1
|
Shelp GV, Dong J, Orlov NO, Malysheva OV, Bender E, Shoveller AK, Bakovic M, Cho CE. Exposure to prenatal excess or imbalanced micronutrients leads to long-term perturbations in one-carbon metabolism, trimethylamine-N-oxide and DNA methylation in Wistar rat offspring. FASEB J 2024; 38:e70032. [PMID: 39212230 DOI: 10.1096/fj.202401018rr] [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: 05/10/2024] [Revised: 08/05/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Prenatal multivitamins, including folic acid, are commonly consumed in excess, whereas choline, an essential nutrient and an important source of labile methyl groups, is underconsumed. Here, we characterized profiles of one-carbon metabolism and related pathways and patterns of DNA methylation in offspring exposed to excess or imbalanced micronutrients prenatally. Pregnant Wistar rats were fed either recommended 1× vitamins (RV), high 10× vitamins (HV), high 10× folic acid with recommended choline (HFolRC), or high 10× folic acid with no choline (HFolNC). Offspring were weaned to a high-fat diet for 12 weeks. Circulating metabolites were analyzed with a focus on the hypothalamus, an area known to be under epigenetic regulation. HV, HFolRC, and HFolNC males had higher body weight (BW) and lower plasma choline and methionine consistent with lower hypothalamic S-adenosylmethionine (SAM):S-adenosylhomocysteine (SAH) and global DNA methylation compared with RV. HV and HFolNC females had higher BW and lower plasma 5-methyltetrahydrofolate and methionine consistent with lower hypothalamic global DNA methylation compared with RV. Plasma dimethylglycine (DMG) and methionine were higher as with hypothalamic SAM:SAH and global DNA methylation in HFolRC females without changes in BW compared with RV. Plasma trimethylamine and trimethylamine-N-oxide were higher in males but lower in females from HFolRC compared with RV. Network modeling revealed a link between the folate-dependent pathway and SAH, with most connections through DMG. Final BW was negatively correlated with choline, DMG, and global DNA methylation. In conclusion, prenatal intake of excess or imbalanced micronutrients induces distinct metabolic and epigenetic perturbations in offspring that reflect long-term nutritional programming of health.
Collapse
Affiliation(s)
- Gia V Shelp
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Jianzhang Dong
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Nikolai O Orlov
- Department of Chemistry, University of Guelph, Guelph, Ontario, Canada
| | - Olga V Malysheva
- Division of Nutritional Sciences, Human Metabolic Research Unit, Cornell University, Ithaca, New York, USA
| | - Erica Bender
- Division of Nutritional Sciences, Human Metabolic Research Unit, Cornell University, Ithaca, New York, USA
| | - Anna K Shoveller
- Department of Animal Biosciences, University of Guelph, Guelph, Ontario, Canada
| | - Marica Bakovic
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Clara E Cho
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
2
|
Wiedeman AM, Miliku K, Moraes TJ, Mandhane PJ, Simons E, Subbarao P, Turvey SE, Zwicker JG, Devlin AM. Women in Canada are consuming above the upper intake level of folic acid but few are meeting dietary choline recommendations in the second trimester of pregnancy: data from the CHILD cohort study. Appl Physiol Nutr Metab 2024; 49:868-873. [PMID: 38320263 DOI: 10.1139/apnm-2023-0258] [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] [Indexed: 02/08/2024]
Abstract
There is concern that during a low-risk pregnancy, women are consuming more than recommended (400 µg/day) supplemental folic acid and may not meet recommendations for other nutrients. The objective of this study was to determine folic acid supplement use and dietary folate intakes in the second trimester (week 18) of pregnancy in women (n = 2996) in the Canadian CHILD cohort study. Vitamin B12 and choline intakes were also assessed because they are metabolically related to folate. The majority of participants (71.6%) were consuming a daily prenatal supplement. Twenty-eight percent of women (n = 847) reported consuming a folic acid supplement and of these women, 45.3% had daily supplemental folic acid intakes above the upper intake level (UL; 1000 µg/day). Daily dietary folate intakes were (mean (SD)) 575 (235) DFE µg/day. In contrast, only 24.8% of women met the dietary choline adequate intake (AI) recommendation (AI ≥ 450 mg/day) with a mean (SD) intake of 375 (151) mg/day. Further understanding of the impact of supplemental folic acid intake above the UL and low choline intake during pregnancy 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, BC V5Z 4H4, Canada
| | - Kozeta Miliku
- Department of Nutritional Sciences, University of Toronto, Toronto, ON M5T 3M7, Canada
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Theo J Moraes
- Department of Pediatrics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | | | - Elinor Simons
- Section of Allergy and Immunology, University of Manitoba, Winnipeg, MB R3E 3P5, Canada
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3A 1S1, Canada
| | - Padmaja Subbarao
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Pediatrics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Stuart E Turvey
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Jill G Zwicker
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Occupational Science & Occupational Therapy, The University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Angela M Devlin
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| |
Collapse
|
3
|
Alvarenga L, Kemp JA, Baptista BG, Ribeiro M, Lima LS, Mafra D. Production of Toxins by the Gut Microbiota: The Role of Dietary Protein. Curr Nutr Rep 2024; 13:340-350. [PMID: 38587573 DOI: 10.1007/s13668-024-00535-x] [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] [Accepted: 03/26/2024] [Indexed: 04/09/2024]
Abstract
PURPOSE OF REVIEW This narrative review will discuss how the intake of specific protein sources (animal and vegetable) providing specific amino acids can modulate the gut microbiota composition and generate toxins. A better understanding of these interactions could lead to more appropriate dietary recommendations to improve gut health and mitigate the risk of complications promoted by the toxic metabolites formed by the gut microbiota. RECENT FINDINGS Gut microbiota is vital in maintaining human health by influencing immune function and key metabolic pathways. Under unfavorable conditions, the gut microbiota can produce excess toxins, which contribute to inflammation and the breakdown of the integrity of the intestinal barrier. Genetic and environmental factors influence gut microbiota diversity, with diet playing a crucial role. Emerging evidence indicates that the gut microbiota significantly metabolizes amino acids from dietary proteins, producing various metabolites with beneficial and harmful effects. Amino acids such as choline, betaine, l-carnitine, tyrosine, phenylalanine, and tryptophan can increase the production of uremic toxins when metabolized by intestinal bacteria. The type of food source that provides these amino acids affects the production of toxins. Plant-based diets and dietary fiber are associated with lower toxin formation than animal-based diets due to the high amino acid precursors in animal proteins.
Collapse
Affiliation(s)
- Livia Alvarenga
- Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro (RJ), Brazil.
- Graduate Program in Nutrition Science, Federal Fluminense University, Niteroi, Rio de Janeiro (RJ), Brazil.
| | - Julie A Kemp
- Graduate Program in Nutrition Science, Federal Fluminense University, Niteroi, Rio de Janeiro (RJ), Brazil
| | - Beatriz G Baptista
- Graduate Program in Medical Science, Federal Fluminense University, Niteroi, Rio de Janeiro (RJ), Brazil
| | - Marcia Ribeiro
- Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro (RJ), Brazil
| | - Ligia Soares Lima
- Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro (RJ), Brazil
| | - Denise Mafra
- Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro (RJ), Brazil
- Graduate Program in Nutrition Science, Federal Fluminense University, Niteroi, Rio de Janeiro (RJ), Brazil
- Graduate Program in Medical Science, Federal Fluminense University, Niteroi, Rio de Janeiro (RJ), Brazil
| |
Collapse
|
4
|
Rusnak T, Azarcoya-Barrera J, Makarowski A, Jacobs RL, Richard C. Plant- and Animal-Derived Dietary Sources of Phosphatidylcholine Have Differential Effects on Immune Function in The Context of A High-Fat Diet in Male Wistar Rats. J Nutr 2024; 154:1936-1944. [PMID: 38582387 PMCID: PMC11217025 DOI: 10.1016/j.tjnut.2024.04.004] [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: 10/26/2023] [Revised: 03/20/2024] [Accepted: 04/02/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Phosphatidylcholine (PC) derived from eggs has been shown to beneficially modulate T cell response and intestinal permeability under the context of a high-fat diet. OBJECTIVES The objective of this study was to determine whether there is a differential effect of plant and animal-derived sources of PC on immune function. METHODS Four-week-old male Wistar rats were randomly assigned to consume 1 of 4 diets (n = 10/group) for 12 wk, all containing 1.5 g of total choline/kg of diet but differing in choline forms: 1-Control Low-Fat [CLF, 20% fat, 100% free choline (FC)]; 2-Control High-Fat (CHF, 50% fat, 100% FC); 3-High-Fat Egg-derived PC (EPC, 50% fat, 100% Egg-PC); 4-High-Fat Soy-derived PC (SPC, 50% fat, 100% Soy-PC). Immune cell functions and phenotypes were measured in splenocytes by ex vivo cytokine production after mitogen stimulation and flow cytometry, respectively. RESULTS The SPC diet increased splenocyte IL-2 production after PMA+I stimulation compared with the CHF diet. However, the SPC group had a lower proportion of splenocytes expressing the IL-2 receptor (CD25+, P < 0.05). After PMA+I stimulation, feeding EPC normalized splenocyte production of IL-10 relative to the CLF diet, whereas SPC did not (P < 0.05). In mesenteric lymph node lymphocytes, the SPC diet group produced more IL-2 and TNF-α after PMA+I stimulation than the CHF diet, whereas the EPC diet group did not. CONCLUSIONS Our results suggest that both egg- and soy-derived PC may attenuate high-fat diet-induced T cell dysfunction. However, egg-PC enhances, to a greater extent, IL-10, a cytokine involved in promoting the resolution phase of inflammation, whereas soy-PC appears to elicit a greater effect on gut-associated immune responses.
Collapse
Affiliation(s)
- Tianna Rusnak
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Jessy Azarcoya-Barrera
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Alexander Makarowski
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - René L Jacobs
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Caroline Richard
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
5
|
Stråvik M, Hartvigsson O, Noerman S, Sandin A, Wold AE, Barman M, Sandberg AS. Biomarker Candidates of Habitual Food Intake in a Swedish Cohort of Pregnant and Lactating Women and Their Infants. Metabolites 2024; 14:256. [PMID: 38786733 PMCID: PMC11123206 DOI: 10.3390/metabo14050256] [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: 02/29/2024] [Revised: 04/19/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
Circulating food metabolites could improve dietary assessments by complementing traditional methods. Here, biomarker candidates of food intake were identified in plasma samples from pregnancy (gestational week 29, N = 579), delivery (mothers, N = 532; infants, N = 348), and four months postpartum (mothers, N = 477; breastfed infants, N = 193) and associated to food intake assessed with semi-quantitative food frequency questionnaires. Families from the Swedish birth cohort Nutritional impact on Immunological maturation during Childhood in relation to the Environment (NICE) were included. Samples were analyzed using untargeted liquid chromatography-mass spectrometry (LC-MS)-based metabolomics. Both exposure and outcome were standardized, and relationships were investigated using a linear regression analysis. The intake of fruits and berries and fruit juice were both positively related to proline betaine levels during pregnancy (fruits and berries, β = 0.23, FDR < 0.001; fruit juice, β = 0.27, FDR < 0.001), at delivery (fruit juice, infants: β = 0.19, FDR = 0.028), and postpartum (fruits and berries, mothers: β = 0.27, FDR < 0.001, infants: β = 0.29, FDR < 0.001; fruit juice, mothers: β = 0.37, FDR < 0.001). Lutein levels were positively related to vegetable intake during pregnancy (β = 0.23, FDR < 0.001) and delivery (mothers: β = 0.24, FDR < 0.001; newborns: β = 0.18, FDR = 0.014) and CMPF with fatty fish intake postpartum (mothers: β = 0.20, FDR < 0.001). No clear relationships were observed with the expected food sources of the remaining metabolites (acetylcarnitine, choline, indole-3-lactic acid, pipecolic acid). Our study suggests that plasma lutein could be useful as a more general food group intake biomarker for vegetables and fruits during pregnancy and delivery. Also, our results suggest the application of proline betaine as an intake biomarker of citrus fruit during gestation and lactation.
Collapse
Affiliation(s)
- Mia Stråvik
- Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden (A.-S.S.)
| | - Olle Hartvigsson
- Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden (A.-S.S.)
| | - Stefania Noerman
- Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden (A.-S.S.)
| | - Anna Sandin
- Pediatrics, Department of Clinical Science, Sunderby Research Unit, Umeå University, 901 87 Umeå, Sweden
| | - Agnes E. Wold
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Malin Barman
- Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden (A.-S.S.)
| | - Ann-Sofie Sandberg
- Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden (A.-S.S.)
| |
Collapse
|
6
|
Nguyen HT, Oktayani PPI, Lee SD, Huang LC. Choline in pregnant women: a systematic review and meta-analysis. Nutr Rev 2024:nuae026. [PMID: 38607338 DOI: 10.1093/nutrit/nuae026] [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] [Indexed: 04/13/2024] Open
Abstract
CONTEXT Choline is a critical nutrient. Inadequate choline intake during pregnancy increases the risk of adverse maternal and offspring health. OBJECTIVE A systematic review and meta-analysis were conducted to examine the current recommendations for choline intake by pregnant women, estimate the overall prevalence of pregnant women with adequate choline intake, and explore associations between maternal choline level and adverse pregnancy outcomes (APOs). METHODS Choline recommendations for pregnant women were assessed from eight nutrient guidelines of the United States, United Kingdom, Canada, Australia, Asia, International Federation of Gynecology and Obstetrics, and World Health Organization. Data on the prevalence of pregnant women with adequate choline intake and the association between maternal choline level and APOs were collected from 5 databases up to May 2023. Meta-analyses with random effects and subgroup analyses were performed for the pooled estimate of prevalence and association. RESULTS Five recent nutrition guidelines from the United States (United States Department of Agriculture), United States (Food and Drug Administration), Canada, Australia, and the International Federation of Gynecology and Obstetrics have emphasized the importance of adequate choline intake for pregnant women. Of 27 publications, 19 articles explored the prevalence and 8 articles explored the association. Meta-analysis of 12 prevalence studies revealed a concerning 11.24% (95% confidence interval, 6.34-17.26) prevalence of pregnant women with adequate choline intake recommendations. A meta-analysis of 6 studies indicated a significant association between high maternal choline levels and a reduced risk of developing APOs, with an odds ratio of 0.51 (95% confidence interval, 0.40-0.65). CONCLUSION The existing guidelines highlight the importance of choline in supporting maternal health and fetal development during pregnancy. Furthermore, a high maternal choline level was likely to be associated with a lower risk of APOs. However, 88.76% of pregnant women do not achieve the optimal choline intake. Therefore, specific policies and actions may be necessary to improve choline intake in pregnant women's care and support the well-being of pregnant women. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CDR42023410561.
