P = 0.018); its interaction with time was not significant (P = 0.068). Compared with controls, the intervention group exhibited higher (P = 0.007; main effect) plasma enrichment of d3-PC (d3-PC/total PC). Moreover, the ratio of d3-PC to d9-PC was higher (+50-67%; P < 0.001) in the choline intervention arm (vs. control) at GW 20-24, GW 28-32, and delivery. CONCLUSIONS Prenatal choline supplementation improves hepatic DHA export and biomarkers of DHA status by bolstering methyl group supply for PEMT activity among pregnant participants consuming supplemental DHA. This trial is registered at www.clinicaltrials.gov as NCT03194659.
Collapse
Affiliation(s)
| | | | - Olga V Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Siraphat Taesuwan
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA,Faculty of Agro-Industry, Chiang Mai University, Chiang Mai, Thailand
| | | | - Julie E H Nevins
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Kara Beckman
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Ruchika Bhawal
- Proteomics and Metabolomics Facility, Cornell University, Ithaca, NY, USA
| | - Elizabeth Anderson
- Proteomics and Metabolomics Facility, Cornell University, Ithaca, NY, USA
| | - Sheng Zhang
- Proteomics and Metabolomics Facility, Cornell University, Ithaca, NY, USA
| | - Erica Bender
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | | | - D Janette King
- The Analytical Core for Metabolomics and Nutrition, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Roger A Dyer
- The Analytical Core for Metabolomics and Nutrition, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | - J Thomas Brenna
- Department of Pediatrics, University of Texas, Austin, TX, USA
| | | |
Collapse
|
26
|
Steane SE, Kumar V, Cuffe JSM, Moritz KM, Akison LK. Prenatal Choline Supplementation Alters One Carbon Metabolites in a Rat Model of Periconceptional Alcohol Exposure. Nutrients 2022; 14:nu14091874. [PMID: 35565848 PMCID: PMC9100923 DOI: 10.3390/nu14091874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/16/2022] Open
Abstract
Prenatal alcohol exposure disturbs fetal and placental growth and can alter DNA methylation (DNAm). Supplementation with the methyl donor choline can increase fetal and placental growth and restore DNAm, suggesting converging effects on one-carbon metabolism (1CM). We investigated the impact of periconceptional ethanol (PCE) exposure and prenatal choline supplementation on 1CM in maternal, placental, and fetal compartments. Female Sprague Dawley rats were given a liquid diet containing 12.5% ethanol (PCE) or 0% ethanol (control) for 4 days before and 4 days after conception. Dams were then placed on chow with different concentrations of choline (1.6 g, 2.6 g, or 7.2 g choline/kg chow). Plasma and tissues were collected in late gestation for the analysis of 1CM components by means of mass spectrometry and real-time PCR. PCE reduced placental components of 1CM, particularly those relating to folate metabolism, resulting in a 3−7.5-fold reduction in the ratio of s-adenosylmethionine:s-adenosylhomocysteine (SAM:SAH) (p < 0.0001). Choline supplementation increased placental 1CM components and the SAM:SAH ratio (3.5−14.5-fold, p < 0.0001). In the maternal and fetal compartments, PCE had little effect, whereas choline increased components of 1CM. This suggests that PCE impairs fetal development via altered placental 1CM, highlighting its role in modulating nutritional inputs to optimize fetal development.
Collapse
Affiliation(s)
- Sarah E. Steane
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia; (S.E.S.); (V.K.); (J.S.M.C.); (K.M.M.)
| | - Vinod Kumar
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia; (S.E.S.); (V.K.); (J.S.M.C.); (K.M.M.)
| | - James S. M. Cuffe
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia; (S.E.S.); (V.K.); (J.S.M.C.); (K.M.M.)
| | - Karen M. Moritz
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia; (S.E.S.); (V.K.); (J.S.M.C.); (K.M.M.)
- Child Health Research Centre, The University of Queensland, South Brisbane, QLD 4101, Australia
| | - Lisa K. Akison
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia; (S.E.S.); (V.K.); (J.S.M.C.); (K.M.M.)
- Child Health Research Centre, The University of Queensland, South Brisbane, QLD 4101, Australia
- Correspondence:
| |
Collapse
|
27
|
Maternal and neonatal one-carbon metabolites and the epigenome-wide infant response. J Nutr Biochem 2022; 101:108938. [PMID: 35017001 PMCID: PMC8847320 DOI: 10.1016/j.jnutbio.2022.108938] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 11/10/2021] [Accepted: 12/22/2021] [Indexed: 12/24/2022]
Abstract
Maternal prenatal status, as encapsulated by that to which a mother is exposed through diet and environment, is a key determinant of offspring health and disease. Alterations in DNA methylation (DNAm) may be a mechanism through which suboptimal prenatal conditions confer disease risk later in life. One-carbon metabolism (OCM) is critical to both fetal development and in supplying methyl donors needed for DNAm. Plasma concentrations of one-carbon metabolites across maternal first trimester (M1), maternal term (M3), and infant cord blood (CB) at birth were tested for association with DNAm patterns in CB from the Michigan Mother and Infant Pairs (MMIP) pregnancy cohort. The Illumina Infinium MethylationEPIC BeadChip was used to quantitatively evaluate DNAm across the epigenome. Global and single-site DNAm and metabolite models were adjusted for infant sex, estimated cell type proportions, and batch as covariates. Change in mean metabolite concentration across pregnancy (M1 to M3) was significantly different for S-adenosylhomocysteine (SAH), S-adenosylmethionine (SAM), betaine, and choline. Both M1 SAH and CB SAH were significantly associated with the global distribution of DNAm in CB, with indications of a shift toward less methylation. M3 SAH and CB SAH also displayed significant associations with locus-specific DNAm in infant CB (FDR<0.05). Our findings underscore the role of maternal one-carbon metabolites in shifting the global DNAm pattern in CB and emphasizes the need to closely evaluate how dietary status influences cellular methylation potential and ultimately offspring health.
Collapse
|
28
|
Korsmo HW, Dave B, Trasino S, Saxena A, Liu J, Caviglia JM, Edwards K, Dembitzer M, Sheeraz S, Khaldi S, Jiang X. Maternal Choline Supplementation and High-Fat Feeding Interact to Influence DNA Methylation in Offspring in a Time-Specific Manner. Front Nutr 2022; 9:841787. [PMID: 35165655 PMCID: PMC8837519 DOI: 10.3389/fnut.2022.841787] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/07/2022] [Indexed: 11/18/2022] Open
Abstract
Maternal methyl donor supplementation during pregnancy has demonstrated lasting influence on offspring DNA methylation. However, it is unknown whether an adverse postnatal environment, such as high-fat (HF) feeding, overrides the influence of prenatal methyl donor supplementation on offspring epigenome. In this study, we examined whether maternal supplementation of choline (CS), a methyl donor, interacts with prenatal and postnatal HF feeding to alter global and site-specific DNA methylation in offspring. We fed wild-type C57BL/6J mouse dams a HF diet with or without CS throughout gestation. After weaning, the offspring were exposed to HF feeding for 6 weeks resembling a continued obesogenic environment. Our results suggest that maternal CS under the HF condition (HFCS) increased global DNA methylation and DNA methyltransferase 1 (Dnmt1) expression in both fetal liver and brain. However, during the postnatal period, HFCS offspring demonstrated lower global DNA methylation and Dnmt1 expression was unaltered in both the liver and visceral adipose tissue. Site-specific DNA methylation analysis during both fetal and postnatal periods demonstrated that HFCS offspring had higher methylation of CpGs in the promoter of Srebf1, a key mediator of de novo lipogenesis. In conclusion, the influence of maternal CS on offspring DNA methylation is specific to HF feeding status during prenatal and postnatal periods. Without continued CS during the postnatal period, global DNA methylation enhanced by prenatal CS in the offspring was overridden by postnatal HF feeding.
