1
|
Guo W, Li X, Qin K, Zhang P, He J, Liu Y, Yang X, Wu S. Nanopore sequencing demonstrates the roles of spermatozoal DNA N6-methyladenine in mediating transgenerational lipid metabolism disorder induced by excessive folate consumpton. Poult Sci 2024; 103:103953. [PMID: 38945000 PMCID: PMC11267017 DOI: 10.1016/j.psj.2024.103953] [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: 02/07/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 07/02/2024] Open
Abstract
Increased consumption of folic acid is prevalent due to its beneficial effects, but growing evidence emphasizes the side effects pointing to excessive dietary folate intake. The effects of excessive paternal folic acid consumption on offspring and its transgenerational inheritance mechanism have not been elucidated. We hypothesize that excessive folic acid consumption will alter sperm DNA N6-methyladenine (6mA) and 5-methylcytosine (5mC) methylation and heritably influence offspring metabolic homeostasis. Here, we fed roosters either folic acid-control or folic acid-excess diet throughout life. Paternal chronic folic acid excessive supplementation increased hepatic lipogenesis and lipid accumulation but reduced lipolysis both in the roosters and their offspring, which was further confirmed to be induced by one-carbon metabolism inhibition and gene expression alteration associated with the Peroxisome proliferator-activated receptor pathway. Based on the spermatozoal genome-wide DNA methylome identified by Nanopore sequencing, multi-omics association analysis of spermatozoal and hepatic DNA methylome, transcriptome, and metabolome suggested that differential spermatozoal DNA 6mA and 5mC methylation could be involved in regulating lipid metabolism-related gene expression in offspring chickens. This model suggests that sperm DNA N6-methyladenine and 5-methylcytosine methylation were involved in epigenetic transmission and that paternal dietary excess folic acid leads to hepatic lipid accumulation in offspring.
Collapse
Affiliation(s)
- Wei Guo
- Jiangsu Institute of Poultry Science, Yangzhou, Jiangsu Province, 225125, China; College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xinyi Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China; Department of Medicine, Karolinska Institutet, Solna, Stockholm, 17165, Sweden
| | - Kailong Qin
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Peilin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jinhui He
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Shengru Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China; Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17165, Sweden.
| |
Collapse
|
2
|
Wang F, Wang X, Wang J, Liu B. Associations of single and multiple vitamin exposure with childhood eczema: data from the national health and nutrition examination survey. Front Pediatr 2024; 12:1328592. [PMID: 38813547 PMCID: PMC11133564 DOI: 10.3389/fped.2024.1328592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 04/29/2024] [Indexed: 05/31/2024] Open
Abstract
Aim The association between vitamins and eczema has garnered attention, yet few studies have evaluated the effects of co-exposure to multiple vitamins on this condition. This study aims to assess the association of vitamin mixtures with eczema in children. Methods This cross-sectional study analyzed data from 2,244 children aged 6-17 years from the National Health and Nutrition Examination Surveys. Eczema served as the primary outcome. Six serum vitamins, namely, vitamins A, B6, B12, C, D, and E, were the main variables. Weighted multivariate logistic regression was adopted to analyze the association between each serum vitamin and eczema. Odds ratios (OR) with a 95% confidence interval (CI) were calculated. Bayesian kernel machine regression (BKMR) analysis and the quantile g-computation (qgcomp) model were used to evaluate the association of co-exposure to multiple vitamins with eczema. Results In total, 10.83% of children (n = 243) developed eczema. After adjusting for confounding factors, we observed that compared with the reference group (vitamin B12 with second quartile), the OR for eczema was 0.604 (95% CI: 0.373-0.978, P = 0.041) for the first quartile of vitamin B12. Both BKMR analysis and the qgcomp model consistently showed that co-exposure to the six vitamins was positively correlated with the risk of eczema, with vitamin B6 contributing most to the overall effect. In BKMR analyses, we observed an interaction between vitamins B6 and B12 concerning eczema risk. Conclusion Co-exposure to vitamins A, C, B6, B12, D, and E was found to be associated with an increased risk of eczema in children, with vitamin B6 as the greatest positive contributor driving the overall effect.
Collapse
Affiliation(s)
- Fang Wang
- Department of Dermatology, Naval Hospital of Eastern Theater of PLA, Zhoushan, Zhejiang, China
| | - Xiaolie Wang
- Department of Dermatology, Naval Hospital of Eastern Theater of PLA, Zhoushan, Zhejiang, China
| | - Jiayan Wang
- Department of Emergency, Naval Hospital of Eastern Theater of PLA, Zhoushan, Zhejiang, China
| | - Biqing Liu
- Department of Dermatology, Naval Hospital of Eastern Theater of PLA, Zhoushan, Zhejiang, China
| |
Collapse
|
3
|
Gurugubelli KR, Ballambattu VB. Perspectives on folate with special reference to epigenetics and neural tube defects. Reprod Toxicol 2024; 125:108576. [PMID: 38479591 DOI: 10.1016/j.reprotox.2024.108576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/07/2024] [Accepted: 03/10/2024] [Indexed: 03/22/2024]
Abstract
Folate is a micronutrient essential for DNA synthesis, cell division, fetal growth and development. Folate deficiency leads to genomic instability. Inadequate intake of folate during conception may lead to neural tube defects (NTDs) in the offspring. Folate influences the DNA methylation, histone methylation and homocysteine mediated gene methylation. DNA methylation influences the expression of microRNAs (miRNAs). Folate deficiency may be associated with miRNAs misregulation leading to NTDs. Mitochondrial epigenetics and folate metabolism has proved to be involved in embryogenesis and neural tube development. Folate related genetic variants also cause the occurrence of NTDs. Unmetabolized excessive folate may affect health adversely. Hence estimation of folate levels in the blood plays an important role in high-risk cases.