Collapse
Affiliation(s)
- Hoan Thi Nguyen
- College of Health Care Science, China Medical University, Taichung, Taiwan
- Nursing and Medical Technology, University of Medicine and Pharmacy, Ho Chi Minh City, VietNam
| | | | - Shin-Da Lee
- College of Health Care Science, China Medical University, Taichung, Taiwan
- Department of Physical Therapy, China Medical University, Taichung, Taiwan
| | - Li-Chi Huang
- College of Health Care Science, China Medical University, Taichung, Taiwan
- School of Nursing, China Medical University, Taichung, Taiwan
- Department of Nursing, China Medical University Children Hospital, Taichung, Taiwan
| |
Collapse
|
7
|
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
|
8
|
Rusnak T, Azarcoya-Barrera J, Wollin B, Makarowski A, Nelson R, Field CJ, Jacobs RL, Richard C. A Physiologically Relevant Dose of 50% Egg-Phosphatidylcholine Is Sufficient in Improving Gut Permeability while Attenuating Immune Cell Dysfunction Induced by a High-Fat Diet in Male Wistar Rats. J Nutr 2023; 153:3131-3143. [PMID: 37586605 DOI: 10.1016/j.tjnut.2023.08.010] [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: 06/01/2023] [Revised: 08/02/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Obesity is associated with increased intestinal permeability and a diminished immune response. Phosphatidylcholine (PC), a form of choline found in eggs, has been shown to beneficially modulate T-cell response in the context of obesity when provided as the sole form of choline in the diet. OBJECTIVE This study aimed to determine the impact of varying doses of PC as part of a high-fat diet (HFD) on immune cell function and intestinal permeability. METHODS Male Wistar rats 4 wk of age were randomly assigned to consume 1 of 6 diets for 12 wk containing the same amount of total choline but differing in the forms of choline: 1-control low-fat (CLF, 20% fat, 100% free choline [FC]); 2-control high-fat (CHF, 50% fat, 100% FC); 3-100% PC (100PC, 50% fat, 100% egg-PC); 4-75% PC (75PC, 50% fat, 75% egg-PC+25% FC); 5-50% PC (50PC, 50% fat, 50% egg-PC+50% FC); and 6-25% PC (25PC; 50% fat, 25% egg-PC+75% FC). Intestinal permeability was measured by fluorescein isothiocyanate-dextran. Immune function was assessed by ex vivo cytokine production of splenocytes and cells isolated from the mesenteric lymph node (MLN) after stimulation with different mitogens. RESULTS Feeding the CHF diet increased intestinal permeability compared with the CLF diet, and doses of PC 50% or greater returned permeability to levels similar to that of the CLF diet. Feeding the CHF diet lowered splenocyte production of interleukin (IL)-1β, IL-2, IL-10, and tumor necrosis factor-alpha, and MLN production of IL-2 compared with the CLF group. The 50PC diet most consistently significantly improved cytokine levels (IL-2, IL-10, tumor necrosis factor-alpha) compared with the CHF diet. CONCLUSIONS Our results show that a dose of 50% of total choline derived from egg-PC can ameliorate HFD-induced intestinal permeability and immune cell dysfunction.
Collapse
Affiliation(s)
- Tianna Rusnak
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Jessy Azarcoya-Barrera
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Bethany Wollin
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Alexander Makarowski
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Randal Nelson
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - René L Jacobs
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Caroline Richard
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
9
|
Dewey D, Martin JW, MacDonald AM, Kinniburgh DW, Letourneau N, Giesbrecht GF, Field CJ, Bell RC, England-Mason G. Sex-specific associations between maternal phthalate exposure and neurodevelopmental outcomes in children at 2 years of age in the APrON cohort. Neurotoxicology 2023; 98:48-60. [PMID: 37517784 DOI: 10.1016/j.neuro.2023.07.005] [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: 04/29/2023] [Revised: 07/05/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
BACKGROUND There is inconsistent evidence regarding the sex-specific associations between prenatal phthalate exposure and children's neurodevelopment. This could be due to differences in the phthalate exposures investigated and the neurodevelopmental domains assessed. OBJECTIVE To evaluate the associations between prenatal phthalate exposure and sex-specific outcomes on measures of cognition, language, motor, executive function, and behaviour in children 2 years of age in the Alberta Pregnancy Outcomes and Nutrition (APrON) cohort. METHODS We evaluated the associations between prenatal phthalate exposure and sex-specific neurodevelopmental outcomes in children at 2 years of age using data from 448 mothers and their children (222 girls, 226 boys). Nine phthalate metabolites were measured in maternal urine collected in the second trimester of pregnancy. Children's cognitive, language, and motor outcomes were assessed using the Bayley Scales of Infant Development - Third Edition (Bayley-III). Parents completed questionnaires on children's executive function and behavior, the Behavior Rating Inventory of Executive Function- Preschool Version (BRIEF-P) and Child Behavior Checklist (CBCL), respectively. Sex-stratified robust multivariate regressions were performed. RESULTS Higher maternal concentrations of ΣDEHP and its metabolites were associated with lower scores on the Bayley-III Cognitive (β's from -11.8 to -0.07 95% CI's from -21.3 to -0.01), Language (β's from -11.7 to -0. 09, 95% CI's from -22.3 to -0.02) and Motor (β's from -10.9 to -0.07, 95% CI from -20.4 to -0.01) composites in boys. The patterns of association in girls were in the opposite direction on the Cognitive and Language composites; on the Motor composite they were in the same direction as boys, but of reduced strength. Higher concentrations of ΣDEHP and its metabolites were associated with higher scores (i.e., more difficulties) on all measures of executive function in girls: inhibitory self-control (B's from 0.05 to 0.11, 95% CI s from -0.01 to 0.15), flexibility (B's from 0.04 to 0.11, 95% CI s from 0.01 to 0.21) and emergent metacognition (B's from -0.01 to 0.06, 95% CIs from -0.01 to 0.20). Similar patterns of attenuated associations were seen in boys. Higher concentrations of ΣDEHP and its metabolites were associated with more Externalizing Problems in girls and boys (B's from 0.03 to 6.82, 95% CIs from -0.08 to 12.0). Two phthalates, MMP and MBP, had sex-specific adverse associations on measures of executive function and behaviour, respectively, while MEP was positively associated with boys' cognitive, language, and motor performance. Limited associations were observed between mixtures of maternal phthalates and sex-specific neurodevelopmental outcomes. CONCLUSIONS Maternal prenatal concentrations of DEHP phthalates were associated with sex specific difference on measures of cognition and language at 2 years of age, specifically, poorer outcomes in boys. Higher exposure to DEHP was associated with poorer motor, executive function, and behavioural outcomes in girls and boys but the strength of these associations differed by sex. Limited associations were noted between phthalate mixtures and child neurodevelopment.
Collapse
Affiliation(s)
- Deborah Dewey
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Owerko Centre, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
| | - Jonathan W Martin
- Department of Environmental Science, Science for Life Laboratory, Stockholm University, Stockholm, Sweden
| | - Amy M MacDonald
- Alberta Centre for Toxicology, University of Calgary, Calgary, Alberta, Canada
| | - David W Kinniburgh
- Alberta Centre for Toxicology, University of Calgary, Calgary, Alberta, Canada; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Nicole Letourneau
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Owerko Centre, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Faculty of Nursing, Univerity of Calgary, Calgary, Alberta, Canada; Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gerald F Giesbrecht
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Owerko Centre, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Psychology, Faculty of Arts, University of Calgary, Calgary, Alberta, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutrition Science, University of Alberta, Edmonton, Alberta, Canada
| | - Rhonda C Bell
- Department of Agricultural, Food and Nutrition Science, University of Alberta, Edmonton, Alberta, Canada
| | - Gillian England-Mason
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Owerko Centre, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
10
|
Irvine N, England-Mason G, Field CJ, Letourneau N, Bell RC, Giesbrecht GF, Kinniburgh DW, MacDonald AM, Martin JW, Dewey D. Associations between maternal folate status and choline intake during pregnancy and neurodevelopment at 3-4 years of age in the Alberta Pregnancy Outcomes and Nutrition (APrON) study. J Dev Orig Health Dis 2023; 14:402-414. [PMID: 36939090 PMCID: PMC10202845 DOI: 10.1017/s2040174423000041] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
Folate and choline are methyl donor nutrients that may play a role in fetal brain development. Animal studies have reported that prenatal folate and choline supplementation are associated with better cognitive outcomes in offspring and that these nutrients may interact and affect brain development. Human studies that have investigated associations between maternal prenatal folate or choline levels and neurodevelopmental outcomes have reported contradictory findings and no human studies have examined the potential interactive effect of folate and choline on children's neurodevelopment. During the second trimester of pregnancy, maternal red blood cell folate was measured from blood samples and choline intake was estimated using a 24-h dietary recall in 309 women in the APrON cohort. At 3-5 years of age, their children's neurodevelopment was assessed using the Wechsler Preschool and Primary Scales of Intelligence - Fourth EditionCND, NEPSY-II language and memory subtests, four behavioral executive function tasks, and the Movement Assessment Battery for Children - Second Edition. Adjusted regressions revealed no associations between maternal folate and choline levels during pregnancy and most of the child outcomes. On the Dimensional Change Card Sort, an executive function task, there was an interaction effect; at high levels of choline intake (i.e., 1 SD above the mean; 223.03 mg/day), higher maternal folate status was associated with decreased odds of receiving a passing score (β = -0.44; 95%CI -0.81, -0.06). In conclusion, maternal folate status and choline intake during the second trimester of pregnancy were not associated with children's intelligence, language, memory, or motor outcomes at 3-4 years of age; however, their interaction may have an influence children's executive functions.
Collapse
Affiliation(s)
- Nathalie Irvine
- Bachelor of Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gillian England-Mason
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Owerko Centre, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Nicole Letourneau
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Owerko Centre, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Faculty of Nursing, University of Calgary, Calgary, Alberta, Canada
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rhonda C Bell
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Gerald F Giesbrecht
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Owerko Centre, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
| | - David W Kinniburgh
- Alberta Centre for Toxicology, University of Calgary, Calgary, Alberta, Canada
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Amy M MacDonald
- Alberta Centre for Toxicology, University of Calgary, Calgary, Alberta, Canada
| | - Jonathan W Martin
- Science for Life Laboratory, Department of Environmental Sciences, Stockholm University, Stockholm, Sweden
| | - Deborah Dewey
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Owerko Centre, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
11
|
Hesselink A, Winkvist A, Lindahl B, Ueland PM, Schneede J, Johansson I, Karlsson T. Healthy Nordic diet and associations with plasma concentrations of metabolites in the choline oxidation pathway: a cross-sectional study from Northern Sweden. Nutr J 2023; 22:26. [PMID: 37198607 DOI: 10.1186/s12937-023-00853-w] [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: 09/06/2022] [Accepted: 05/10/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND The choline oxidation pathway and metabolites involved have been linked to diseases including cardiovascular disease, type 2 diabetes and cancer. A healthy Nordic diet is a recently defined dietary pattern associated with decreased risk for these diseases. Our aim was to explore associations between adherence to a healthy Nordic diet and plasma concentrations of metabolites of the choline oxidation pathway. METHODS The Healthy Nordic Food Index (HNFI) and Baltic Sea Diet Score (BSDS) were applied to cross-sectional data (n = 969) from the Västerbotten Intervention Programme in Northern Sweden to score adherence to a healthy Nordic diet. Data included responses to a dietary questionnaire and blood sample analyses (1991-2008). Associations of diet scores with plasma concentrations of metabolites of the choline oxidation pathway and total homocysteine (tHcy), seven metabolites in total, were evaluated with linear regression, adjusting for age, BMI, education and physical activity. RESULTS HNFI scores showed linear relationships with plasma choline (β = 0.11), betaine (β = 0.46), serine (β = 0.98) and tHcy (β = - 0.38), and BSDS scores with betaine (β = 0.13) and tHcy (β = - 0.13); unstandardized beta coefficients, all significant at P < 0.05. The regression models predicted changes in plasma metabolite concentrations (± 1 SD changes in diet score) in the range of 1-5% for choline, betaine, serine and tHcy. No other statistically significant associations were observed. CONCLUSIONS A healthy Nordic diet was associated with plasma concentrations of several metabolites of the choline oxidation pathway. Although relationships were statistically significant, effect sizes were moderate. Further research is warranted to explore the underlying mechanisms and associations with health outcomes.