Collapse
Affiliation(s)
- Hunter W. Korsmo
- Department of Biochemistry, The Graduate Center, City University of New York (CUNY), New York, NY, United States
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Bhoomi Dave
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Steven Trasino
- School of Urban Public Health, Hunter College of the CUNY, New York, NY, United States
| | - Anjana Saxena
- Department of Biochemistry, The Graduate Center, City University of New York (CUNY), New York, NY, United States
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Jia Liu
- Advanced Science Research Center at the Graduate Center of the CUNY, New York, NY, United States
| | - Jorge Matias Caviglia
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Kaydine Edwards
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Moshe Dembitzer
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Shameera Sheeraz
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Sarah Khaldi
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Xinyin Jiang
- Department of Biochemistry, The Graduate Center, City University of New York (CUNY), New York, NY, United States
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| |
Collapse
|
29
|
Zhong W, Hu L, Zhao Y, Li Z, Zhuo Y, Jiang X, Li J, Zhao X, Che L, Feng B, Lin Y, Xu S, Fang Z, Wu D. Effects of Dietary Choline Levels During Pregnancy on Reproductive Performance, Plasma Metabolome and Gut Microbiota of Sows. Front Vet Sci 2022; 8:771228. [PMID: 35141305 PMCID: PMC8818960 DOI: 10.3389/fvets.2021.771228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/15/2021] [Indexed: 12/05/2022] Open
Abstract
This study investigated the effects of dietary choline levels during gestation on reproductive performance of sows. In addition, the plasma metabolome and gut microbiota of sows was studied. A total of 260 multiparous sows were allocated to five dietary treatment groups with increasing choline concentrations (1,050, 1,450, 1,850, 2,250, and 2,650 mg/kg) in a randomized complete block design. The sows were fed experimental diets from breeding until farrowing and a common lactating diet during lactation. The results showed that the backfat (BF) gain of sows during gestation, individual birth weight for total piglets born, piglets born alive, average piglet weight at weaning increased linearly (P < 0.05), whereas the within-litter birth weight variation coefficient (CV) of piglets born alive and suckling piglet mortality decreased linearly (P < 0.05) as dietary choline level increased. A quadratic effect of dietary choline level was observed for the average daily feed intake (ADFI) of sows during lactation (P < 0.05). ADFI was maximized when the dietary choline concentration reached 1,910 mg/kg. Plasma H2O2 concentration at day 30 of gestation in the 1,050 mg/kg group was greater than that in the 1,850 and 2,650 mg/kg groups (P < 0.05). Plasma metabolomics identified 46 metabolites among the three groups. Specifically, plasma concentrations of trimethylamine-N-oxide (TMAO), dopamine, and L-proline increased while 1-methylhistidine concentration decreased as dietary choline levels increased. In addition, bacterial observed species and richness (Chao 1 and ACE) at day 110 of gestation decreased as dietary choline levels increased (P < 0.05). For the gut microbiota composition, the enhanced dietary choline level decreased the abundance of phylum Proteobacteria (P < 0.05) and increased the abundance of phylum Actinobacteria (P < 0.05) at day 30 of gestation. Compared with the 1,050 mg/kg group, the abundance of genus Terrisporobacter was less in the 1,850 mg/kg group, and genera Bacillus and Cellulomonas were greater in the 2,650 mg/kg group. In summary, increasing dietary choline levels improved the birth weight, uniformity of neonatal piglets and litter performance during lactation. This may be associated with better antioxidant capability, metabolic status, and gut microbiota of sows during gestation.
Collapse
Affiliation(s)
- Wei Zhong
- Key Laboratory for Animal Disease-Resistance Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Liang Hu
- College of Food Science, Sichuan Agricultural University, Ya'an, China
| | - Yang Zhao
- Key Laboratory for Animal Disease-Resistance Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Zhen Li
- Key Laboratory for Animal Disease-Resistance Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Yong Zhuo
- Key Laboratory for Animal Disease-Resistance Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Xuemei Jiang
- Key Laboratory for Animal Disease-Resistance Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Jian Li
- Key Laboratory for Animal Disease-Resistance Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Xilun Zhao
- Key Laboratory for Animal Disease-Resistance Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Lianqiang Che
- Key Laboratory for Animal Disease-Resistance Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Bin Feng
- Key Laboratory for Animal Disease-Resistance Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Yan Lin
- Key Laboratory for Animal Disease-Resistance Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Shengyu Xu
- Key Laboratory for Animal Disease-Resistance Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Zhengfeng Fang
- Key Laboratory for Animal Disease-Resistance Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - De Wu
- Key Laboratory for Animal Disease-Resistance Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
30
|
OUP accepted manuscript. Nutr Rev 2022; 80:1985-2001. [DOI: 10.1093/nutrit/nuac015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
31
|
Bahnfleth CL, Strupp BJ, Caudill MA, Canfield RL. Prenatal choline supplementation improves child sustained attention: A 7-year follow-up of a randomized controlled feeding trial. FASEB J 2021; 36:e22054. [PMID: 34962672 PMCID: PMC9303951 DOI: 10.1096/fj.202101217r] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/22/2021] [Accepted: 11/08/2021] [Indexed: 01/23/2023]
Abstract
Numerous rodent studies demonstrate developmental programming of offspring cognition by maternal choline intake, with prenatal choline deprivation causing lasting adverse effects and supplemental choline producing lasting benefits. Few human studies have evaluated the effect of maternal choline supplementation on offspring cognition, with none following children to school age. Here, we report results from a controlled feeding study in which pregnant women were randomized to consume 480 mg choline/d (approximately the Adequate Intake [AI]) or 930 mg choline/d during the 3rd trimester. Sustained attention was assessed in the offspring at age 7 years (n = 20) using a signal detection task that showed benefits of maternal choline supplementation in a murine model. Children in the 930 mg/d group showed superior performance (vs. 480 mg/d group) on the primary endpoint (SAT score, p = .02) and a superior ability to maintain correct signal detections (hits) across the 12‐min session (p = .02), indicative of improved sustained attention. This group difference in vigilance decrement varied by signal duration (p = .04). For the briefest (17 ms) signals, the 480 mg/d group showed a 22.9% decline in hits across the session compared to a 1.5% increase in hits for the 930 mg/d group (p = .04). The groups did not differ in vigilance decrement for 29 or 50 ms signals. This pattern suggests an enhanced ability to sustain perceptual amplification of a brief low‐contrast visual signal by children in the 930 mg/d group. This inference of improved sustained attention by the 930 mg/d group is strengthened by the absence of group differences for false alarms, omissions, and off‐task behaviors. This pattern of results indicates that maternal 3rd trimester consumption of the choline AI for pregnancy (vs. double the AI) produces offspring with a poorer ability to sustain attention—reinforcing concerns that, on average, choline consumption by pregnant women is approximately 70% of the AI.
Collapse
Affiliation(s)
| | - Barbara J Strupp
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA.,Department of Psychology, Cornell University, Ithaca, New York, USA
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | - Richard L Canfield
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| |
Collapse
|
32
|
Taesuwan S, McDougall MQ, Malysheva OV, Bender E, Nevins JEH, Devapatla S, Vidavalur R, Caudill MA, Klatt KC. Choline metabolome response to prenatal choline supplementation across pregnancy: A randomized controlled trial. FASEB J 2021; 35:e22063. [PMID: 34820909 PMCID: PMC10911820 DOI: 10.1096/fj.202101401rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/04/2021] [Accepted: 11/09/2021] [Indexed: 12/31/2022]
Abstract
Pregnancy places a unique stress upon choline metabolism, requiring adaptations to support both maternal and fetal requirements. The impact of pregnancy and prenatal choline supplementation on choline and its metabolome in free-living, healthy adults is relatively uncharacterized. This study investigated the effect of prenatal choline supplementation on maternal and fetal biomarkers of choline metabolism among free-living pregnant persons consuming self-selected diets. Participants were randomized to supplemental choline (as choline chloride) intakes of 550 mg/d (500 mg/d d0-choline + 50 mg/d methyl-d9-choline; intervention) or 25 mg/d d9-choline (control) from gestational week (GW) 12-16 until Delivery. Fasting blood and 24-h urine samples were obtained at study Visit 1 (GW 12-16), Visit 2 (GW 20-24), and Visit 3 (GW 28-32). At Delivery, maternal and cord blood and placental tissue samples were collected. Participants randomized to 550 (vs. 25) mg supplemental choline/d achieved higher (p < .05) plasma concentrations of free choline, betaine, dimethylglycine, phosphatidylcholine (PC), and sphingomyelin at one or more study timepoint. Betaine was most responsive to prenatal choline supplementation with increases (p ≤ .001) in maternal plasma observed at Visit 2-Delivery (relative to Visit 1 and control), as well as in the placenta and cord plasma. Notably, greater plasma enrichments of d3-PC and LDL-C were observed in the intervention (vs. control) group, indicating enhanced PC synthesis through the de novo phosphatidylethanolamine N-methyltransferase pathway and lipid export. Overall, these data show that prenatal choline supplementation profoundly alters the choline metabolome, supporting pregnancy-related metabolic adaptations and revealing biomarkers for use in nutritional assessment and monitoring during pregnancy.