Collapse
Affiliation(s)
- André Hesselink
- Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, Box 459, 405 30, Gothenburg, Sweden
| | - Anna Winkvist
- Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, Box 459, 405 30, Gothenburg, Sweden
- Department of Public Health and Clinical Medicine, Sustainable Health, Umeå University, Umeå, Sweden
| | - Bernt Lindahl
- Department of Public Health and Clinical Medicine, Sustainable Health, Umeå University, Umeå, Sweden
| | - Per M Ueland
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Bevital AS, Bergen, Norway
| | - Jörn Schneede
- Department of Clinical Pharmacology, Pharmacology and Clinical Neurosciences, Umeå University, Umeå, Sweden
| | | | - Therese Karlsson
- Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, Box 459, 405 30, Gothenburg, Sweden.
- Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden.
| |
Collapse
|
12
|
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
|
13
|
Azarcoya-Barrera J, Lewis ED, Field CJ, Goruk S, Makarowski A, Pouliot Y, Jacobs RL, Richard C. The Lipid-Soluble Forms of Choline Enhance Ex Vivo Responses from the Gut-Associated Immune System in Young Female Rat Offspring. J Nutr 2022; 152:2604-2614. [PMID: 36774126 DOI: 10.1093/jn/nxac180] [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: 03/29/2022] [Revised: 06/16/2022] [Accepted: 08/10/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND In humans, the development of gut-associated lymphoid tissue (GALT) occurs in the first years of life and can be influenced by diet. OBJECTIVES The objective of this study was to determine the effect of dietary choline on the development of gut-associated lymphoid tissue (GALT). METHODS Three feeding trials were conducted in female Sprague-Dawley rats. Beginning 3 d before parturition (studies 1 and 3) or at day 10 of gestation (study 2), control dams consumed a 100% free choline (FC) diet until the end of the lactation period. In studies 1 and 3, test dams consumed a high-glycerophosphocholine (HGPC) diet [75% glycerophosphocholine (GPC), 12.5% phosphatidylcholine (PC), 12.5% FC] and a 100% PC diet, respectively (both 1 g of choline/kg diet). In study 2, test dams consumed a high-sphingomyelin (SM) and PC (SMPC) diet (34% SM, 37% PC, 17% GPC, 7% FC, 5% phosphocholine) or a 50% PC diet (50% PC, 25% FC, 25% GPC), both 1.7 g of choline/kg diet. Immune cell phenotypes and ex vivo cytokine production by mitogen-stimulated immune cells were measured. RESULTS Feeding of the HGPC diet lowered T-cell IL-2 (44%), IFN-γ (34%), and TNF-α (55%) production in mesenteric lymph nodes (MLNs) compared with control. Feeding both SMPC and 50% PC diets during the lactation and weaning periods increased IL-2 (54%) and TNF-α (46%) production after T-cell stimulation compared with control. There was a lower production of IL-2 (46%), IL-6 (66%), and TNF-α (45%), and a higher production of IL-10 (44%) in both SMPC and 50% PC groups following ovalbumin stimulation compared with control in MLNs. Feeding a diet containing 100% PC increased the production of IFN-γ by 52% after T-cell stimulation compared with control. CONCLUSION Feeding a diet containing a mixture of choline forms with a high content of lipid-soluble forms during both the lactation and weaning periods enhances ex vivo immune responses from the GALT in female Sprague-Dawley offspring.
Collapse
Affiliation(s)
- Jessy Azarcoya-Barrera
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Erin D Lewis
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Susan Goruk
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Alexander Makarowski
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Yves Pouliot
- STELA Dairy Research Center, Institute of Nutrition and Functional Foods, Université Laval, Québec, Canada
| | - René L Jacobs
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Caroline Richard
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
14
|
Probst Y, Sulistyoningrum DC, Netting MJ, Gould JF, Wood S, Makrides M, Best KP, Green TJ. Estimated Choline Intakes and Dietary Sources of Choline in Pregnant Australian Women. Nutrients 2022; 14:nu14183819. [PMID: 36145195 PMCID: PMC9503354 DOI: 10.3390/nu14183819] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/29/2022] Open
Abstract
(1) Background: Despite the postulated importance of choline during pregnancy, little is known about the choline intake of Australians during pregnancy. In this study, we estimated dietary intakes of choline in early and late pregnancy, compared those intakes to recommendations, and investigated food sources of choline in a group of pregnant women in Australia. (2) Methods: 103 pregnant women enrolled in a randomized controlled trial. In early pregnancy (12−16 weeks gestation) and late pregnancy (36 weeks gestation), women completed a food frequency questionnaire designed to assess dietary intake over the previous month. (3) Results: Choline intakes and sources were similar in early and late pregnancy. Median choline intake in early pregnancy was 362 mg/day. Of the women, 39% and 25% had choline intakes above the Australian National Health and Medical Research Council (NHMRC) adequate intake (AI) of >440 mg/day and the European Food Safety Authority (EFSA) AI of >480 mg/day for choline in pregnancy, respectively. Eggs, red meat, nuts, legumes, and dairy accounted for 50% of choline intake, with eggs being the most significant contributor at 17%. (4) Conclusions: Few pregnant women in our study met the AI recommended by the NHMRC and EFSA. In Australia, choline intake in pregnancy may need to be improved, but further work to define choline requirements in pregnancy is required.
Collapse
Affiliation(s)
- Yasmine Probst
- School of Medical, Indigenous and Health Sciences, University of Wollongong, Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Dian C. Sulistyoningrum
- Discipline of Paediatrics, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Merryn J. Netting
- Discipline of Paediatrics, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Jacqueline F. Gould
- Discipline of Paediatrics, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Simon Wood
- Factors Group of Nutritional Products Inc. Research & Development, Burnaby, BC V3K 6Y2, Canada
- School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia
| | - Maria Makrides
- Discipline of Paediatrics, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Karen P. Best
- Discipline of Paediatrics, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Tim J. Green
- Discipline of Paediatrics, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Correspondence: ; Tel.: +61-45-244-8438
| |
Collapse
|
15
|
Robb L, Jordaan EM, Joubert G, Ngounda J, Walsh CM. Sociodemographic Indicators, Household Food Security and Associations with Choline Intake in Pregnant Women: The NuEMI Study. Ecol Food Nutr 2022; 61:669-686. [PMID: 36083168 DOI: 10.1080/03670244.2022.2118734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Higher sociodemographic status is associated with adequate nutrient intake and food security. Adequate choline intake is vital during pregnancy to support fetal development. In this cross-sectional study, information was obtained from 682 pregnant women attending an ante-natal clinic in Bloemfontein, South Africa. Sociodemographic indicators that were bivariately significantly associated with a choline intake below the adequate intake level (AI) included a higher household density ratio, no access to own flush toilets at home, household not owning a refrigerator or microwave, as well as a lower level of education. Approximately one-third of participants were severely food-insecure. Logistic regression analysis, adjusted for energy intake, showed that household overcrowding increased the odds of a choline intake below the AI (OR 1.71).
Collapse
Affiliation(s)
- Liska Robb
- Department of Nutrition and Dietetics, School of Health and Rehabilitation Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein, Republic of South Africa
| | - Elizabeth Margaretha Jordaan
- Department of Nutrition and Dietetics, School of Health and Rehabilitation Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein, Republic of South Africa
| | - Gina Joubert
- Department of Biostatistics, School of Biomedical Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein, Republic of South Africa
| | - Jennifer Ngounda
- Department of Nutrition and Dietetics, School of Health and Rehabilitation Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein, Republic of South Africa
| | - Corinna May Walsh
- Department of Nutrition and Dietetics, School of Health and Rehabilitation Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein, Republic of South Africa
| |
Collapse
|
16
|
Carter RC, Senekal M, Duggan CP, Dodge NC, Meintjes EM, Molteno CD, Jacobson JL, Jacobson SW. Gestational weight gain and dietary energy, iron, and choline intake predict severity of fetal alcohol growth restriction in a prospective birth cohort. Am J Clin Nutr 2022; 116:460-469. [PMID: 35441212 PMCID: PMC9348980 DOI: 10.1093/ajcn/nqac101] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/11/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Animal models have demonstrated that maternal nutrition can alter fetal vulnerability to prenatal alcohol exposure (PAE). Few human studies have examined the role of nutrition in fetal alcohol spectrum disorders (FASD). OBJECTIVES Our objectives were to examine whether fetal vulnerability to PAE-related growth restriction is modified by: 1) rate of gestational weight gain; or prenatal dietary intakes of 2) energy, 3) iron, or 4) choline. METHODS In a prospective longitudinal birth cohort in Cape Town, South Africa, 118 heavy-drinking and 71 abstaining/light-drinking pregnant women were weighed and interviewed regarding demographics, alcohol, cigarette/other drug use, and diet at prenatal visits. Infant length, weight, and head circumference were measured at 2 wk and 12 mo postpartum. RESULTS Heavy-drinking mothers reported a binge pattern of drinking [Mean = 129 mL (∼7.2 drinks)/occasion on 1.3 d/wk). Rate of gestational weight gain and average daily dietary energy, iron, and choline intakes were similar between heavy-drinking women and controls. In regression models adjusting for maternal age, socioeconomic status, cigarette use, and weeks gestation at delivery, PAE [ounces (30 mL) absolute alcohol per day] was related to smaller 2-wk length and head circumference and 12-mo length, weight, and head circumference z-scores (β = -0.43 to -0.67; all P values <0.05). In stratified analyses for each maternal nutritional measure (inadequate compared with adequate weight gain; tertiles for dietary energy, iron, and choline intakes), PAE-related growth restriction was more severe in women with poorer nutrition, with effect modification seen by weight gain, energy, iron, and/or choline for several anthropometric outcomes. CONCLUSIONS Gestational weight gain and dietary intakes of energy, choline, and iron appeared to modify fetal vulnerability to PAE-related growth restriction. These findings suggest a need for screening programs for pregnant women at higher risk of having a child with FASD to identify alcohol-using women who could benefit from nutritional interventions.
Collapse
Affiliation(s)
| | - Marjanne Senekal
- Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| | - Christopher P Duggan
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Neil C Dodge
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ernesta M Meintjes
- Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| | - Christopher D Molteno
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA, USA,Departments of Nutrition and Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA, USA,Department of Psychiatry and Mental Health, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| | - Joseph L Jacobson
- Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa,Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Sandra W Jacobson
- Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa,Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA,Department of Psychiatry and Mental Health, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| |
Collapse
|
17
|
She Y, Mangat R, Tsai S, Proctor SD, Richard C. The Interplay of Obesity, Dyslipidemia and Immune Dysfunction: A Brief Overview on Pathophysiology, Animal Models, and Nutritional Modulation. Front Nutr 2022; 9:840209. [PMID: 35252310 PMCID: PMC8891442 DOI: 10.3389/fnut.2022.840209] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/20/2022] [Indexed: 12/14/2022] Open
Abstract
Obesity has emerged as a leading global health concern. It is characterized by chronic low-grade inflammation, which impairs insulin signaling, lipid metabolism and immune function. Recent findings from animal and clinical studies have begun to elucidate the underlying mechanisms of immune dysfunction seen in the context of obesity. Here, we provide a brief review on the current understanding of the interplay between obesity, dyslipidemia and immunity. We also emphasize the advantages and shortcomings of numerous applicable research models including rodents and large animal swine that aim at unraveling the molecular basis of disease and clinical manifestations. Although there is no perfect model to answer all questions at once, they are often used to complement each other. Finally, we highlight some emerging nutritional strategies to improve immune function in the context of obesity with a particular focus on choline and foods that contains high amounts of choline.
Collapse
Affiliation(s)
- Yongbo She
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Rabban Mangat
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Sue Tsai
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Spencer D. Proctor
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Caroline Richard
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
18
|
Letourneau N, Aghajafari F, Bell RC, Deane AJ, Dewey D, Field C, Giesbrecht G, Kaplan B, Leung B, Ntanda H. The Alberta Pregnancy Outcomes and Nutrition (APrON) longitudinal study: cohort profile and key findings from the first three years. BMJ Open 2022; 12:e047503. [PMID: 35131812 PMCID: PMC8823238 DOI: 10.1136/bmjopen-2020-047503] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
PURPOSE The objectives of the ongoing Canadian longitudinal cohort called the Alberta Pregnancy Outcomes and Nutrition (APrON) study are to: (1) determine the relationship between maternal nutrient intake and status before, during, after pregnancy, and (a) maternal mental health, (b) pregnancy and birth outcomes, and (c) infant/child neurodevelopment and behavior; (2) identify maternal mental health and nutrient predictors of child behaviour; and (3) establish a DNA biobank to explore genomic predictors of children's neurodevelopment and behavior. The purpose of this paper is to describe the participants, measures, and key findings on maternal and paternal mental health, maternal nutrition, and child outcomes to when children are 3 years of age. PARTICIPANTS Participants included mothers and their children (n=2189) and mothers' partners (usually fathers; n=1325) from whom data were collected during the period from pregnancy to when children were 3 years of age, in Alberta, Canada. More than 88% of families have been retained to take part in completed data collection at 8 years of age. FINDINGS TO DATE Data comprise: questionnaires completed by pregnant women/mothers and their partners on mothers', fathers' and children's health; dietary interviews; clinical assessments; linkage to hospital obstetrical records; and biological samples such as DNA. Key findings on mental health, nutrition and child outcomes are presented. APrON women who consumed more selenium and omega-3 were less likely to develop symptoms of perinatal depression. Higher prenatal consumption of choline rich foods such as eggs and milk were recommended as was vitamin D supplementation for both mothers and children to meet guidelines. Couples in which both mothers and fathers were affected by perinatal depression reported lower incomes and higher maternal prenatal depressive symptoms and lower support from fathers postnatally and their children presented with the most behavioural problems. Maternal experiences of early adversity predicted increased likelihood of perinatal depression and anxiety and children's behavioural problems. FUTURE PLANS The APrON cohort offers a unique opportunity to advance understanding of the developmental origins of health and disease. There is a planned follow-up to collect data at 12 years of age.
Collapse
Affiliation(s)
- Nicole Letourneau
- Faculty of Nursing, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine (Departments of Paediatrics, Psychiatry & Community Health Sciences), University of Calgary, Calgary, Alberta, Canada
| | - Fariba Aghajafari
- Cumming School of Medicine (Family Medicine & Community Health Sciences), University of Calgary, Calgary, Alberta, Canada
| | - Rhonda C Bell
- Faculty of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Andrea J Deane
- Cumming School of Medicine (Pediatrics), University of Calgary, Calgary, Alberta, Canada
| | - Deborah Dewey
- Cumming School of Medicine (Pediatrics), University of Calgary, Calgary, Alberta, Canada
| | - Catherine Field
- Faculty of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Gerald Giesbrecht
- Cumming School of Medicine (Pediatrics & Community Health Sciences), University of Calgary, Calgary, Alberta, Canada
| | - Bonnie Kaplan
- Cumming School of Medicine (Pediatrics & Community Health Sciences), University of Calgary, Calgary, Alberta, Canada
| | - Brenda Leung
- Health Science - Public Health Program, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Henry Ntanda
- Cumming School of Medicine (Pediatrics), University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
19
|
Azarcoya-Barrera J, Wollin B, Veida-Silva H, Makarowski A, Goruk S, Field CJ, Jacobs RL, Richard C. Egg-Phosphatidylcholine Attenuates T-Cell Dysfunction in High-Fat Diet Fed Male Wistar Rats. Front Nutr 2022; 9:811469. [PMID: 35187037 PMCID: PMC8847771 DOI: 10.3389/fnut.2022.811469] [Citation(s) in RCA: 5] [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: 11/08/2021] [Accepted: 01/10/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity is associated with immune dysfunction including an impaired T-cell function characterized by a lower IL-2 (proliferation marker) production after stimulation. Phosphatidylcholine (PC), a form of choline mostly found in eggs, has been shown to beneficially modulate T-cell responses during the lactation period by increasing the production of IL-2. To determine the impact of egg-PC as part of a high-fat diet on immune function we randomly fed male Wistar rats one of three diets containing the same amount of total choline but differing in the form of choline: 1-Control low fat [CLF, 10% wt/wt fat, 100% free choline (FC)]; 2- Control high-fat (CHF, 25% wt/wt fat, 100% FC); 3- PC high-fat (PCHF, 25% wt/wt, 100% PC). After 9 weeks of feeding, rats were euthanized. Cell phenotypes and ex vivo cytokine production by splenocytes stimulated with phorbol 12-myristate 13-acetate plus ionomycin (PMA+I), lipopolysaccharide (LPS) and pokeweed (PWM) were measured by flow cytometry and ELISA, respectively. Rats fed the PCHF diet had a lower proportion of CD3+ cells when compared to both the CLF and the CHF. Following PMA+I stimulation, splenocytes from the CHF group produced less IL-2 and TNF-α compared to CLF and PCHF groups. No significant differences in cytokine production were found among groups after LPS and PWM stimulation. Our results show that feeding a high-fat diet impairs T-cell responses, as measured by ex vivo cytokine production, which can be attenuated by providing egg-PC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Caroline Richard
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
20
|
Irvine N, England-Mason G, Field CJ, Dewey D, Aghajafari F. Prenatal Folate and Choline Levels and Brain and Cognitive Development in Children: A Critical Narrative Review. Nutrients 2022; 14:nu14020364. [PMID: 35057545 PMCID: PMC8778665 DOI: 10.3390/nu14020364] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 01/13/2023] Open
Abstract
Women’s nutritional status during pregnancy can have long-term effects on children’s brains and cognitive development. Folate and choline are methyl-donor nutrients and are important for closure of the neural tube during fetal development. They have also been associated with brain and cognitive development in children. Animal studies have observed that prenatal folate and choline supplementation is associated with better cognitive outcomes in offspring and that these nutrients may have interactive effects on brain development. Although some human studies have reported associations between maternal folate and choline levels and child cognitive outcomes, results are not consistent, and no human studies have investigated the potential interactive effects of folate and choline. This lack of consistency could be due to differences in the methods used to assess folate and choline levels, the gestational trimester at which they were measured, and lack of consideration of potential confounding variables. This narrative review discusses and critically reviews current research examining the associations between maternal levels of folate and choline during pregnancy and brain and cognitive development in children. Directions for future research that will increase our understanding of the effects of these nutrients on children’s neurodevelopment are discussed.
Collapse
Affiliation(s)
- Nathalie Irvine
- O’Brien Centre for the Bachelor of Health Sciences, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada;
| | - Gillian England-Mason
- Owerko Centre, Alberta Children’s Hospital Research Institute, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada; (G.E.-M.); (D.D.)
- Department of Pediatrics, Cumming School of Medicine, Alberta Children’s Hospital, 28 Oki Drive NW, Calgary, AB T3B 6A8, Canada
| | - Catherine J. Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, 4-126C Li Ka Shing Centre for Research, 11203-87th Ave NW, Edmonton, AB T6G 2H5, Canada;
| | - Deborah Dewey
- Owerko Centre, Alberta Children’s Hospital Research Institute, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada; (G.E.-M.); (D.D.)
- Department of Pediatrics, Cumming School of Medicine, Alberta Children’s Hospital, 28 Oki Drive NW, Calgary, AB T3B 6A8, Canada
- Hotchkiss Brain Institute, Health Research Innovation Centre, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, 3D10, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada
| | - Fariba Aghajafari
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, 3D10, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada
- Department of Family Medicine, Cumming School of Medicine, University of Calgary, G012, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
- Correspondence:
| |
Collapse
|
21
|
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: 20] [Impact Index Per Article: 6.7] [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
|
22
|
Mild Choline Deficiency and MTHFD1 Synthetase Deficiency Interact to Increase Incidence of Developmental Delays and Defects in Mice. Nutrients 2021; 14:nu14010127. [PMID: 35011003 PMCID: PMC8747146 DOI: 10.3390/nu14010127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 01/07/2023] Open
Abstract
Folate and choline are interconnected metabolically. The MTHFD1 R653Q SNP is a risk factor for birth defects and there are concerns that choline deficiency may interact with this SNP and exacerbate health risks. 80–90% of women do not meet the Adequate Intake (AI) for choline. The objective of this study was to assess the effects of choline deficiency on maternal one-carbon metabolism and reproductive outcomes in the MTHFD1-synthetase deficient mouse (Mthfd1S), a model for MTHFD1 R653Q. Mthfd1S+/+ and Mthfd1S+/− females were fed control (CD) or choline-deficient diets (ChDD; 1/3 the amount of choline) before mating and during pregnancy. Embryos were evaluated for delays and defects at 10.5 days gestation. Choline metabolites were measured in the maternal liver, and total folate measured in maternal plasma and liver. ChDD significantly decreased choline, betaine, phosphocholine, and dimethylglycine in maternal liver (p < 0.05, ANOVA), and altered phosphatidylcholine metabolism. Maternal and embryonic genotype, and diet-genotype interactions had significant effects on defect incidence. Mild choline deficiency and Mthfd1S+/− genotype alter maternal one-carbon metabolism and increase incidence of developmental defects. Further study is required to determine if low choline intakes contribute to developmental defects in humans, particularly in 653QQ women.
Collapse
|
23
|
Robb L, Joubert G, Jordaan EM, Ngounda J, Walsh CM. Choline intake and associations with egg and dairy consumption among pregnant women attending a high-risk antenatal clinic in South Africa: the NuEMI study. BMC Pregnancy Childbirth 2021; 21:833. [PMID: 34906117 PMCID: PMC8670152 DOI: 10.1186/s12884-021-04314-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 12/01/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The importance of adequate choline intake during pregnancy has been well documented, but low intake is common. Total choline intake, main food sources of choline, as well as associations between choline intake and egg and dairy consumption were determined in a sample of pregnant women attending the high-risk antenatal clinic at a regional hospital in Bloemfontein, South Africa. METHODS A cross-sectional study design was used. Trained fieldworkers collected dietary intake data using a validated quantified food frequency questionnaire (QFFQ), after which all food items were matched to foods in the USDA Database for the Choline Content of Common Foods (Release 2) to quantify choline intake. Logistic regression with backward selection (p < 0.05) was used to determine whether egg and dairy consumption were independently associated with a choline intake below the adequate intake (AI) level. RESULTS The median daily intake of choline was 275 mg (interquartile range 185 mg - 387 mg) (N = 681). Most participants (84.7%) consumed less than the AI of 450 mg/day for choline. Meat and meat products, cereals, eggs and dairy contributed mostly to choline intake. Food items that contributed most to choline intake included full-cream milk, maize porridge, brown bread, deep-fried potatoes and deep-fried dough (vetkoek). A choline intake below the AI was significantly associated with lower egg and dairy intakes (p < 0.0001 and p = 0.0002 respectively). CONCLUSION Most pregnant women in the current study had choline intakes below the AI. It is recommended that public health messaging targeted at pregnant women promote the consumption of foods that can significantly contribute to choline intake, such as eggs and dairy.
Collapse
Affiliation(s)
- Liska Robb
- Department of Nutrition and Dietetics, School of Health and Rehabilitation Sciences, Faculty of Health Sciences, University of the Free State, PO Box 339, internal box G24, Bloemfontein, 9300, Free State, Republic of South Africa.
| | - Gina Joubert
- Department of Biostatistics, School of Biomedical Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein, Republic of South Africa
| | - Elizabeth Margaretha Jordaan
- Department of Nutrition and Dietetics, School of Health and Rehabilitation Sciences, Faculty of Health Sciences, University of the Free State, PO Box 339, internal box G24, Bloemfontein, 9300, Free State, Republic of South Africa
| | - Jennifer Ngounda
- Department of Nutrition and Dietetics, School of Health and Rehabilitation Sciences, Faculty of Health Sciences, University of the Free State, PO Box 339, internal box G24, Bloemfontein, 9300, Free State, Republic of South Africa
| | - Corinna May Walsh
- Department of Nutrition and Dietetics, School of Health and Rehabilitation Sciences, Faculty of Health Sciences, University of the Free State, PO Box 339, internal box G24, Bloemfontein, 9300, Free State, Republic of South Africa
| |
Collapse
|
24
|
Derbyshire E, Obeid R, Schön C. Habitual Choline Intakes across the Childbearing Years: A Review. Nutrients 2021; 13:nu13124390. [PMID: 34959942 PMCID: PMC8709092 DOI: 10.3390/nu13124390] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 11/20/2022] Open
Abstract
Choline is an important nutrient during the first 1000 days post conception due to its roles in brain function. An increasing number of studies have measured choline intakes at the population level. We collated the evidence focusing on habitual choline intakes in the preconceptual, pregnancy, and lactation life stages. We conducted a review including studies published from 2004 to 2021. Twenty-six relevant publications were identified. After excluding studies with a high choline intake (>400 mg/day; two studies) or low choline intake (<200 mg/day; one study), average choline intake in the remaining 23 studies ranged from 233 mg/day to 383 mg/day, even with the inclusion of choline from supplements. Intakes were not higher in studies among pregnant and lactating women compared with studies in nonpregnant women. To conclude, during the childbearing years and across the globe, habitual intakes of choline from foods alone and foods and supplements combined appear to be consistently lower than the estimated adequate intakes for this target group. Urgent measures are needed to (1) improve the quality of choline data in global food composition databases, (2) encourage the reporting of choline intakes in dietary surveys, (3) raise awareness about the role(s) of choline in foetal–maternal health, and (4) consider formally advocating the use of choline supplements in women planning a pregnancy, pregnant, or lactating.
Collapse
Affiliation(s)
| | - Rima Obeid
- Department of Clinical Chemistry and Laboratory Medicine, University Hospital of the Saarland, D-66420 Homburg, Germany;
| | - Christiane Schön
- BioTeSys GmbH, Schelztorstrasse 54-56, D-73728 Esslingen, Germany;
| |
Collapse
|
25
|
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: 3.7] [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
|
26
|
Van Parys A, Brække MS, Karlsson T, Vinknes KJ, Tell GS, Haugsgjerd TR, Ueland PM, Øyen J, Dierkes J, Nygård O, Lysne V. Assessment of Dietary Choline Intake, Contributing Food Items, and Associations with One-Carbon and Lipid Metabolites in Middle-Aged and Elderly Adults: The Hordaland Health Study. J Nutr 2021; 152:513-524. [PMID: 34643705 PMCID: PMC8826836 DOI: 10.1093/jn/nxab367] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/19/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Choline is an essential nutrient for humans and is involved in various physiologic functions. Through its metabolite betaine, it is closely connected to the one-carbon metabolism, and the fat-soluble choline form phosphatidylcholine is essential for VLDL synthesis and secretion in the liver connecting choline to the lipid metabolism. Dietary recommendations for choline are not available in the Nordic countries primarily due to data scarcity. OBJECTIVES The aim of this study was to investigate the dietary intake of total choline and individual choline forms, dietary sources, and the association of total choline intake with circulating one-carbon metabolites and lipids. METHODS We included 5746 participants in the Hordaland Health Study, a survey including community-dwelling adults born in 1925-1927 (mean age 72 y, 55% women) and 1950-1951 (mean age 48 y, 57% women). Dietary data were obtained using a 169-item FFQ, and choline content was calculated using the USDA Database for Choline Content of Common Foods, release 2. Metabolites of the one-carbon and lipid metabolism were measured in a nonfasting blood sample obtained at baseline, and the association with total choline intake was assessed using polynomial splines. RESULTS The geometric mean (95% prediction interval) energy-adjusted total choline intake was 260 (170, 389) mg/d, with phosphatidylcholine being the main form (44%). The major food items providing dietary choline were eggs, low-fat milk, potatoes, and leafy vegetables. Dietary total choline was inversely associated with circulating concentrations of total homocysteine, glycine, and serine and positively associated with choline, methionine, cystathionine, cysteine, trimethyllysine, trimethylamine-N-oxide, and dimethylglycine. A weak association was observed between choline intake and serum lipids. CONCLUSIONS Phosphatidylcholine was the most consumed choline form in community-dwelling adults in Norway. Our findings suggest that choline intake is associated with the concentration of most metabolites involved in the one-carbon and lipid metabolism.