Collapse
Affiliation(s)
- Siraphat Taesuwan
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
- Cluster of Agro Bio-Circular-Green Industry, Faculty of Agro-Industry, Chiang Mai University, Chiang Mai, Thailand
| | | | - Olga V. Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | - Erica Bender
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | - Julie E. H. Nevins
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | | | | | - Marie A. Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | - Kevin C. Klatt
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
- Children’s Nutrition Research Center, Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
33
|
Powers BE, Velazquez R, Strawderman MS, Ginsberg SD, Mufson EJ, Strupp BJ. Maternal Choline Supplementation as a Potential Therapy for Down Syndrome: Assessment of Effects Throughout the Lifespan. Front Aging Neurosci 2021; 13:723046. [PMID: 34690739 PMCID: PMC8527982 DOI: 10.3389/fnagi.2021.723046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/20/2021] [Indexed: 12/23/2022] Open
Abstract
Maternal choline supplementation (MCS) has emerged as a promising therapy to lessen the cognitive and affective dysfunction associated with Down syndrome (DS). Choline is an essential nutrient, especially important during pregnancy due to its wide-ranging ontogenetic roles. Using the Ts65Dn mouse model of DS, our group has demonstrated that supplementing the maternal diet with additional choline (4-5 × standard levels) during pregnancy and lactation improves spatial cognition, attention, and emotion regulation in the adult offspring. The behavioral benefits were associated with a rescue of septohippocampal circuit atrophy. These results have been replicated across a series of independent studies, although the magnitude of the cognitive benefit has varied. We hypothesized that this was due, at least in part, to differences in the age of the subjects at the time of testing. Here, we present new data that compares the effects of MCS on the attentional function of adult Ts65Dn offspring, which began testing at two different ages (6 vs. 12 months of age). These data replicate and extend the results of our previous reports, showing a clear pattern indicating that MCS has beneficial effects in Ts65Dn offspring throughout life, but that the magnitude of the benefit (relative to non-supplemented offspring) diminishes with aging, possibly because of the onset of Alzheimer's disease-like neuropathology. In light of growing evidence that increased maternal choline intake during pregnancy is beneficial to the cognitive and affective functioning of all offspring (e.g., neurotypical and DS), the addition of this nutrient to a prenatal vitamin regimen would be predicted to have population-wide benefits and provide early intervention for fetuses with DS, notably including babies born to mothers unaware that they are carrying a fetus with DS.
Collapse
Affiliation(s)
- Brian E. Powers
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
- Edward Hines Jr. VA Hospital, Hines, IL, United States
| | - Ramon Velazquez
- Department of Psychology, Cornell University, Ithaca, NY, United States
- Arizona State University-Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, United States
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Myla S. Strawderman
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Stephen D. Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
- Department Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
- New York University Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
| | - Elliott J. Mufson
- Departments of Translational Neuroscience and Neurology, Barrow Neurological Institute, St. Joseph's Medical Center, Phoenix, AZ, United States
| | - Barbara J. Strupp
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
- Department of Psychology, Cornell University, Ithaca, NY, United States
| |
Collapse
|
34
|
Moltó-Puigmartí C, Obeid R, Mommers M, Eussen SJ, Thijs C. Maternal plasma choline and betaine in late pregnancy and child growth up to age 8 years in the KOALA Birth Cohort Study. Am J Clin Nutr 2021; 114:1438-1446. [PMID: 34113974 PMCID: PMC8488875 DOI: 10.1093/ajcn/nqab177] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/05/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Sufficient choline and betaine during pregnancy are needed for fetal growth and development. OBJECTIVES We aimed to investigate the associations between maternal plasma choline and betaine in the third trimester of pregnancy and child growth from birth up to 8 years of age. METHODS Concentrations of choline and betaine were measured in plasma of 1331 pregnant women from the KOALA (Kind, Ouders en gezondheid: Aandacht voor Leefstijl en Aanleg) Birth Cohort Study in the Netherlands. Child weight and height were measured at birth and at 1 (91% complete), 2 (86%), and 6-8 y (76%). Birth weight, weight gain in the first year, and z scores for weight and height at 1 and 2 y were used as continuous outcome variables. BMI z scores at 1 and 2 y were used as continuous and dichotomous outcomes, and BMI z scores at age 6-8 y were used to study overweight at that age. RESULTS Each 1-µmol/L increase of maternal plasma choline was associated with a mean 20-g (95% CI: 1.1, 38.0 g) higher weight gain in the first year of life, and a higher BMI z score (β: 0.02; 95% CI: 0.00, 0.04) and slightly higher odds of BMI z score >85th percentile (OR: 1.08; 95% CI: 1.03, 1.10) at 1-2 y. Each 1-µmol/L increase of plasma betaine was associated with a mean 12-g (95% CI: 0.8, 23.9 g) higher weight gain in the first year of life and higher odds of BMI z score >85th percentile at 1-2 y (OR: 1.03; 95% CI: 1.00, 1.07). Lastly, betaine was associated with overweight at 6-8 y (OR: 1.17; 95% CI: 1.02, 1.34), only in boys. CONCLUSIONS Third-trimester pregnancy plasma choline and betaine were positively associated with childhood anthropometric measures. In boys, some of the associations may have persisted up to 8 y of age. Further studies may investigate the validity of maternal plasma choline and betaine concentrations as markers of maternal intake and fetal transfer.
Collapse
Affiliation(s)
| | - Rima Obeid
- Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, Homburg, Germany
| | - Monique Mommers
- Department of Epidemiology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Simone Jpm Eussen
- Department of Epidemiology, Maastricht University Medical Centre, Maastricht, Netherlands,Care and Public Health Research Institute, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| | - Carel Thijs
- Department of Epidemiology, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
35
|
Hoffman MC, Freedman R, Law AJ, Clark AM, Hunter SK. Maternal nutrients and effects of gestational COVID-19 infection on fetal brain development. Clin Nutr ESPEN 2021; 43:1-8. [PMID: 34024500 PMCID: PMC8144544 DOI: 10.1016/j.clnesp.2021.04.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 04/20/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Maternal gestational infection is a well-characterized risk factor for offsprings' development of mental disorders including schizophrenia, autism, and attention deficit disorder. The inflammatory response elicited by the infection is partly directed against the placenta and fetus and is the putative pathogenic mechanism for fetal brain developmental abnormalities. Fetal brain abnormalities are generally irreversible after birth and increase risk for later mental disorders. Maternal immune activation in animals models this pathophysiology. SARS-CoV-2 produces maternal inflammatory responses during pregnancy similar to previously studied common respiratory viruses. METHOD Choline, folic acid, Vitamin D, and n-3 polyunsaturated fatty acids are among the nutrients that have been studied as possible mitigating factors for effects of maternal infection and inflammation on fetal development. Clinical and animal studies relevant to their use in pregnant women who have been infected are reviewed. RESULTS Higher maternal choline levels have positive effects on the development of brain function for infants of mothers who experienced viral infections in early pregnancy. No other nutrient has been studied in the context of viral inflammation. Vitamin D reduces pro-inflammatory cytokines in some, but not all, studies. Active folic acid metabolites decrease anti-inflammatory cytokines. N-3 polyunsaturated fatty acids have no effect. CONCLUSIONS Vitamin D and folic acid are already supplemented in food additives and in prenatal vitamins. Despite recommendations by several public health agencies and medical societies, choline intake is often inadequate in early gestation when the brain is forming. A public health initiative for choline supplements during the pandemic could be helpful for women planning or already pregnant who also become exposed or infected with SARS-CoV-2.
Collapse
Affiliation(s)
- M Camille Hoffman
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of Colorado Denver School of Medicine, Mail Stop F-546, Anschutz Medical Center, Aurora, CO, 80045, USA; Department of Psychiatry, University of Colorado Denver School of Medicine, Mail Stop F-546, Anschutz Medical Center, Aurora, CO, 80045, USA.
| | - Robert Freedman
- Department of Psychiatry, University of Colorado Denver School of Medicine, Mail Stop F-546, Anschutz Medical Center, Aurora, CO, 80045, USA.
| | - Amanda J Law
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of Colorado Denver School of Medicine, Mail Stop F-546, Anschutz Medical Center, Aurora, CO, 80045, USA; Department of Psychiatry, University of Colorado Denver School of Medicine, Mail Stop F-546, Anschutz Medical Center, Aurora, CO, 80045, USA; Department of Cell and Developmental Biology, University of Colorado Denver School of Medicine, Mail Stop F-546, Anschutz Medical Center, Aurora, CO, 80045, USA; Department of Medicine, University of Colorado Denver School of Medicine, Mail Stop F-546, Anschutz Medical Center, Aurora, CO, 80045, USA.
| | - Alena M Clark
- Department of Nutrition and Dietetics, Campus Box 93, University of Northern Colorado, Greeley, CO, 80639, USA.
| | - Sharon K Hunter
- Department of Psychiatry, University of Colorado Denver School of Medicine, Mail Stop F-546, Anschutz Medical Center, Aurora, CO, 80045, USA.
| |
Collapse
|
36
|
Jackson KH, Klatt KC, Caudill MA, McDougall MQ, West AA, Perry CA, Malysheva OV, Harris WS. Baseline red blood cell and breast milk DHA levels affect responses to standard dose of DHA in lactating women on a controlled feeding diet. Prostaglandins Leukot Essent Fatty Acids 2021; 166:102248. [PMID: 33516092 DOI: 10.1016/j.plefa.2021.102248] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND The importance of providing the newborn infant with docosahexaenoic acid (DHA) from breast milk is well established. However, women in the United States, on average, have breast milk DHA levels of 0.20%, which is below the worldwide average (and proposed target) of >0.32%. Additionally, the relationship between maternal red blood cell (RBC) and breast milk DHA levels may provide insight into the sufficiency of DHA recommendations during lactation. Whether the standard recommendation of at least 200 mg/day of supplemental DHA during lactation is sufficient for most women to achieve a desirable RBC and breast milk DHA status is unknown. METHODS Lactating women (n = 27) at about 5 weeks postpartum were enrolled in a 10-12 week controlled feeding study that included randomization to 480 or 930 mg choline/d (diet plus supplementation). As part of the intervention, all participants were required to consume a 200 mg/d of microalgal DHA. RBC and breast milk DHA levels were measured by capillary gas chromatography in an exploratory analysis. RESULTS Median RBC DHA was 5.0% (95% CI: 4.3, 5.5) at baseline and 5.1% (4.6, 5.4) after 10 weeks of supplementation (P = 0.6). DHA as a percent of breast milk fatty acids increased from 0.19% (0.18, 0.33) to 0.34% (0.27, 0.38) after supplementation (P<0.05). The proportion of women meeting the target RBC DHA level of >5% was unchanged (52% at baseline and week 10). The proportion of women achieving a breast milk DHA level of >0.32% approximately doubled from 30% to 56% (p = 0.06). Baseline RBC and breast milk DHA levels affected their responses to supplementation. Those with baseline RBC and breast milk DHA levels above the median (5% and 0.19%, respectively) experienced no change or a slight decrease in levels, while those below the median had a significant increase. Choline supplementation did not significantly influence final RBC or breast milk DHA levels. CONCLUSIONS On average, the standard prenatal DHA dose of 200 mg/d did not increase RBC DHA but did increase breastmilk DHA over 10 weeks in a cohort of lactating women in a controlled-feeding study. Baseline DHA levels in RBC and breast milk affected the response to DHA supplementation, with lower levels being associated with a greater increase and higher levels with no change or a slight decrease. Additional larger, dose-response DHA trials accounting for usual intakes and baseline DHA status are needed to determine how to best achieve target breast milk DHA levels and to identify additional modifiers of the variable breast milk DHA response to maternal DHA supplementation.
Collapse
Affiliation(s)
- Kristina Harris Jackson
- OmegaQuant Analytics, LLC. Sioux Falls, SD, 57105, USA; Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, 57105, USA.
| | - Kevin C Klatt
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Marie A Caudill
- Division of Nutritional Science, Cornell University, Ithaca, NY, 14853, USA
| | | | - Allyson A West
- Division of Nutritional Science, Cornell University, Ithaca, NY, 14853, USA
| | - Cydne A Perry
- Division of Nutritional Science, Cornell University, Ithaca, NY, 14853, USA
| | - Olga V Malysheva
- Division of Nutritional Science, Cornell University, Ithaca, NY, 14853, USA
| | - William S Harris
- Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, 57105, USA; Fatty Acid Research Institute, Sioux Falls, SD, 57105, USA
| |
Collapse
|
37
|
Hammoud R, Pannia E, Kubant R, Wasek B, Bottiglieri T, Malysheva OV, Caudill MA, Anderson GH. Choline and Folic Acid in Diets Consumed during Pregnancy Interact to Program Food Intake and Metabolic Regulation of Male Wistar Rat Offspring. J Nutr 2021; 151:857-865. [PMID: 33561219 PMCID: PMC8030718 DOI: 10.1093/jn/nxaa419] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/26/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND North American women consume high folic acid (FA), but most are not meeting the adequate intakes for choline. High-FA gestational diets induce an obesogenic phenotype in rat offspring. It is unclear if imbalances between FA and other methyl-nutrients (i.e., choline) account for these effects. OBJECTIVE This study investigated the interaction of choline and FA in gestational diets on food intake, body weight, one-carbon metabolism, and hypothalamic gene expression in male Wistar rat offspring. METHODS Pregnant Wistar rats were fed an AIN-93G diet with recommended choline and FA [RCRF; 1-fold, control] or high (5-fold) FA with choline at 0.5-fold [low choline and high folic acid (LCHF)], 1-fold [recommended choline and high folic acid (RCHF)], or 2.5-fold [high choline and high folic acid (HCHF)]. Male offspring were weaned to an RCRF diet for 20 wk. Food intake, weight gain, plasma energy-regulatory hormones, brain and plasma one-carbon metabolites, and RNA sequencing (RNA-seq) in pup hypothalamuses were assessed. RESULTS Adult offspring from LCHF and RCHF, but not HCHF, gestational diets had 10% higher food intake and weight gain than controls (P < 0.01). HCHF newborn pups had lower plasma insulin and leptin compared with LCHF and RCHF pups (P < 0.05), respectively. Pup brain choline (P < 0.05) and betaine (P < 0.01) were 22-33% higher in HCHF pups compared with LCHF pups; methionine was ∼23% lower after all high FA diets compared with RCRF (P < 0.01). LCHF adult offspring had lower brain choline (P < 0.05) than all groups and lower plasma 5-methyltetrahydrofolate (P < 0.05) than RCRF and RCHF groups. HCHF adult offspring had lower plasma cystathionine (P < 0.05) than LCHF adult offspring and lower homocysteine (P < 0.01) than RCHF and RCRF adult offspring. RNA-seq identified 144 differentially expressed genes in the hypothalamus of HCHF newborns compared with controls. CONCLUSIONS Increased choline in gestational diets modified the programming effects of high FA on long-term food intake regulation, plasma energy-regulatory hormones, one-carbon metabolism, and hypothalamic gene expression in male Wistar rat offspring, emphasizing a need for more attention to the choline and FA balance in maternal diets.
Collapse
Affiliation(s)
- Rola Hammoud
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Emanuela Pannia
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Ruslan Kubant
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Brandi Wasek
- Institute of Metabolic Disease, Baylor Scott & White Health, Austin, TX, USA
| | - Teodoro Bottiglieri
- Institute of Metabolic Disease, Baylor Scott & White Health, Austin, TX, USA
| | - Olga V Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | | |
Collapse
|
38
|
Zhu Y, Mordaunt CE, Durbin‐Johnson BP, Caudill MA, Malysheva OV, Miller JW, Green R, James SJ, Melnyk SB, Fallin MD, Hertz‐Picciotto I, Schmidt RJ, LaSalle JM. Expression Changes in Epigenetic Gene Pathways Associated With One-Carbon Nutritional Metabolites in Maternal Blood From Pregnancies Resulting in Autism and Non-Typical Neurodevelopment. Autism Res 2021; 14:11-28. [PMID: 33159718 PMCID: PMC7894157 DOI: 10.1002/aur.2428] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/16/2020] [Accepted: 10/21/2020] [Indexed: 12/15/2022]
Abstract
The prenatal period is a critical window for the development of autism spectrum disorder (ASD). The relationship between prenatal nutrients and gestational gene expression in mothers of children later diagnosed with ASD or non-typical development (Non-TD) is poorly understood. Maternal blood collected prospectively during pregnancy provides insights into the effects of nutrition, particularly one-carbon metabolites, on gene pathways and neurodevelopment. Genome-wide transcriptomes were measured with microarrays in 300 maternal blood samples in Markers of Autism Risk in Babies-Learning Early Signs. Sixteen different one-carbon metabolites, including folic acid, betaine, 5'-methyltretrahydrofolate (5-MeTHF), and dimethylglycine (DMG) were measured. Differential expression analysis and weighted gene correlation network analysis (WGCNA) were used to compare gene expression between children later diagnosed as typical development (TD), Non-TD and ASD, and to one-carbon metabolites. Using differential gene expression analysis, six transcripts (TGR-AS1, SQSTM1, HLA-C, and RFESD) were associated with child outcomes (ASD, Non-TD, and TD) with genome-wide significance. Genes nominally differentially expressed between ASD and TD significantly overlapped with seven high confidence ASD genes. WGCNA identified co-expressed gene modules significantly correlated with 5-MeTHF, folic acid, DMG, and betaine. A module enriched in DNA methylation functions showed a suggestive protective association with folic acid/5-MeTHF concentrations and ASD risk. Maternal plasma betaine and DMG concentrations were associated with a block of co-expressed genes enriched for adaptive immune, histone modification, and RNA processing functions. These results suggest that the prenatal maternal blood transcriptome is a sensitive indicator of gestational one-carbon metabolite status and changes relevant to children's later neurodevelopmental outcomes. LAY SUMMARY: Pregnancy is a time when maternal nutrition could interact with genetic risk for autism spectrum disorder. Blood samples collected during pregnancy from mothers who had a prior child with autism were examined for gene expression and nutrient metabolites, then compared to the diagnosis of the child at age three. Expression differences in gene pathways related to the immune system and gene regulation were observed for pregnancies of children with autism and non-typical neurodevelopment and were associated with maternal nutrients.