Collapse
Affiliation(s)
| | - Maria Sandvik Brække
- Centre for Nutrition, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Therese Karlsson
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kathrine J Vinknes
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Grethe S Tell
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Teresa R Haugsgjerd
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | | | | | - Jutta Dierkes
- Mohn Nutrition Research Laboratory, University of Bergen, Bergen, Norway,Centre for Nutrition, Department of Clinical Medicine, University of Bergen, Bergen, Norway,Department of Laboratory Medicine and Pathology, Haukeland University Hospital, Bergen, Norway
| | - Ottar Nygård
- Centre for Nutrition, Department of Clinical Science, University of Bergen, Bergen, Norway,Mohn Nutrition Research Laboratory, University of Bergen, Bergen, Norway,Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Vegard Lysne
- Centre for Nutrition, Department of Clinical Science, University of Bergen, Bergen, Norway,Mohn Nutrition Research Laboratory, University of Bergen, Bergen, Norway,Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
27
|
Liu J, Martin LJ, Dinu I, Field CJ, Dewey D, Martin JW. Interaction of prenatal bisphenols, maternal nutrients, and toxic metal exposures on neurodevelopment of 2-year-olds in the APrON cohort. ENVIRONMENT INTERNATIONAL 2021; 155:106601. [PMID: 33962233 DOI: 10.1016/j.envint.2021.106601] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Epidemiological studies suggest that Bisphenol-A (BPA) is a developmental neurotoxicant, but the modifying effects of maternal nutrient status or neurotoxicant metal co-exposures have not been reported. Bisphenol-S (BPS) is being used as a BPA-alternative, but few epidemiological studies have evaluated its effects. OBJECTIVES To examine if prenatal maternal BPA or BPS exposure are associated with children's neurodevelopment at two years of age while adjusting for effect-measure modification by sex, maternal nutrients, and co-exposure to neurotoxic metals. METHODS Total BPA and BPS concentrations were analyzed in spot maternal urine from the second trimester; metals and maternal nutrient status were analyzed in blood. Child neurodevelopment was evaluated with the Bayley Scales of Infant Development-III (Bayley-III) at age 2 (394 maternal-child pairs) and linear regression was used to investigate associations. RESULTS Among nutrients and neurotoxic metals, selenium (Se) and cadmium (Cd) were the most significant predictors of Bayley-III scale scores. Higher maternal Cd was significantly correlated with poorer motor performance (p < 0.01), and higher levels of maternal Se were significantly associated with poorer performance on the cognitive, motor, and adaptive behavior scales (p < 0.05). While maternal Cd did not modify relationships between bisphenol exposures and Bayley-III scores, both maternal Se and child sex were significant effect-measure modifiers. Associations between BPA exposure and social emotional scores were negative for boys (p = 0.056) but positive for girls (p = 0.046). Higher exposure to bisphenols was associated with lower motor scores among children with lower levels of maternal Se. CONCLUSION Higher maternal Cd was associated with poorer motor development, but it was not an effect-measure modifier of bisphenols' effects on motor development. Maternal Se may be protective against adverse effects of bisphenols, and additional nutrient-bisphenol interaction studies examining sex-specific effects of BPA and BPS on child development are warranted.
Collapse
Affiliation(s)
- Jiaying Liu
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Canada; Department of Nutrition and Health, China Agricultural University, Beijing, China
| | | | - Irina Dinu
- School of Public Health, University of Alberta, Canada
| | - Catherine J Field
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Canada
| | - Deborah Dewey
- Departments of Paediatrics and Community Health Sciences and the Owerko Centre at the Alberta Children's Hospital Research Institute, University of Calgary, Canada
| | - Jonathan W Martin
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Canada; Science for Life Laboratory, Department of Environmental Sciences, Stockholm University, Stockholm, Sweden.
| |
Collapse
|
28
|
Warton FL, Molteno CD, Warton CMR, Wintermark P, Lindinger NM, Dodge NC, Zöllei L, van der Kouwe AJW, Carter RC, Jacobson JL, Jacobson SW, Meintjes EM. Maternal choline supplementation mitigates alcohol exposure effects on neonatal brain volumes. Alcohol Clin Exp Res 2021; 45:1762-1774. [PMID: 34342017 DOI: 10.1111/acer.14672] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/07/2021] [Accepted: 07/01/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) is associated with smaller regional and global brain volumes. In rats, gestational choline supplementation mitigates adverse developmental effects of ethanol exposure. Our recent randomized, double-blind, placebo-controlled maternal choline supplementation trial showed improved somatic and functional outcomes in infants at 6.5 and 12 months postpartum. Here, we examined whether maternal choline supplementation protected the newborn brain from PAE-related volume reductions and, if so, whether these volume changes were associated with improved infant recognition memory. METHODS Fifty-two infants born to heavy-drinking women who had participated in a choline supplementation trial during pregnancy underwent structural magnetic resonance imaging with a multi-echo FLASH protocol on a 3T Siemens Allegra MRI (median age = 2.8 weeks postpartum). Subcortical regions were manually segmented. Recognition memory was assessed at 12 months on the Fagan Test of Infant Intelligence (FTII). We examined the effects of choline on regional brain volumes, whether choline-related volume increases were associated with higher FTII scores, and the degree to which the regional volume increases mediated the effects of choline on the FTII. RESULTS Usable MRI data were acquired in 50 infants (choline: n = 27; placebo: n = 23). Normalized volumes were larger in six of 12 regions in the choline than placebo arm (t ≥ 2.05, p ≤ 0.05) and were correlated with the degree of maternal choline adherence (β ≥ 0.28, p ≤ 0.04). Larger right putamen and corpus callosum were related to higher FTII scores (r = 0.36, p = 0.02) with a trend toward partial mediation of the choline effect on recognition memory. CONCLUSIONS High-dose choline supplementation during pregnancy mitigated PAE-related regional volume reductions, with larger volumes associated with improved 12-month recognition memory. These results provide the first evidence that choline may be neuroprotective against PAE-related brain structural deficits in humans.
Collapse
Affiliation(s)
- Fleur L Warton
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Biomedical Engineering Research Centre, Division of Biomedical Engineering, University of Cape Town, Cape Town, South Africa.,Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Christopher D Molteno
- Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Christopher M R Warton
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Pia Wintermark
- Department of Pediatrics, Montreal Children's Hospital, McGill University, Montreal, Canada
| | - Nadine M Lindinger
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,ACSENT Laboratory, Department of Psychology, University of Cape Town, Cape Town, South Africa
| | - Neil C Dodge
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Lilla Zöllei
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Andre J W van der Kouwe
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - R Colin Carter
- Division of Pediatric Emergency Medicine, Columbia University Medical Center, New York, New York, USA
| | - Joseph L Jacobson
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Sandra W Jacobson
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Ernesta M Meintjes
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Biomedical Engineering Research Centre, Division of Biomedical Engineering, University of Cape Town, Cape Town, South Africa.,Neuroscience Institute, University of Cape Town, Cape Town, South Africa.,Cape Universities Body Imaging Centre, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
29
|
Azarcoya-Barrera J, Field CJ, Goruk S, Makarowski A, Curtis JM, Pouliot Y, Jacobs RL, Richard C. Buttermilk: an important source of lipid soluble forms of choline that influences the immune system development in Sprague-Dawley rat offspring. Eur J Nutr 2021; 60:2807-2818. [PMID: 33416979 DOI: 10.1007/s00394-020-02462-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE To determine the effect of feeding buttermilk-derived choline metabolites on the immune system development in Sprague-Dawley rat pups. METHODS Sprague-Dawley dams were randomized to one of the three diets containing 1.7 g/kg choline: 1-Control (100% free choline (FC)), 2-Buttermilk (BM, 37% phosphatidylcholine (PC), 34% sphingomyelin (SM), 17% glycerophosphocholine (GPC), 7% FC, 5% phosphocholine), and 3-Placebo (PB, 50% PC, 25% FC, 25% GPC) until the end of the lactation period. At weaning, pups continued on the same diet as their mom. Cell phenotypes and cytokine production by mitogen-stimulated splenocytes isolated from 3- and 10-week-old pups were measured. RESULTS At 3 weeks, BM-pups had a higher proportion of cytotoxic T cells (CTL; CD3 + CD8 +) while both BM- and PB-pups had an increased proportion of cells expressing CD28 + , CD86 + and CD27 + (all p > 0.05). Following ConA stimulation, splenocytes from BM- and PB-pups produced more TNF-α and IFN-γ and after LPS stimulation produced more IL-10 and TNF-α (all p > 0.05). Starting at week 6 of age, BM-pups had a higher body weight. At 10 weeks, both the BM- and PB-pups had a higher proportion of CTL expressing CD27 + . After ConA stimulation, splenocytes from BM- and PB-pups produced more IL-2, IFN-γ and IL-6 and more IL-10 after LPS stimulation (all p > 0.05). CONCLUSION The proportion of lipid soluble forms of choline in the diet during lactation and weaning periods influence the immune system development in rat offspring.
Collapse
Affiliation(s)
- Jessy Azarcoya-Barrera
- Department of Agricultural, Food and Nutritional Science, Center for Health Research Innovation, University of Alberta, 4-002G Li Ka Shing, Edmonton, AB, T6G 2E1, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, Center for Health Research Innovation, University of Alberta, 4-002G Li Ka Shing, Edmonton, AB, T6G 2E1, Canada
| | - Susan Goruk
- Department of Agricultural, Food and Nutritional Science, Center for Health Research Innovation, University of Alberta, 4-002G Li Ka Shing, Edmonton, AB, T6G 2E1, Canada
| | - Alexander Makarowski
- Department of Agricultural, Food and Nutritional Science, Center for Health Research Innovation, University of Alberta, 4-002G Li Ka Shing, Edmonton, AB, T6G 2E1, Canada
| | - Jonathan M Curtis
- Department of Agricultural, Food and Nutritional Science, Center for Health Research Innovation, University of Alberta, 4-002G Li Ka Shing, Edmonton, AB, T6G 2E1, Canada
| | - Yves Pouliot
- STELA Dairy Research Center, Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, Canada
| | - René L Jacobs
- Department of Agricultural, Food and Nutritional Science, Center for Health Research Innovation, University of Alberta, 4-002G Li Ka Shing, Edmonton, AB, T6G 2E1, Canada
| | - Caroline Richard
- Department of Agricultural, Food and Nutritional Science, Center for Health Research Innovation, University of Alberta, 4-002G Li Ka Shing, Edmonton, AB, T6G 2E1, Canada.
| |
Collapse
|
30
|
The role of maternal nutrition during pregnancy in the intergenerational transmission of childhood adversity. Psychoneuroendocrinology 2021; 130:105283. [PMID: 34082275 DOI: 10.1016/j.psyneuen.2021.105283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 01/30/2023]
Abstract
Adverse childhood experiences (ACEs) of a woman can lead to dysregulated hypothalamus-pituitary-adrenal (HPA) axis during pregnancy, which can in turn adversely affect her offspring HPA axis function. Choline and docosahexaenoic acid (DHA) are dietary factors with the potential to favorably modify the stress response system. The current study aimed to investigate whether maternal choline intake and DHA status moderate the effects of maternal ACEs exposure on maternal and infant HPA axes function. Participants were a sub-sample of the prospective longitudinal Alberta Pregnancy Outcomes and Nutrition (APrON) study consisting of 340 mothers and 238 infants. We collected data on maternal ACEs, maternal choline intake (24-hour dietary recall) and serum phospholipid DHA concentrations (at each trimester). Women self-collected saliva samples on two consecutive days (at waking, +30 min, 1100 h, and 2100 h) in each trimester to calculate the cortisol awakening response (CAR) and total daytime cortisol. Infants' salivary cortisol was measured before and after (20, and 40 min) exposure to a blood draw stressor 3 months postpartum. During pregnancy, choline intake moderated (reduced) the association between maternal ACEs and CAR (β = -0.003; 95% CI -0.006, -0.003), but not total daytime cortisol. DHA status did not moderate the association between ACEs and CAR or total daytime cortisol. Choline intake also moderated (reduced) the association between maternal CAR and infant cortisol during a stress task (β = -0.0001; 95% CI -0.0002, -0.00003). Maternal DHA status revealed no modifying effects on these associations. Our findings suggest that maternal choline intake, but not DHA status, can buffer the associations between ACEs and maternal HPA axis, as well as maternal and infant HPA axes function.