Collapse
Affiliation(s)
- Yihui Zhu
- Department of Medical Microbiology and Immunology, Genome Center, and Perinatal Origins of Disparities CenterUniversity of CaliforniaDavisCaliforniaUSA
- MIND Institute, School of MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Charles E. Mordaunt
- Department of Medical Microbiology and Immunology, Genome Center, and Perinatal Origins of Disparities CenterUniversity of CaliforniaDavisCaliforniaUSA
- MIND Institute, School of MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | | | - Marie A. Caudill
- Division of Nutritional SciencesCornell UniversityIthacaNew YorkUSA
| | | | - Joshua W. Miller
- Department of Nutritional SciencesRutgers UniversityNew BrunswickNew JerseyUSA
| | - Ralph Green
- Department of Pathology and Laboratory MedicineUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - S. Jill James
- Department of Pediatrics, University of Arkansas for Medical SciencesArkansas Children's Research InstituteLittle RockArkansasUSA
| | - Stepan B. Melnyk
- Department of Pediatrics, University of Arkansas for Medical SciencesArkansas Children's Research InstituteLittle RockArkansasUSA
| | - M. Daniele Fallin
- Department of Mental Health, Bloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Irva Hertz‐Picciotto
- MIND Institute, School of MedicineUniversity of CaliforniaDavisCaliforniaUSA
- Department of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Rebecca J. Schmidt
- MIND Institute, School of MedicineUniversity of CaliforniaDavisCaliforniaUSA
- Department of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Janine M. LaSalle
- Department of Medical Microbiology and Immunology, Genome Center, and Perinatal Origins of Disparities CenterUniversity of CaliforniaDavisCaliforniaUSA
- MIND Institute, School of MedicineUniversity of CaliforniaDavisCaliforniaUSA
| |
Collapse
|
39
|
Choline Content of Term and Preterm Infant Formulae Compared to Expressed Breast Milk-How Do We Justify the Discrepancies? Nutrients 2020; 12:nu12123815. [PMID: 33322176 PMCID: PMC7763895 DOI: 10.3390/nu12123815] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/10/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
Choline/phosphatidylcholine concentrations are tightly regulated in all organs and secretions. During rapid organ growth in the third trimester, choline requirement is particularly high. Adequate choline intake is 17–18 mg/kg/day in term infants, whereas ~50–60 mg/kg/day is required to achieve fetal plasma concentrations in preterm infants. Whereas free choline is supplied via the placenta, other choline carriers characterize enteral feeding. We therefore quantified the concentrations and types of choline carriers and choline-related components in various infant formulae and fortifiers compared to breast milk, and calculated the supply at full feeds (150 mL/kg/day) using tandem mass spectrometry. Choline concentration in formula ranged from values below to far above that of breastmilk. Humana 0-VLB (2015: 60.7 mg/150 mL; 2020: 27.3 mg/150 mL), Aptamil-Prematil (2020: 34.7 mg/150 mL), Aptamil-Prematil HA (2020: 37.6 mg/150 mL) for preterm infants with weights < 1800 g, and Humana 0 (2020: 41.6 mg/150 mL) for those > 1800 g, comprised the highest values in formulae studied. Formulae mostly were rich in free choline or phosphatidylcholine rather than glycerophosphocholine and phosphocholine (predominating in human milk). Most formulae (150 mL/kg/day) do not supply the amounts and physiologic components of choline required to achieve fetal plasma choline concentrations. A revision of choline content in formulae and breast milk fortifiers and a clear declaration of the choline components in formulae is required to enable informed choices.
Collapse
|
40
|
Ilozumba MN, Cheng TYD, Neuhouser ML, Miller JW, Beresford SAA, Duggan DJ, Toriola AT, Song X, Zheng Y, Bailey LB, Shadyab AH, Liu S, Malysheva O, Caudill MA, Ulrich CM. Associations between Plasma Choline Metabolites and Genetic Polymorphisms in One-Carbon Metabolism in Postmenopausal Women: The Women's Health Initiative Observational Study. J Nutr 2020; 150:2874-2881. [PMID: 32939549 PMCID: PMC7675024 DOI: 10.1093/jn/nxaa266] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/12/2020] [Accepted: 08/10/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Choline plays an integral role in one-carbon metabolism in the body, but it is unclear whether genetic polymorphisms are associated with variations in plasma choline and its metabolites. OBJECTIVES This study aimed to evaluate the association of genetic variants in choline and one-carbon metabolism with plasma choline and its metabolites. METHODS We analyzed data from 1423 postmenopausal women in a case-control study nested within the Women's Health Initiative Observational Study. Plasma concentrations of choline, betaine, dimethylglycine (DMG), and trimethylamine N-oxide were determined in 12-h fasting blood samples collected at baseline (1993-1998). Candidate and tagging single-nucleotide polymorphisms (SNPs) were genotyped in betaine-homocysteine S-methyltransferase (BHMT), BHMT2, 5,10-methylenetetrahydrofolate reductase (MTHFR), methylenetetrahydrofolate dehydrogenase (NADP+ dependent 1) (MTHFD1), 5-methyltetrahydrofolate-homocysteine methyltransferase (MTR), and 5-methyltetrahydrofolate-homocysteine methyltransferase reductase (MTRR). Linear regression was used to derive percentage difference in plasma concentrations per variant allele, adjusting for confounders, including B-vitamin biomarkers. Potential effect modification by plasma vitamin B-12, vitamin B-6, and folate concentrations and folic-acid fortification periods was examined. RESULTS The candidate SNP BHMT R239Q (rs3733890) was associated with lower concentrations of plasma betaine and DMG concentrations (-4.00% and -6.75% per variant allele, respectively; both nominal P < 0.05). Another candidate SNP, BHMT2 rs626105 A>G, was associated with higher plasma DMG concentration (13.0%; P < 0.0001). Several tagSNPs in these 2 genes were associated with plasma concentrations after correction for multiple comparisons. Vitamin B-12 status was a significant effect modifier of the association between the genetic variant BHMT2 rs626105 A>G and plasma DMG concentration. CONCLUSIONS Genetic variations in metabolic enzymes were associated with plasma concentrations of choline and its metabolites. Our findings contribute to the knowledge on the variation in blood nutrient concentrations in postmenopausal women.
Collapse
Affiliation(s)
| | - Ting-Yuan D Cheng
- Department of Epidemiology, University of Florida, Gainesville, FL, USA
| | - Marian L Neuhouser
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Joshua W Miller
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Shirley A A Beresford
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - David J Duggan
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Adetunji T Toriola
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoling Song
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Yingye Zheng
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lynn B Bailey
- Department of Foods and Nutrition, University of Georgia, Athens, GA, USA
| | - Aladdin H Shadyab
- Department of Family Medicine and Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Simin Liu
- Department of Epidemiology, Brown University, Providence, RI, USA
- Department of Medicine, Brown University, Providence, RI, USA
| | - Olga Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Cornelia M Ulrich
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
41
|
The Mechanisms of Improving IVF Outcomes of Liu-Wei-Di-Huang Pill Acting on DOR Patients. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:5183017. [PMID: 33178317 PMCID: PMC7648682 DOI: 10.1155/2020/5183017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 07/26/2020] [Accepted: 10/19/2020] [Indexed: 11/17/2022]
Abstract
Diminished ovarian reserve (DOR) is the weakening of ovarian oocyte production and quality. It will further become premature ovarian failure without timely cure. However, disease pathology and diagnostic markers are still incompletely understood. Liu-Wei-Di-Huang (LWDH) pill, a traditional Chinese medicine formula, is commonly used in the treatment of DOR in China. To explore the mechanism of the effect of LWDH on in vitro fertilization (IVF) outcomes in patients with DOR, a pseudotargeted metabolomics study combined with multivariate data processing strategy was carried out. A liquid chromatography tandem mass spectrometry-based metabolomics approach was applied to characterize metabolic biomarker candidates. Multiple pattern recognition was used to determine groups and confirm important variables. A total of 21 potential biomarkers were characterized, and related metabolic pathways were identified. The study displayed that the established pseudotargeted metabolomics strategy is a powerful approach for investigating the mechanism of DOR and LWDH. In addition, the approach may highlight biomarkers and metabolic pathways and can capture subtle metabolite changes from headache, which may lead to an improved mechanism understanding of DOR diseases and LWDH treatment.