Collapse
|
31
|
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: 13] [Impact Index Per Article: 4.3] [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
|
32
|
Van Parys A, Karlsson T, Vinknes KJ, Olsen T, Øyen J, Dierkes J, Nygård O, Lysne V. Food Sources Contributing to Intake of Choline and Individual Choline Forms in a Norwegian Cohort of Patients With Stable Angina Pectoris. Front Nutr 2021; 8:676026. [PMID: 34055860 PMCID: PMC8160433 DOI: 10.3389/fnut.2021.676026] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/22/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Choline is an essential nutrient involved in a wide range of physiological functions. It occurs in water- and lipid-soluble forms in the body and diet. Foods with a known high choline content are eggs, beef, chicken, milk, fish, and selected plant foods. An adequate intake has been set in the US and Europe, however, not yet in the Nordic countries. A higher intake of lipid-soluble choline forms has been associated with increased risk of acute myocardial infarction, highlighting the need for knowledge about food sources of the individual choline forms. In general, little is known about the habitual intake and food sources of choline, and individual choline forms. Objective: Investigate foods contributing to the intake of total choline and individual choline forms. Design: The study population consisted of 1,929 patients with stable angina pectoris from the Western Norway B Vitamin Intervention Trial. Dietary intake data was obtained through a 169-item food frequency questionnaire. Intake of total choline and individual choline forms was quantified using the USDA database, release 2. Results: The geometric mean (95% prediction interval) total choline intake was 287 (182, 437) mg/d. Phosphatidylcholine accounted for 42.5% of total choline intake, followed by free choline (25.8%) and glycerophosphocholine (21.2%). Phosphocholine and sphingomyelin contributed 4.2 and 4.5%, respectively. The main dietary choline sources were eggs, milk, fresh vegetables, lean fish, and bread. In general, animal food sources were the most important contributors to choline intake. Conclusion: This study is, to the best of our knowledge, the first to assess the intake of all choline forms and their dietary sources in a European population. Most choline was consumed in the form of phosphatidylcholine and animal food sources contributed most to choline intake. There is a need for accurate estimates of the dietary intake of this essential nutrient to issue appropriate dietary recommendations.
Collapse
Affiliation(s)
- Anthea Van Parys
- Centre for Nutrition, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Therese Karlsson
- Department of Internal Medicine and Clinical Nutrition, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kathrine J. Vinknes
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Jutta Dierkes
- Mohn Nutrition Research Laboratory, University of Bergen, Bergen, Norway
- Centre for Nutrition, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Laboratory Medicine and Pathology, Haukeland University Hospital, Bergen, Norway
| | - Ottar Nygård
- Centre for Nutrition, Department of Clinical Science, University of Bergen, Bergen, Norway
- Mohn Nutrition Research Laboratory, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Vegard Lysne
- Centre for Nutrition, Department of Clinical Science, University of Bergen, Bergen, Norway
- Mohn Nutrition Research Laboratory, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
33
|
Azarcoya-Barrera J, Goruk S, Lewis ED, Pouliot Y, Curtis JM, Steele R, Wadge E, Field CJ, Jacobs RL, Richard C. Feeding Buttermilk-Derived Choline Forms During Gestation and Lactation Modulates Ex Vivo T-Cell Response in Rat Dams. J Nutr 2020; 150:1958-1965. [PMID: 32271922 DOI: 10.1093/jn/nxaa089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/07/2020] [Accepted: 03/12/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Buttermilk contains a mixture of choline forms; it is high in phosphatidylcholine (PC) and sphingomyelin (SM), which could have an impact on immune system development and function. OBJECTIVES We aimed to determine the effect of feeding buttermilk-derived choline forms during pregnancy and lactation on maternal immune function. METHODS Sprague Dawley dams (n = 8 per diet) were randomly assigned midway through pregnancy (10 d of gestation) to 1 of 3 experimental diets, containing 1.7 g/kg choline: control [100% free choline (FC)]; buttermilk [37% PC, 34% SM, 17% glycerophosphocholine (GPC), 7% FC, 5% phosphocholine]; or placebo (50% PC, 25% FC, 25% GPC). Dams consumed the same diet until the end of the lactation period (21 d after parturition). Cell phenotypes and cytokine production by mitogen-stimulated splenocytes were measured and compared using 1-factor ANOVA test in order to asses the effect of diet on immune fuction of lactating dams (main outcome). RESULTS After ConA stimulation, splenocytes from dams in the buttermilk group produced more IL-2 (30%), TNF-α (30%), and IFN-γ (42%) compared with both the placebo and control diets. Placebo-fed dams had a higher proportion of CD8+ cells expressing CD152+ (22%) in spleen, and splenocytes from dams that were fed the buttermilk and the placebo diets produced about 50% and 53% more IL-10 after LPS and OVA stimulation, respectively, compared with the control group. CONCLUSIONS Feeding buttermilk-derived choline forms during pregnancy and lactation had a beneficial impact on the immune system of Sprague Dawley rat dams, especially on T-cell function.
Collapse
Affiliation(s)
- Jessy Azarcoya-Barrera
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Susan Goruk
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Erin D Lewis
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Yves Pouliot
- STELA Dairy Research Center, Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, Québec, Canada
| | - Jonathan M Curtis
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Reid Steele
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Emily Wadge
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - René L Jacobs
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Caroline Richard
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
34
|
Derbyshire E, Obeid R. Choline, Neurological Development and Brain Function: A Systematic Review Focusing on the First 1000 Days. Nutrients 2020; 12:E1731. [PMID: 32531929 PMCID: PMC7352907 DOI: 10.3390/nu12061731] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/22/2022] Open
Abstract
The foundations of neurodevelopment across an individual's lifespan are established in the first 1000 days of life (2 years). During this period an adequate supply of nutrients are essential for proper neurodevelopment and lifelong brain function. Of these, evidence for choline has been building but has not been widely collated using systematic approaches. Therefore, a systematic review was performed to identify the animal and human studies looking at inter-relationships between choline, neurological development, and brain function during the first 1000 days of life. The database PubMed was used, and reference lists were searched. In total, 813 publications were subject to the title/abstract review, and 38 animal and 16 human studies were included after evaluation. Findings suggest that supplementing the maternal or child's diet with choline over the first 1000 days of life could subsequently: (1) support normal brain development (animal and human evidence), (2) protect against neural and metabolic insults, particularly when the fetus is exposed to alcohol (animal and human evidence), and (3) improve neural and cognitive functioning (animal evidence). Overall, most offspring would benefit from increased choline supply during the first 1000 days of life, particularly in relation to helping facilitate normal brain development. Health policies and guidelines should consider re-evaluation to help communicate and impart potential choline benefits through diet and/or supplementation approaches across this critical life stage.
Collapse
Affiliation(s)
| | - Rima Obeid
- Department of Clinical Chemistry, University Hospital of the Saarland, Building 57, 66424 Homburg, Germany;
| |
Collapse
|
35
|
Hammoud R, Pannia E, Kubant R, Liao CS, Ho M, Yang NV, Chatterjee D, Caudill MA, Malysheva OV, Pausova Z, Anderson GH. Maternal Choline Intake Programs Hypothalamic Energy Regulation and Later-Life Phenotype of Male Wistar Rat Offspring. Mol Nutr Food Res 2020; 64:e1901178. [PMID: 32110848 DOI: 10.1002/mnfr.201901178] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/21/2020] [Indexed: 12/13/2022]
Abstract
SCOPE High-folic-acid diets during pregnancy result in obesity in the offspring, associated with altered DNA-methylation of hypothalamic food intake neurons. Like folic acid, the methyl-donor choline modulates foetal brain development, but its long-term programing effects on energy regulation remain undefined. This study aims to describe the effect of choline intake during pregnancy on offspring phenotype and hypothalamic energy-regulatory mechanisms. METHODS AND RESULTS Wistar rat dams are fed an AIN-93G diet with recommended choline (RC, 1 g kg-1 diet), low choline (LC, 0.5-fold), or high choline (HC, 2.5-fold) during pregnancy. Male pups are terminated at birth and 17 weeks post-weaning. Brain 1-carbon metabolites, body weight, food intake, energy expenditure, plasma hormones, and protein expression of hypothalamic neuropeptides are measured. HC pups have higher expression of the orexigenic neuropeptide-Y neurons at birth, consistent with higher cumulative food intake and body weight gain post-weaning compared to RC and LC offspring. LC pups have lower leptin receptor expression at birth and lower energy expenditure and activity during adulthood. CONCLUSION Choline content of diets that are consumed by rats during pregnancy affects the later-life phenotype of offspring, associated with altered in utero programing of hypothalamic food intake regulation.
Collapse
Affiliation(s)
- Rola Hammoud
- Department of Nutritional Sciences, University of Toronto, 1 King's College Circle, Rm. 5360, Toronto, Ontario, M5S1A8, Canada
| | - Emanuela Pannia
- Department of Nutritional Sciences, University of Toronto, 1 King's College Circle, Rm. 5360, Toronto, Ontario, M5S1A8, Canada
| | - Ruslan Kubant
- Department of Nutritional Sciences, University of Toronto, 1 King's College Circle, Rm. 5360, Toronto, Ontario, M5S1A8, Canada
| | - Chih-Sheng Liao
- Department of Nutritional Sciences, University of Toronto, 1 King's College Circle, Rm. 5360, Toronto, Ontario, M5S1A8, Canada
| | - Mandy Ho
- Department of Nutritional Sciences, University of Toronto, 1 King's College Circle, Rm. 5360, Toronto, Ontario, M5S1A8, Canada
| | - Neil V Yang
- Department of Nutritional Sciences, University of Toronto, 1 King's College Circle, Rm. 5360, Toronto, Ontario, M5S1A8, Canada
| | - Diptendu Chatterjee
- Department of Nutritional Sciences, University of Toronto, 1 King's College Circle, Rm. 5360, Toronto, Ontario, M5S1A8, Canada
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, 228 Savage Hall, Ithaca, NY, 14850, USA
| | - Olga V Malysheva
- Division of Nutritional Sciences, Cornell University, 228 Savage Hall, Ithaca, NY, 14850, USA
| | - Zdenka Pausova
- Department of Nutritional Sciences, University of Toronto, 1 King's College Circle, Rm. 5360, Toronto, Ontario, M5S1A8, Canada
- Department of Physiology, University of Toronto, 1 King's College Circle, Rm. 5360, Toronto, Ontario, M5S1A8, Canada
- Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, 686 Bay Street, Rm. 109705, Toronto, Ontario, M5G0A4, Canada
| | - G Harvey Anderson
- Department of Nutritional Sciences, University of Toronto, 1 King's College Circle, Rm. 5360, Toronto, Ontario, M5S1A8, Canada
- Department of Physiology, University of Toronto, 1 King's College Circle, Rm. 5360, Toronto, Ontario, M5S1A8, Canada
| |
Collapse
|
36
|
Perrin MT, Pawlak R, Allen LH, Hampel D. Total Water-Soluble Choline Concentration Does Not Differ in Milk from Vegan, Vegetarian, and Nonvegetarian Lactating Women. J Nutr 2020; 150:512-517. [PMID: 32133524 DOI: 10.1093/jn/nxz257] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/26/2019] [Accepted: 09/26/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Choline is an essential nutrient for brain growth and other processes in the developing neonate. The impact of a maternal plant-based diet on the choline composition of breast milk is unknown. OBJECTIVE We assessed the water-soluble choline content of milk from lactating women in the United States following 3 dietary patterns: vegan, vegetarian, and nonvegetarian. METHODS We conducted a cross-sectional study of 74 healthy lactating women who provided a single breast-milk sample using a standardized collection protocol. Participants completed a food-frequency screener and were classified as follows: nonvegetarians (NONVEG) consumed meat; vegetarians (VEGT) consumed milk, dairy, and/or fish; and vegans (VEGAN) consumed animal products less than monthly. Primary outcomes measured were the concentration (in milligrams per liter) and distribution (percentage) of choline from the following water-soluble forms: free choline, phosphocholine (PCho), and glycerophosphocholine (GPC). Differences between diet groups were evaluated with ANOVA. RESULTS There was a wide range in breast-milk total water-soluble choline (4-301 mg/L), with no significant difference (P > 0.05) by maternal diet pattern. There were differences in choline forms, with VEGAN having a greater mean ± SD concentration and distribution of choline derived from GPC (62.7 ± 25.3 mg/L) than VEGT (47.7 ± 21.2 mg/L) and NONVEG (42.4 ± 14.9 mg/L) (P = 0.0052). There was a lower mean ± SD percentage of choline from PCho (P = 0.0106) in VEGAN (32.5% ± 18.3%) than in VEGT (46.1% ± 18.3%) and NONVEG (44.8% ± 15.7%). Lactation stage and maternal BMI were significantly associated with some choline forms. CONCLUSIONS There was a wide range of water-soluble choline concentrations in the milk of healthy lactating women following vegan, vegetarian, and nonvegetarian diets, with no observed difference in total water-soluble choline concentration by maternal diet. This suggests that maternal plant-based diet by itself is not a risk factor for low breast-milk choline.
Collapse
Affiliation(s)
- Maryanne T Perrin
- Department of Nutrition, University of North Carolina Greensboro, Greensboro, NC, USA
| | - Roman Pawlak
- Department of Nutrition Science, East Carolina University, Greenville, NC, USA
| | - Lindsay H Allen
- USDA/Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, USA.,Department of Nutrition, University of California-Davis, Davis, CA, USA
| | - Daniela Hampel
- USDA/Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, USA.,Department of Nutrition, University of California-Davis, Davis, CA, USA
| |
Collapse
|
37
|
Moore CJ, Perreault M, Mottola MF, Atkinson SA. Diet in Early Pregnancy: Focus on Folate, Vitamin B12, Vitamin D, and Choline. CAN J DIET PRACT RES 2019; 81:58-65. [PMID: 31512510 DOI: 10.3148/cjdpr-2019-025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Purpose: Prenatal multivitamins are recommended in pregnancy. This study assessed food and supplement intakes of folate, vitamin B12 (B12), vitamin D, and choline in pregnant women living in Southern Ontario in comparison with current recommendations. Methods: Women recruited to the Be Healthy in Pregnancy RCT (NCT01693510) completed 3-day diet/supplement records at 12-17 weeks gestation. Intakes of folate, B12, vitamin D, and choline were quantified and compared with recommendations for pregnant women. Results: Folate intake (median (min, max)) was 1963 μg/day dietary folate equivalents (153, 10 846); 90% of women met the Estimated Average Requirement (EAR) but 77% exceeded the Tolerable Upper Intake Level (UL) (n = 232). B12 intake was 12.1 μg/day (0.3, 2336); 96% of women met the EAR with 7% exceeding the EAR 100-fold (n = 232). Vitamin D intake was 564 IU/day (0.0, 11 062); 83% met the EAR, whereas 1.7% exceeded the UL (n = 232). Choline intake was 338 mg/day (120, 1016); only 18% met the Adequate Intake and none exceeded the UL (n = 158). Conclusion: To meet the nutrient requirements of pregnancy many women rely on prenatal vitamins. Reformulating prenatal multivitamin supplements to provide doses of vitamins within recommendations to complement a balanced healthy diet would ensure appropriate micronutrient intakes for pregnant women.
Collapse
|
38
|
Derbyshire E. Could we be overlooking a potential choline crisis in the United Kingdom? BMJ Nutr Prev Health 2019; 2:86-89. [PMID: 33235962 PMCID: PMC7664488 DOI: 10.1136/bmjnph-2019-000037] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/17/2019] [Accepted: 06/21/2019] [Indexed: 11/18/2022] Open
|
39
|
Probst Y, Guan V, Neale E. Development of a Choline Database to Estimate Australian Population Intakes. Nutrients 2019; 11:nu11040913. [PMID: 31018620 PMCID: PMC6521034 DOI: 10.3390/nu11040913] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/18/2019] [Accepted: 04/22/2019] [Indexed: 11/16/2022] Open
Abstract
The AUSNUT 2011-13 food composition database was expanded to include Australian choline values. The development began with a systematic literature review of published studies. Analytical data from the food studies were extracted and aligned with their equivalent AUSNUT food identification code. Global food composition databases containing choline values were matched to the remaining AUSNUT food codes, following the FAO INFOODS food matching guidelines, including adjustments for moisture and protein composition. Composite foods, and not further-specified foods, were developed using the Food Standards Australia New Zealand (FSANZ) recipe files. The completed choline database was then employed to analyse the Australian National Nutrition and Physical Activity Survey 2011-12, with population and sampling weightings applied. Survey respondents were classified into categories based on their level of choline intake and compared with the Australian Adequate Intake levels. Food sources of intake were also explored. Multiple linear regression models were developed for food group contributors to choline intake. Mean choline intakes varied from 151.50 mg for pregnant 14-18 years old, to 310.54 mg for 19-64 year old males. Less than 10% of the population by age and gender were achieving the Adequate Intake for choline. Eggs and their contributing food groups were the top ranked food sources of choline for the population.
Collapse
Affiliation(s)
- Yasmine Probst
- Smart Foods Centre, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia.
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.
| | - Vivienne Guan
- Smart Foods Centre, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia.
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.
| | - Elizabeth Neale
- Smart Foods Centre, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia.
| |
Collapse
|
40
|
van Lee L, Crozier SR, Aris IM, Tint MT, Sadananthan SA, Michael N, Quah PL, Robinson SM, Inskip HM, Harvey NC, Barker M, Cooper C, Velan SS, Lee YS, Fortier MV, Yap F, Gluckman PD, Tan KH, Shek LP, Chong YS, Godfrey KM, Chong MFF. Prospective associations of maternal choline status with offspring body composition in the first 5 years of life in two large mother-offspring cohorts: the Southampton Women's Survey cohort and the Growing Up in Singapore Towards healthy Outcomes cohort. Int J Epidemiol 2019; 48:433-444. [PMID: 30649331 PMCID: PMC6751083 DOI: 10.1093/ije/dyy291] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Choline status has been positively associated with weight and fat mass in animal and human studies. As evidence examining maternal circulating choline concentrations and offspring body composition in human infants/children is lacking, we investigated this in two cohorts. METHODS Maternal choline concentrations were measured in the UK Southampton Women's Survey (SWS; serum, n = 985, 11 weeks' gestation) and Singapore Growing Up Towards healthy Outcomes (GUSTO); n = 955, 26-28 weeks' gestation) mother-offspring cohorts. Offspring anthropometry was measured at birth and up to age 5 years. Body fat mass was determined using dual-energy x-ray absorptiometry at birth and age 4 years for SWS; and using air-displacement plethysmography at birth and age 5 years for GUSTO. Linear-regression analyses were performed, adjusting for confounders. RESULTS In SWS, higher maternal choline concentrations were associated with higher neonatal total body fat mass {β = 0.60 standard deviation [SD]/5 µmol/L maternal choline [95% confidence interval (CI) 0.04-1.16]} and higher subscapular skinfold thickness [β = 0.55 mm/5 µmol/L (95% CI, 0.12-1.00)] at birth. In GUSTO, higher maternal choline concentrations were associated with higher neonatal body mass index-for-age z-score [β = 0.31 SD/5 µmol/L (0.10-0.51)] and higher triceps [β = 0.38 mm/5 µmol/L (95% CI, 0.11-0.65)] and subscapular skinfold thicknesses [β = 0.26 mm/5 µmol/L (95% CI, 0.01-0.50)] at birth. No consistent trends were observed between maternal choline and offspring gain in body mass index, skinfold thicknesses, abdominal circumference, weight, length/height and adiposity measures in later infancy and early childhood. CONCLUSION Our study provides evidence that maternal circulating choline concentrations during pregnancy are positively associated with offspring BMI, skinfold thicknesses and adiposity at birth, but not with growth and adiposity through infancy and early childhood to the age of 5 years.
Collapse
Affiliation(s)
- Linde van Lee
- Singapore Institute for Clinical Science, Agency for Science, Technology, and Research, Singapore
| | - Sarah R Crozier
- MRC Lifecourse Epidemiology Unit, University of Southampton, United Kingdom
| | - Izzuddin M Aris
- Singapore Institute for Clinical Science, Agency for Science, Technology, and Research, Singapore
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mya T Tint
- Singapore Institute for Clinical Science, Agency for Science, Technology, and Research, Singapore
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Suresh Anand Sadananthan
- Singapore Institute for Clinical Science, Agency for Science, Technology, and Research, Singapore
| | - Navin Michael
- Singapore Institute for Clinical Science, Agency for Science, Technology, and Research, Singapore
| | - Phaik Ling Quah
- Singapore Institute for Clinical Science, Agency for Science, Technology, and Research, Singapore
| | - Sian M Robinson
- MRC Lifecourse Epidemiology Unit, University of Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University of Southampton and Hospital Southampton NHS Foundation Trust, United Kingdom
| | - Hazel M Inskip
- MRC Lifecourse Epidemiology Unit, University of Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University of Southampton and Hospital Southampton NHS Foundation Trust, United Kingdom
| | - Nicholas C Harvey
- MRC Lifecourse Epidemiology Unit, University of Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University of Southampton and Hospital Southampton NHS Foundation Trust, United Kingdom
| | - Mary Barker
- MRC Lifecourse Epidemiology Unit, University of Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University of Southampton and Hospital Southampton NHS Foundation Trust, United Kingdom
| | - Cyrus Cooper
- MRC Lifecourse Epidemiology Unit, University of Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University of Southampton and Hospital Southampton NHS Foundation Trust, United Kingdom
| | - Sendhil S Velan
- Singapore Institute for Clinical Science, Agency for Science, Technology, and Research, Singapore
- Laboratory of Molecular Imaging, Singapore Bioimaging Consortium, Agency for Science Technology and Research, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yung Seng Lee
- Singapore Institute for Clinical Science, Agency for Science, Technology, and Research, Singapore
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore, Singapore
| | - Marielle V Fortier
- Singapore Institute for Clinical Science, Agency for Science, Technology, and Research, Singapore
- Department of Diagnostic and Interventional Imaging, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Fabian Yap
- Duke-NUS Medical School, Singapore, Nanyang Technological University, Singapore, Singapore
- Department of Pediatrics, KK Women’s and Children’s Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Peter D Gluckman
- Singapore Institute for Clinical Science, Agency for Science, Technology, and Research, Singapore
- Liggings Institute, University of Auckland, New Zealand
| | - Kok Hian Tan
- Department of Reproductive Medicine, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Lynette P Shek
- Singapore Institute for Clinical Science, Agency for Science, Technology, and Research, Singapore
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yap-Seng Chong
- Singapore Institute for Clinical Science, Agency for Science, Technology, and Research, Singapore
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Unit, University of Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University of Southampton and Hospital Southampton NHS Foundation Trust, United Kingdom
| | - Mary FF Chong
- Singapore Institute for Clinical Science, Agency for Science, Technology, and Research, Singapore
- Clinical Nutrition Research Centre, Agency for Science, Technology, and Research, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| |
Collapse
|
41
|
Development and validation of a quantitative choline food frequency questionnaire for use with drinking and non-drinking pregnant women in Cape Town, South Africa. Nutr J 2018; 17:108. [PMID: 30466439 PMCID: PMC6251124 DOI: 10.1186/s12937-018-0411-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/24/2018] [Indexed: 12/27/2022] Open
Abstract
Background Although animal and human studies have demonstrated interactions between dietary choline and fetal alcohol spectrum disorders, dietary choline deficiency in pregnancy is common in the US and worldwide. We sought to develop and validate a quantitative food frequency questionnaire (QFFQ) to estimate usual daily choline intake in pregnant mothers. Methods A panel of nutrition experts developed a Choline-QFFQ food item list, including sources with high choline content and the most commonly consumed choline-containing foods in the target population. A data base for choline content of each item was compiled. For reliability and validity testing in a prospective longitudinal cohort, 123 heavy drinking Cape Coloured pregnant women and 83 abstaining/light-drinking controls were recruited at their first antenatal clinic visit. At 3 prenatal study visits, each gravida was interviewed about alcohol, smoking, and drug use, and administered a 24-hour recall interview and the Choline-QFFQ. Results Across all visits and assessments, > 78% of heavy drinkers and controls reported choline intake below the Dietary Reference Intakes adequate intake level (450 mg/day). Women reported a decrease in choline intake over time on the QFFQ. Reliability of the QFFQ across visits was good-to-acceptable for 2 of 4 group-level tests and 4 of 5 individual-level tests for both drinkers and controls. When compared with 24-hr recall data, validity of the QFFQ was good-to-acceptable for 3 of 4 individual-level tests and 3 of 5 group-level tests. For controls, validity was good-to-acceptable for all 4 individual-level tests and all 5 group-level tests. Conclusions To our knowledge, this is the first quantitative choline food frequency screening questionnaire to be developed and validated for use with both heavy and non-drinking pregnant women and the first to be used in the Cape Coloured community in South Africa. Given the high prevalence of inadequate choline intake and the growing evidence that maternal choline supplementation can mitigate some of the adverse effects of prenatal alcohol exposure, this tool may be useful for both research and future clinical outreach programs.
Collapse
|
42
|
Dietary Choline Intake: Current State of Knowledge Across the Life Cycle. Nutrients 2018; 10:nu10101513. [PMID: 30332744 PMCID: PMC6213596 DOI: 10.3390/nu10101513] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/10/2018] [Accepted: 10/13/2018] [Indexed: 12/13/2022] Open
Abstract
Choline, an essential dietary nutrient for humans, is required for the synthesis of the neurotransmitter, acetylcholine, the methyl group donor, betaine, and phospholipids; and therefore, choline is involved in a broad range of critical physiological functions across all stages of the life cycle. The current dietary recommendations for choline have been established as Adequate Intakes (AIs) for total choline; however, dietary choline is present in multiple different forms that are both water-soluble (e.g., free choline, phosphocholine, and glycerophosphocholine) and lipid-soluble (e.g., phosphatidylcholine and sphingomyelin). Interestingly, the different dietary choline forms consumed during infancy differ from those in adulthood. This can be explained by the primary food source, where the majority of choline present in human milk is in the water-soluble form, versus lipid-soluble forms for foods consumed later on. This review summarizes the current knowledge on dietary recommendations and assessment methods, and dietary choline intake from food sources across the life cycle.