Collapse
|
42
|
Abstract
Pregnancy is a time where expectant mothers often focus on their diet to improve their own health and to preserve the future health of their children. There is much conflicting information in the public domain about the safety and/or efficacy of nutritional supplements during pregnancy. Despite this, the market for supplements is growing. This review discusses the roles of critical nutrients in pregnancy and the available evidence on the use of supplements to reduce risks and improve maternal and fetal outcomes. Recommendations are made for pregnant women, taking into account safety data and tolerable upper intakes set for pregnant women. It is important for dieticians, nutritionists, physicians, and other healthcare providers to be able to offer accurate and evidence-based advice on supplement use in pregnancy. Routine supplementation may not be necessary for all, but individuals at risk are identified.
Collapse
|
43
|
Agam G, Taylor Z, Vainer E, Golan HM. The influence of choline treatment on behavioral and neurochemical autistic-like phenotype in Mthfr-deficient mice. Transl Psychiatry 2020; 10:316. [PMID: 32948746 PMCID: PMC7501861 DOI: 10.1038/s41398-020-01002-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 12/22/2022] Open
Abstract
Imbalanced one carbon metabolism and aberrant autophagy is robustly reported in patients with autism. Polymorphism in the gene methylenetetrahydrofolate reductase (Mthfr), encoding for a key enzyme in this pathway is associated with an increased risk for autistic-spectrum-disorders (ASDs). Autistic-like core and associated behaviors have been described, with contribution of both maternal and offspring Mthfr+/- genotype to the different domains of behavior. Preconception and prenatal supplementation with methyl donor rich diet to human subjects and mice reduced the risk for developing autism and autistic-like behavior, respectively. Here we tested the potential of choline supplementation to Mthfr-deficient mice at young-adulthood to reduce behavioral and neurochemical changes reminiscent of autism characteristics. We show that offspring of Mthfr+/- mothers, whether wildtype or heterozygote, exhibit autistic-like behavior, altered brain p62 protein levels and LC3-II/LC3-I levels ratio, both, autophagy markers. Choline supplementation to adult offspring of Mthfr+/- mothers for 14 days counteracted characteristics related to repetitive behavior and anxiety both in males and in females and improved social behavior solely in male mice. Choline treatment also normalized deviant cortical levels of the autophagy markers measured in male mice. The results demonstrate that choline supplementation even at adulthood, not tested previously, to offspring of Mthfr-deficient mothers, attenuates the autistic-like phenotype. If this proof of concept is replicated it might promote translation of these results to treatment recommendation for children with ASDs bearing similar genetic/metabolic make-up.
Collapse
Affiliation(s)
- Galila Agam
- grid.7489.20000 0004 1937 0511Faculty of Health Sciences, Department of Clinical Biochemistry and Pharmacology and Psychiatry Research Unit, Ben-Gurion University of the Negev and Mental Health Center, Beer-Sheva, Israel ,grid.7489.20000 0004 1937 0511Zlotowski Center for Neurosciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Zoe Taylor
- grid.7489.20000 0004 1937 0511Faculty of Health Sciences, Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ella Vainer
- grid.7489.20000 0004 1937 0511Faculty of Health Sciences, Department of Clinical Biochemistry and Pharmacology and Psychiatry Research Unit, Ben-Gurion University of the Negev and Mental Health Center, Beer-Sheva, Israel
| | - Hava M. Golan
- grid.7489.20000 0004 1937 0511Zlotowski Center for Neurosciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel ,grid.7489.20000 0004 1937 0511Faculty of Health Sciences, Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
44
|
Bozack AK, Howe CG, Hall MN, Liu X, Slavkovich V, Ilievski V, Lomax-Luu AM, Parvez F, Siddique AB, Shahriar H, Uddin MN, Islam T, Graziano JH, Gamble MV. Betaine and choline status modify the effects of folic acid and creatine supplementation on arsenic methylation in a randomized controlled trial of Bangladeshi adults. Eur J Nutr 2020; 60:1921-1934. [PMID: 32918135 DOI: 10.1007/s00394-020-02377-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/28/2020] [Indexed: 01/24/2023]
Abstract
PURPOSE Methylation of ingested inorganic arsenic (InAs) to monomethyl- (MMAs) and dimethyl-arsenical species (DMAs) facilitates urinary arsenic elimination. Folate and creatine supplementation influenced arsenic methylation in a randomized controlled trial. Here, we examine if baseline status of one-carbon metabolism nutrients (folate, choline, betaine, and vitamin B12) modified the effects of FA and creatine supplementation on changes in homocysteine, guanidinoacetate (GAA), total blood arsenic, and urinary arsenic metabolite proportions and indices. METHODS Study participants (N = 622) received 400 or 800 μg FA, 3 g creatine, 400 μg FA + 3 g creatine, or placebo daily for 12 weeks. RESULTS Relative to placebo, FA supplementation was associated with greater mean increases in %DMAs among participants with betaine concentrations below the median than those with levels above the median (FDR < 0.05). 400 μg FA/day was associated with a greater decrease in homocysteine among participants with plasma folate concentrations below, compared with those above, the median (FDR < 0.03). Creatine treatment was associated with a significant decrease in %MMAs among participants with choline concentrations below the median (P = 0.04), but not among participants above the median (P = 0.94); this effect did not significantly differ between strata (P = 0.10). CONCLUSIONS Effects of FA and creatine supplementation on arsenic methylation capacity were greater among individuals with low betaine and choline status, respectively. The efficacy of FA and creatine interventions to facilitate arsenic methylation may be modified by choline and betaine nutritional status. CLINICAL TRIAL REGISTRATION Clinical Trial Registry Identifier: NCT01050556, U.S. National Library of Medicine, https://clinicaltrials.gov ; registered January 15, 2010.
Collapse
Affiliation(s)
- Anne K Bozack
- Division of Pulmonary Medicine, Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th Street, Room 1107E, New York, NY, 10032, USA
| | - Caitlin G Howe
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th Street, Room 1107E, New York, NY, 10032, USA.,Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Megan N Hall
- Department of Epidemiology, Columbia University, New York, NY, USA.,Department of Environmental and Occupational Health Sciences, SUNY Downstate School of Public Health, Brooklyn, NY, USA.,Department of Epidemiology, SUNY Downstate School of Public Health, Brooklyn, NY, USA
| | - Xinhua Liu
- Department of Biostatistics, Columbia University, New York, NY, USA
| | - Vesna Slavkovich
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th Street, Room 1107E, New York, NY, 10032, USA
| | - Vesna Ilievski
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th Street, Room 1107E, New York, NY, 10032, USA
| | - Angela M Lomax-Luu
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th Street, Room 1107E, New York, NY, 10032, USA
| | - Faruque Parvez
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th Street, Room 1107E, New York, NY, 10032, USA
| | - Abu B Siddique
- Columbia University Arsenic Project in Bangladesh, Dhaka, Bangladesh
| | - Hasan Shahriar
- Columbia University Arsenic Project in Bangladesh, Dhaka, Bangladesh
| | - Mohammad N Uddin
- Columbia University Arsenic Project in Bangladesh, Dhaka, Bangladesh
| | - Tariqul Islam
- Columbia University Arsenic Project in Bangladesh, Dhaka, Bangladesh
| | - Joseph H Graziano
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th Street, Room 1107E, New York, NY, 10032, USA
| | - Mary V Gamble
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th Street, Room 1107E, New York, NY, 10032, USA.
| |
Collapse
|
45
|
Prenatal Amino Acid Supplementation to Improve Fetal Growth: A Systematic Review and Meta-Analysis. Nutrients 2020; 12:nu12092535. [PMID: 32825593 PMCID: PMC7551332 DOI: 10.3390/nu12092535] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/16/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022] Open
Abstract
Aberrant fetal growth remains a leading cause of perinatal morbidity and mortality and is associated with a risk of developing non-communicable diseases later in life. We performed a systematic review and meta-analysis combining human and animal studies to assess whether prenatal amino acid (AA) supplementation could be a promising approach to promote healthy fetal growth. PubMed, Embase, and Cochrane libraries were searched to identify studies orally supplementing the following AA groups during gestation: (1) arginine family, (2) branched chain (BCAA), and (3) methyl donors. The primary outcome was fetal/birth weight. Twenty-two human and 89 animal studies were included in the systematic review. The arginine family and, especially, arginine itself were studied the most. Our meta-analysis showed beneficial effects of arginine and (N-Carbamyl) glutamate (NCG) but not aspartic acid and citrulline on fetal/birth weight. However, no effects were reported when an isonitrogenous control diet was included. BCAA and methyl donor supplementation did not affect fetal/birth weight. Arginine family supplementation, in particular arginine and NCG, improves fetal growth in complicated pregnancies. BCAA and methyl donor supplementation do not seem to be as promising in targeting fetal growth. Well-controlled research in complicated pregnancies is needed before ruling out AA supplements or preferring arginine above other AAs.