Collapse
|
43
|
Zia Y, Al Rajabi A, Mi S, Ju T, Leonard KA, Nelson R, Thiesen A, Willing BP, Field CJ, Curtis JM, van der Veen JN, Jacobs RL. Hepatic Expression of PEMT, but Not Dietary Choline Supplementation, Reverses the Protection against Atherosclerosis in Pemt-/-/Ldlr-/- Mice. J Nutr 2018; 148:1513-1520. [PMID: 30281112 DOI: 10.1093/jn/nxy165] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/03/2018] [Indexed: 01/11/2023] Open
Abstract
Background Phosphatidylethanolamine N-methyltransferase (PEMT) converts phosphatidylethanolamine to phosphatidylcholine. Pemt-/-/low density lipoprotein receptor (Ldlr)-/- mice have significantly reduced plasma lipids and are protected against atherosclerosis. Recent studies have shown that choline can be metabolized by the gut flora into trimethylamine-N-oxide (TMAO), which is an emerging risk factor for atherosclerosis. Objective The objective of this study was to determine whether ectopic hepatic PEMT expression or choline supplementation would promote atherosclerosis in Pemt-/-/Ldlr-/- mice. Methods Male 8- to 10-wk-old Pemt+/+/Ldlr-/- (SKO) and Pemt-/-/Ldlr-/- (DKO) mice were injected with an adeno-associated virus (AAV) expressing green fluorescent protein (GFP) or human PEMT and fed a Western diet (40% of calories from fat, 0.5% cholesterol) for 8 wk. In a separate experiment, 8- to 10-wk-old SKO and half of the DKO male mice were fed a Western diet with normal (3 g/kg) choline for 12 wk. The remaining DKO mice [choline-supplemented (CS) DKO] were fed a CS Western diet (10 g choline/kg). Plasma lipid concentrations, choline metabolites, and aortic atherosclerosis were measured. Results Plasma cholesterol, plasma TMAO, and aortic atherosclerosis were reduced by 60%, 40%, and 80%, respectively, in DKO mice compared with SKO mice. AAV-PEMT administration increased plasma cholesterol and TMAO by 30% and 40%, respectively, in DKO mice compared with AAV-GFP-treated DKO mice. Furthermore, AAV-PEMT-injected DKO mice developed atherosclerotic lesions similar to SKO mice. In the second study, there was no difference in atherosclerosis or plasma cholesterol between DKO and CS-DKO mice. However, plasma TMAO concentrations were increased 2.5-fold in CS-DKO mice compared with DKO mice. Conclusions Reintroducing hepatic PEMT reversed the atheroprotective phenotype of DKO mice. Choline supplementation did not increase atherosclerosis or plasma cholesterol in DKO mice. Our data suggest that plasma TMAO does not induce atherosclerosis when plasma cholesterol is low. Furthermore, this is the first report to our knowledge that suggests that de novo choline synthesis alters TMAO status.
Collapse
Affiliation(s)
- Yumna Zia
- Departments of Agricultural, Food, and Nutritional Science
| | - Ala Al Rajabi
- Departments of Agricultural, Food, and Nutritional Science
| | - Si Mi
- Departments of Agricultural, Food, and Nutritional Science
| | - Tingting Ju
- Departments of Agricultural, Food, and Nutritional Science
| | | | | | - Aducio Thiesen
- Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | - René L Jacobs
- Departments of Agricultural, Food, and Nutritional Science.,Biochemistry
| |
Collapse
|
44
|
Conrad Z, Johnson LK, Roemmich JN, Juan W, Jahns L. Nutrient intake disparities in the US: modeling the effect of food substitutions. Nutr J 2018; 17:53. [PMID: 29776365 PMCID: PMC5960152 DOI: 10.1186/s12937-018-0360-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 05/01/2018] [Indexed: 01/30/2023] Open
Abstract
Background Diet quality among federal food assistance program participants remains low, and little research has assessed the diet quality of food insecure non-participants. Further research is needed to assess the extent to which food substitutions can improve the nutritional status of these vulnerable populations. Substituting egg dishes for other commonly consumed dishes at certain eating occasions may be an effective strategy for improving the daily nutrient intake among these groups. Eggs are rich in many important nutrients, and are low-cost and part of a wide range of cultural food menus, which are important considerations for low-income and ethnically diverse populations. To help guide the focus of targeted nutrition interventions and education campaigns for vulnerable populations, the present work begins by 1) estimating the prevalence of nutrient inadequacy among these groups, and then models the effect of consuming egg dishes instead of commonly consumed dishes at each eating occasion on 2) the prevalence of nutrient inadequacy, and 3) the mean intake of nutrients. Methods Dietary data from 34,741 adults ≥ 20 y were acquired from the National Health and Nutrition Examination Survey, 2001–2014. Diet pattern modeling was used to substitute commonly consumed egg dishes for commonly consumed main dishes at breakfast, lunch, and dinner. National Cancer Institute usual intake methods were used to estimate the prevalence of inadequate intake of 31 nutrients pre- and post-substitution, and a novel index was used to estimate change in intake of all nutrients collectively. Results Substituting eggs for commonly consumed main dishes at lunch or dinner did not change total daily nutrient intake for each group (P > 0.05), but decreased the prevalence of vitamin D inadequacy by 1–4 percentage points (P < 0.01). Substituting eggs for commonly consumed foods at breakfast increased the prevalence of folate inadequacy by 8–12 percentage points among each group (P < 0.01). Conclusions When making food substitutions to increase nutrient intake, eating occasion should be an important consideration. Further research is needed to better understand how food substitutions affect diet costs, which may be an important driver of food purchasing decisions among low income individuals with limited food budgets. Electronic supplementary material The online version of this article (10.1186/s12937-018-0360-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zach Conrad
- Grand Forks Human Nutrition Research Center, US Department of Agriculture, Agricultural Research Service, 2420 2nd Ave. N, Grand Forks, ND, 58203, USA.
| | - LuAnn K Johnson
- Grand Forks Human Nutrition Research Center, US Department of Agriculture, Agricultural Research Service, 2420 2nd Ave. N, Grand Forks, ND, 58203, USA
| | - James N Roemmich
- Grand Forks Human Nutrition Research Center, US Department of Agriculture, Agricultural Research Service, 2420 2nd Ave. N, Grand Forks, ND, 58203, USA
| | - WenYen Juan
- Center for Food Safety and Applied Nutrition, US Department of Health and Human Services, Food and Drug Administration, 5001 Campus Drive, College Park, MD, 20740, USA
| | - Lisa Jahns
- Grand Forks Human Nutrition Research Center, US Department of Agriculture, Agricultural Research Service, 2420 2nd Ave. N, Grand Forks, ND, 58203, USA
| |
Collapse
|
45
|
Concentrations of Water-Soluble Forms of Choline in Human Milk from Lactating Women in Canada and Cambodia. Nutrients 2018; 10:nu10030381. [PMID: 29558412 PMCID: PMC5872799 DOI: 10.3390/nu10030381] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/12/2018] [Accepted: 03/16/2018] [Indexed: 02/07/2023] Open
Abstract
Choline has critical roles during periods of rapid growth and development, such as infancy. In human milk, choline is mostly present in water-soluble forms (free choline, phosphocholine, and glycerophosphocholine). It is thought that milk choline concentration is influenced by maternal choline intake, and the richest food sources for choline are of animal origin. Scarce information exists on milk choline from countries differing in animal-source food availability. In this secondary analysis of samples from previous trials, the concentrations of the water-soluble forms of choline were quantified by liquid chromatography-tandem mass spectrometry in mature milk samples collected from lactating women in Canada (n = 301) and in Cambodia (n = 67). None of the water-soluble forms of choline concentrations in milk differed between Canada and Cambodia. For all milk samples (n = 368), free choline, phosphocholine, glycerophosphocholine, and the sum of water-soluble forms of choline concentrations in milk were (mean (95%CI)) 151 (141, 160, 540 (519, 562), 411 (396, 427), and 1102 (1072, 1133) µmol/L, respectively. Theoretically, only 19% of infants would meet the current Adequate Intake (AI) for choline. Our findings suggest that the concentrations in milk of water-soluble forms of choline are similar in Canada and Cambodia, and that the concentration used to set the infant AI might be inaccurate.
Collapse
|
46
|
McGee M, Bainbridge S, Fontaine-Bisson B. A crucial role for maternal dietary methyl donor intake in epigenetic programming and fetal growth outcomes. Nutr Rev 2018. [DOI: 10.1093/nutrit/nuy006] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Meghan McGee
- Department of Nutritional Sciences, University of Toronto, and Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Shannon Bainbridge
- Interdisciplinary School of Health Sciences and the Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Bénédicte Fontaine-Bisson
- School of Nutrition Sciences, University of Ottawa, and the Institut du savoir Montfort, Ottawa, Ontario, Canada
| |
Collapse
|
47
|
Adherence to Diabetes Dietary Guidelines Assessed Using a Validated Questionnaire Predicts Glucose Control in Adults With Type 2 Diabetes. Can J Diabetes 2018. [DOI: 10.1016/j.jcjd.2017.04.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
48
|
Caudill MA, Strupp BJ, Muscalu L, Nevins JEH, Canfield RL. Maternal choline supplementation during the third trimester of pregnancy improves infant information processing speed: a randomized, double-blind, controlled feeding study. FASEB J 2018; 32:2172-2180. [PMID: 29217669 DOI: 10.1096/fj.201700692rr] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Rodent studies demonstrate that supplementing the maternal diet with choline during pregnancy produces life-long cognitive benefits for the offspring. In contrast, the two experimental studies examining cognitive effects of maternal choline supplementation in humans produced inconsistent results, perhaps because of poor participant adherence and/or uncontrolled variation in intake of choline or other nutrients. We examined the effects of maternal choline supplementation during pregnancy on infant cognition, with intake of choline and other nutrients tightly controlled. Women entering their third trimester were randomized to consume, until delivery, either 480 mg choline/d ( n = 13) or 930 mg choline/d ( n = 13). Infant information processing speed and visuospatial memory were tested at 4, 7, 10, and 13 mo of age ( n = 24). Mean reaction time averaged across the four ages was significantly faster for infants born to mothers in the 930 ( vs. 480) mg choline/d group. This result indicates that maternal consumption of approximately twice the recommended amount of choline during the last trimester improves infant information processing speed. Furthermore, for the 480-mg choline/d group, there was a significant linear effect of exposure duration (infants exposed longer showed faster reaction times), suggesting that even modest increases in maternal choline intake during pregnancy may produce cognitive benefits for offspring.-Caudill, M. A., Strupp, B. J., Muscalu, L., Nevins, J. E. H., Canfield, R. L. Maternal choline supplementation during the third trimester of pregnancy improves infant information processing speed: a randomized, double-blind, controlled feeding study.
Collapse
Affiliation(s)
- Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | - Barbara J Strupp
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA.,Department of Psychology, Cornell University, Ithaca, New York, USA; and
| | - Laura Muscalu
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA.,Department of Psychology, Ithaca College, Ithaca, New York, USA
| | - Julie E H Nevins
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | - Richard L Canfield
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| |
Collapse
|
49
|
Choline Intake and Its Food Sources in the Diet of Romanian Kindergarten Children. Nutrients 2017; 9:nu9080896. [PMID: 28820445 PMCID: PMC5579689 DOI: 10.3390/nu9080896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/12/2017] [Accepted: 08/15/2017] [Indexed: 11/25/2022] Open
Abstract
The objective of this study is to assess the usual intake and food sources of choline in a group of Romanian kindergarten children. A cross-sectional study was performed among 71 children aged 4–6 years from four kindergartens from Cluj-Napoca, Romania. Dietary intake data were collected by means of three-day food records. The mean (SD) daily intake of choline was 215 (32) mg/day, 22.5% of the participants fulfilling the adequate intake (AI) for children 4–6 years of age of 250 mg of choline per day. The main food sources were meat (mainly poultry), eggs, grains, cereals, and baked products (mainly bread), and dairy products (mainly milk). The results of the logistic regression analyses show that an appropriate consumption of choline/day was statistically significantly associated with the consumption of at least one egg per three days (OR = 7.5, p < 0.05), a minimum of two portions of milk or dairy products per day (500 mL milk or yoghurt, or 60 g of cheese/day) (OR = 4.4, p < 0.05), and at least one portion of meat/day (90 g/day) (OR = 14.4, p < 0.05). The results underline the need for future surveys in this field, as well as actions to encourage an appropriate diet for children, including an appropriate content of choline.
Collapse
|
50
|
Usual Choline Intakes Are Associated with Egg and Protein Food Consumption in the United States. Nutrients 2017; 9:nu9080839. [PMID: 28783055 PMCID: PMC5579632 DOI: 10.3390/nu9080839] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 07/19/2017] [Accepted: 08/02/2017] [Indexed: 12/02/2022] Open
Abstract
Choline is an essential nutrient with critical roles in several biological processes including neuronal development, cell signaling, nerve impulse transmission, and lipid transport and metabolism. The National Cancer Institute method was used to assess usual intakes of choline from foods according to data for participants enrolled in the National Health and Nutrition Examination Survey 2009–2014 datasets and pregnant women in the 2005–2014 datasets. Suboptimal intakes of choline are present across many gender and life-stage subpopulations, as well as pregnant women in the U.S. Only 8.03 ± 0.56% of adults and 8.51 ± 2.89% pregnant women meet the AI for choline. Children 2–3 years were the most likely to meet their gender and life-stage specific AI, followed by children 4–8 years. Adults 19+ years who consume eggs were more likely to meet their gender and life-stage AI as compared to non-consumers (57.3 ± 1.45% and 2.43 ± 0.28%). Consumers of eggs had almost double the usual intake of choline as compared to non-consumers (525 ± 5.17 mg/d and 294 ± 1.98; p < 0.0001). Protein food (meat, poultry and seafood) consumption also increased usual choline intakes compared to non-consumers (345 ± 2.21 mg/day and 235 ± 8.81; p < 0.0001) to a lesser degree, but did not result in substantial increases in the percent of individuals meeting the AI. No subpopulation exceeded the UL for choline. This research illustrates that it is extremely difficult to achieve the AI for choline without consuming eggs or taking a dietary supplement.
Collapse
|