Collapse
|
46
|
Bernhard W. Choline in cystic fibrosis: relations to pancreas insufficiency, enterohepatic cycle, PEMT and intestinal microbiota. Eur J Nutr 2020; 60:1737-1759. [PMID: 32797252 DOI: 10.1007/s00394-020-02358-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Cystic Fibrosis (CF) is an autosomal recessive disorder with life-threatening organ manifestations. 87% of CF patients develop exocrine pancreas insufficiency, frequently starting in utero and requiring lifelong pancreatic enzyme substitution. 99% develop progressive lung disease, and 20-60% CF-related liver disease, from mild steatosis to cirrhosis. Characteristically, pancreas, liver and lung are linked by choline metabolism, a critical nutrient in CF. Choline is a tightly regulated tissue component in the form of phosphatidylcholine (Ptd'Cho) and sphingomyelin (SPH) in all membranes and many secretions, particularly of liver (bile, lipoproteins) and lung (surfactant, lipoproteins). Via its downstream metabolites, betaine, dimethylglycine and sarcosine, choline is the major one-carbon donor for methionine regeneration from homocysteine. Methionine is primarily used for essential methylation processes via S-adenosyl-methionine. CLINICAL IMPACT CF patients with exocrine pancreas insufficiency frequently develop choline deficiency, due to loss of bile Ptd'Cho via feces. ~ 50% (11-12 g) of hepatic Ptd'Cho is daily secreted into the duodenum. Its re-uptake requires cleavage to lyso-Ptd'Cho by pancreatic and small intestinal phospholipases requiring alkaline environment. Impaired CFTR-dependent bicarbonate secretion, however, results in low duodenal pH, impaired phospholipase activity, fecal Ptd'Cho loss and choline deficiency. Low plasma choline causes decreased availability for parenchymal Ptd'Cho metabolism, impacting on organ functions. Choline deficiency results in hepatic choline/Ptd'Cho accretion from lung tissue via high density lipoproteins, explaining the link between choline deficiency and lung function. Hepatic Ptd'Cho synthesis from phosphatidylethanolamine by phosphatidylethanolamine-N-methyltransferase (PEMT) partly compensates for choline deficiency, but frequent single nucleotide polymorphisms enhance choline requirement. Additionally, small intestinal bacterial overgrowth (SIBO) frequently causes intraluminal choline degradation in CF patients prior to its absorption. As adequate choline supplementation was clinically effective and adult as well as pediatric CF patients suffer from choline deficiency, choline supplementation in CF patients of all ages should be evaluated.
Collapse
Affiliation(s)
- Wolfgang Bernhard
- Department of Neonatology, University Children's Hospital, Faculty of Medicine, Eberhard-Karls-University, Calwer Straße 7, 72076, Tübingen, Germany.
| |
Collapse
|
47
|
Goss KCW, Goss VM, Townsend JP, Koster G, Clark HW, Postle AD. Postnatal adaptations of phosphatidylcholine metabolism in extremely preterm infants: implications for choline and PUFA metabolism. Am J Clin Nutr 2020; 112:1438-1447. [PMID: 32778895 PMCID: PMC7727469 DOI: 10.1093/ajcn/nqaa207] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/01/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Lipid metabolism in pregnancy delivers PUFAs from maternal liver to the developing fetus. The transition at birth to diets less enriched in PUFA is especially challenging for immature, extremely preterm infants who are typically supported by total parenteral nutrition. OBJECTIVE The aim was to characterize phosphatidylcholine (PC) and choline metabolism in preterm infants and demonstrate the molecular specificity of PC synthesis by the immature preterm liver in vivo. METHODS This MS-based lipidomic study quantified the postnatal adaptations to plasma PC molecular composition in 31 preterm infants <28 weeks' gestational age. Activities of the cytidine diphosphocholine (CDP-choline) and phosphatidylethanolamine-N-methyltransferase (PEMT) pathways for PC synthesis were assessed from incorporations of deuterated methyl-D9-choline chloride. RESULTS The concentration of plasma PC in these infants increased postnatally from median values of 481 (IQR: 387-798) µM at enrollment to 1046 (IQR: 616-1220) µM 5 d later (P < 0.001). Direct incorporation of methyl-D9-choline demonstrated that this transition was driven by an active CDP-choline pathway that synthesized PC enriched in species containing oleic and linoleic acids. A second infusion of methyl-D9-choline chloride at day 5 clearly indicated continued activity of this pathway. Oxidation of D9-choline through D9-betaine resulted in the transfer of 1 deuterated methyl group to S-adenosylmethionine. A very low subsequent transfer of this labeled methyl group to D3-PC indicated that liver PEMT activity was essentially inactive in these infants. CONCLUSIONS This study demonstrated that the preterm infant liver soon after birth, and by extension the fetal liver, was metabolically active in lipoprotein metabolism. The low PEMT activity, which is the only pathway for endogenous choline synthesis and is responsible for hormonally regulated export of PUFAs from adult liver, strongly supports increased supplementation of preterm parenteral nutrition with both choline and PUFAs.
Collapse
Affiliation(s)
- Kevin C W Goss
- Child Health, Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,NIHR Southampton Respiratory Biomedical Research Unit, University Hospitals Southampton, Southampton, United Kingdom
| | - Victoria M Goss
- NIHR Southampton Respiratory Biomedical Research Unit, University Hospitals Southampton, Southampton, United Kingdom
| | - J Paul Townsend
- NIHR Southampton Respiratory Biomedical Research Unit, University Hospitals Southampton, Southampton, United Kingdom
| | - Grielof Koster
- NIHR Southampton Respiratory Biomedical Research Unit, University Hospitals Southampton, Southampton, United Kingdom
| | - Howard W Clark
- Present address for HWC: UCL EGA Institute for Women's Health, Faculty of Population Health Sciences, University College London, London, UK
| | | |
Collapse
|
48
|
Cho CE, Aardema NDJ, Bunnell ML, Larson DP, Aguilar SS, Bergeson JR, Malysheva OV, Caudill MA, Lefevre M. Effect of Choline Forms and Gut Microbiota Composition on Trimethylamine- N-Oxide Response in Healthy Men. Nutrients 2020; 12:nu12082220. [PMID: 32722424 PMCID: PMC7468900 DOI: 10.3390/nu12082220] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Trimethylamine-N-oxide (TMAO), a choline-derived gut microbiota-dependent metabolite, is a newly recognized risk marker for cardiovascular disease. We sought to determine: (1) TMAO response to meals containing free versus lipid-soluble choline and (2) effects of gut microbiome on TMAO response. Methods: In a randomized, controlled, double-blinded, crossover study, healthy men (n = 37) were provided meals containing 600 mg choline either as choline bitartrate or phosphatidylcholine, or no choline control. Results: Choline bitartrate yielded three-times greater plasma TMAO AUC (p = 0.01) and 2.5-times greater urinary TMAO change from baseline (p = 0.01) compared to no choline and phosphatidylcholine. Gut microbiota composition differed (permutational multivariate analysis of variance, PERMANOVA; p = 0.01) between high-TMAO producers (with ≥40% increase in urinary TMAO response to choline bitartrate) and low-TMAO producers (with <40% increase in TMAO response). High-TMAO producers had more abundant lineages of Clostridium from Ruminococcaceae and Lachnospiraceae compared to low-TMAO producers (analysis of composition of microbiomes, ANCOM; p < 0.05). Conclusion: Given that phosphatidylcholine is the major form of choline in food, the absence of TMAO elevation with phosphatidylcholine counters arguments that phosphatidylcholine should be avoided due to TMAO-producing characteristics. Further, development of individualized dietary recommendations based on the gut microbiome may be effective in reducing disease risk
Collapse
Affiliation(s)
- Clara E. Cho
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT 84322, USA; (N.D.J.A.); (M.L.B.); (D.P.L.); (S.S.A.); (J.R.B.); (M.L.)
- Correspondence: ; Tel.: +1-435-797-5369
| | - Niklas D. J. Aardema
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT 84322, USA; (N.D.J.A.); (M.L.B.); (D.P.L.); (S.S.A.); (J.R.B.); (M.L.)
| | - Madison L. Bunnell
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT 84322, USA; (N.D.J.A.); (M.L.B.); (D.P.L.); (S.S.A.); (J.R.B.); (M.L.)
| | - Deanna P. Larson
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT 84322, USA; (N.D.J.A.); (M.L.B.); (D.P.L.); (S.S.A.); (J.R.B.); (M.L.)
| | - Sheryl S. Aguilar
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT 84322, USA; (N.D.J.A.); (M.L.B.); (D.P.L.); (S.S.A.); (J.R.B.); (M.L.)
| | - Janet R. Bergeson
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT 84322, USA; (N.D.J.A.); (M.L.B.); (D.P.L.); (S.S.A.); (J.R.B.); (M.L.)
| | - Olga V. Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; (O.V.M.); (M.A.C.)
| | - Marie A. Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; (O.V.M.); (M.A.C.)
| | - Michael Lefevre
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT 84322, USA; (N.D.J.A.); (M.L.B.); (D.P.L.); (S.S.A.); (J.R.B.); (M.L.)
| |
Collapse
|
49
|
Cosín-Tomás M, Luan Y, Leclerc D, Malysheva OV, Lauzon N, Bahous RH, Christensen KE, Caudill MA, Rozen R. Moderate Folic Acid Supplementation in Pregnant Mice Results in Behavioral Alterations in Offspring with Sex-Specific Changes in Methyl Metabolism. Nutrients 2020; 12:nu12061716. [PMID: 32521649 PMCID: PMC7352339 DOI: 10.3390/nu12061716] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/27/2020] [Accepted: 06/04/2020] [Indexed: 12/24/2022] Open
Abstract
Fifteen to 20% of pregnant women may exceed the recommended intake of folic acid (FA) by more than four-fold. This excess could compromise neurocognitive and motor development in offspring. Here, we explored the impact of an FA-supplemented diet (5× FASD, containing five-fold higher FA than recommended) during pregnancy on brain function in murine offspring, and elucidated mechanistic changes. We placed female C57BL/6 mice for one month on control diets or 5× FASD before mating. Diets were maintained throughout pregnancy and lactation. Behavioural tests were conducted on 3-week-old pups. Pups and mothers were sacrificed at weaning. Brains and livers were collected to examine choline/methyl metabolites and immunoreactive methylenetetrahydrofolate reductase (MTHFR). 5× FASD led to hyperactivity-like behavior and memory impairment in 3-week-old pups of both sexes. Reduced MTHFR protein in the livers of FASD mothers and male pups resulted in choline/methyl metabolite disruptions in offspring liver (decreased betaine) and brain (decreased glycerophosphocholine and sphingomyelin in male pups, and decreased phosphatidylcholine in both sexes). These results indicate that moderate folate supplementation downregulates MTHFR and alters choline/methyl metabolism, contributing to neurobehavioral alterations. Our findings support the negative impact of high FA on brain development, and may lead to improved guidelines on optimal folate levels during pregnancy.
Collapse
Affiliation(s)
- Marta Cosín-Tomás
- Departments of Human Genetics and Pediatrics, Research Institute of the McGill University Health Center, McGill University, Montreal, QC H4A 3J1, Canada; (M.C.-T.); (Y.L.); (D.L.); (R.H.B.); (K.E.C.)
| | - Yan Luan
- Departments of Human Genetics and Pediatrics, Research Institute of the McGill University Health Center, McGill University, Montreal, QC H4A 3J1, Canada; (M.C.-T.); (Y.L.); (D.L.); (R.H.B.); (K.E.C.)
| | - Daniel Leclerc
- Departments of Human Genetics and Pediatrics, Research Institute of the McGill University Health Center, McGill University, Montreal, QC H4A 3J1, Canada; (M.C.-T.); (Y.L.); (D.L.); (R.H.B.); (K.E.C.)
| | - Olga V. Malysheva
- Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY 14853, USA; (O.V.M.); (M.A.C.)
| | - Nidia Lauzon
- Drug Discovery Platform, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada;
| | - Renata H. Bahous
- Departments of Human Genetics and Pediatrics, Research Institute of the McGill University Health Center, McGill University, Montreal, QC H4A 3J1, Canada; (M.C.-T.); (Y.L.); (D.L.); (R.H.B.); (K.E.C.)
| | - Karen E. Christensen
- Departments of Human Genetics and Pediatrics, Research Institute of the McGill University Health Center, McGill University, Montreal, QC H4A 3J1, Canada; (M.C.-T.); (Y.L.); (D.L.); (R.H.B.); (K.E.C.)
| | - Marie A. Caudill
- Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY 14853, USA; (O.V.M.); (M.A.C.)
| | - Rima Rozen
- Departments of Human Genetics and Pediatrics, Research Institute of the McGill University Health Center, McGill University, Montreal, QC H4A 3J1, Canada; (M.C.-T.); (Y.L.); (D.L.); (R.H.B.); (K.E.C.)
- Correspondence: ; Tel.: +15-14934-1934 (ext. 23839)
| |
Collapse
|
50
|
Huang KT, Shen YL, Lee CN, Chu KY, Ku WC, Liu CY, Huang RFS. Using Differential Threshold Effects of Individual and Combined Periconceptional Methyl Donor Status on Maternal Genomic LINE-1 and Imprinted H19 DNA Methylation to Predict Birth Weight Variance in the Taiwan Pregnancy-Newborn Epigenetics (TPNE) Cohort Study. J Nutr 2020; 150:108-117. [PMID: 31504733 DOI: 10.1093/jn/nxz204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Few studies have comprehensively examined the effect of methyl donor status on maternal DNA methylation and birth outcomes. OBJECTIVES This study examined associations between periconceptional methyl donor status and genome-wide and specific imprinted gene methylation and fetal growth indices in the Taiwan Pregnancy-Newborn Epigenetics cohort. METHODS Plasma folate, choline (free form), and betaine concentrations of the participants enrolled at 7-10 weeks of gestation were analyzed. DNA methylation at regulatory sequences of the imprinted H19 gene and genomic long interspersed nuclear element 1 (LINE-1) were measured in maternal lymphocytes using bisulfite/high-resolution melt polymerase chain reaction. Associations with birth weight (BW) were estimated through multiple regressions from 112 mother-newborn pairs. RESULTS A nonlinear "L-shaped" relation and an inverse association between maternal plasma folate in T1 (mean ± SE: 17.6 ± 5.1 nmol/L) and lymphocytic LINE-1 methylation (β: -0.49, P = 0.027) were characterized. After adjusting for LINE-1 methylation, individual maternal folate concentrations were positively associated with BW variance (β = 0.24, P = 0.035), and the association was more pronounced in mothers with choline in T1 (mean ± SE: 5.4 ± 0.6 μmol/L; β: 0.40, P = 0.039). Choline status of the mothers in T2 (mean ± SE: 7.2 ± 0.6 μmol/L) was inversely associated with LINE-1 methylation (β: -0.43, P = 0.035), and a positive association was evident between T1 choline and H19 methylation (β: 0.48, P = 0.011). After adjusting for epigenetic modification, maternal choline status predicted a positive association with BW (β: 0.56, P = 0.005), but the effect was limited to mothers with high betaine concentrations in T3 (mean ± SE: 36.4 ± 8.8 μmol/L), depending on folate status. CONCLUSIONS Our data highlight the differential threshold effects of periconceptional folate, choline, and betaine status on genomic LINE-1 and H19 DNA methylation and how their interplay has a long-term effect on BW variance.
Collapse
Affiliation(s)
- Kuang-Ta Huang
- PhD Program in Nutrition and Food Science, Fu Jen Catholic University, New Taipei City, Taiwan.,Loving Care Maternity and Children's Health Centers, New Taipei City, Taiwan
| | - Yu-Li Shen
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chien-Nan Lee
- Department of Gynecology and Obstetrics, National Taiwan University Hospital, Taipei City, Taiwan
| | - Kuan-Yu Chu
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Wei-Chi Ku
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chieh-Yu Liu
- Biostatistical Consultant Lab, Department of Speech Language Pathology and Audiology, National Taipei University of Nursing and Health Sciences, Taipei City, Taiwan
| | - Rwei-Fen S Huang
- PhD Program in Nutrition and Food Science, Fu Jen Catholic University, New Taipei City, Taiwan.,Department of Nutritional Science, Fu Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|