Collapse
Affiliation(s)
- Krishna Rao Gurugubelli
- Department of Biochemistry, Andhra Medical College (AMC), Visakhapatnam, Andhra Pradesh, India
| | - Vishnu Bhat Ballambattu
- Aarupadai Veedu Medical College & Hospital (AVMC & H), Vinayaka Mission's Research Foundation (DU), Kirumambakkam, Puducherry, India.
| |
Collapse
|
4
|
Gallo LA, Steane SE, Young SL, de Jersey S, Schoenaker DAJM, Borg DJ, Lockett J, Collins CE, Perkins AV, Kumar S, Clifton VL, Wilkinson SA. Dietary supplements, guideline alignment and biochemical nutrient status in pregnancy: Findings from the Queensland Family Cohort pilot study. MATERNAL & CHILD NUTRITION 2024; 20:e13589. [PMID: 37947159 PMCID: PMC10750014 DOI: 10.1111/mcn.13589] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/16/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
In high-income nations, multiple micronutrient (MMN) supplementation during pregnancy is a common practice. We aimed to describe maternal characteristics associated with supplement use and daily dose of supplemental nutrients consumed in pregnancy, and whether guideline alignment and nutrient status are related to supplement use. The Queensland Family Cohort is a prospective, Australian observational longitudinal study. Maternal characteristics, nutrient intake from food and supplements, and biochemical nutrient status were assessed in the second trimester (n = 127). Supplement use was reported by 89% of participants, of whom 91% reported taking an MMN supplement. Participants who received private obstetric care, had private health insurance and had greater alignment to meat/vegetarian alternatives recommendations were more likely to report MMN supplement use. Private obstetric care and general practitioner shared care were associated with higher daily dose of supplemental nutrients consumed compared with midwifery group practice. There was high reliance on supplements to meet nutrient reference values for folate, iodine and iron, but only plasma folate concentrations were higher in MMN supplement versus nonsupplement users. Exceeding the upper level of intake for folic acid and iron was more likely among combined MMN and individual supplement/s users, and associated with higher plasma concentrations of the respective nutrients. Given the low alignment with food group recommendations and potential risks associated with high MMN supplement use, whole food diets should be emphasized. This study confirms the need to define effective strategies for optimizing nutrient intake in pregnancy, especially among those most vulnerable where MMN supplement use may be appropriate.
Collapse
Affiliation(s)
- Linda A. Gallo
- School of HealthUniversity of the Sunshine CoastPetrieQLDAustralia
- School of Biomedical SciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Sarah E. Steane
- School of Biomedical SciencesThe University of QueenslandSt LuciaQLDAustralia
- Mater Research InstituteThe University of QueenslandSouth BrisbaneQLDAustralia
| | - Sophia L. Young
- School of HealthUniversity of the Sunshine CoastPetrieQLDAustralia
| | - Susan de Jersey
- Department of Dietetics and Foodservices, Royal Brisbane and Women's HospitalMetro North Hospital and Health ServiceBrisbaneQLDAustralia
- Centre for Health Services ResearchThe University of QueenslandHerstonQLDAustralia
| | - Danielle A. J. M. Schoenaker
- School of Human Development and Health, Faculty of MedicineUniversity of SouthamptonSouthamptonUK
- MRC Lifecourse Epidemiology CentreUniversity of SouthamptonSouthamptonUK
- NIHR Southampton Biomedical Research CentreUniversity Hospital Southampton NHSSouthamptonUK
| | - Danielle J. Borg
- Mater Research InstituteThe University of QueenslandSouth BrisbaneQLDAustralia
| | - Jack Lockett
- Mater Research InstituteThe University of QueenslandSouth BrisbaneQLDAustralia
| | - Clare E. Collins
- School of Health Sciences, College of Health, Medicine and WellbeingUniversity of NewcastleCallaghanNSWAustralia
- Hunter Medical Research InstituteNew Lambton HeightsNSWAustralia
| | | | - Sailesh Kumar
- Mater Research InstituteThe University of QueenslandSouth BrisbaneQLDAustralia
| | - Vicki L. Clifton
- Mater Research InstituteThe University of QueenslandSouth BrisbaneQLDAustralia
| | - Shelley A. Wilkinson
- School of PharmacyThe University of QueenslandSt LuciaQLDAustralia
- Department of Obstetric MedicineMater Mothers HospitalSouth BrisbaneQLDAustralia
| |
Collapse
|
5
|
Fardous AM, Heydari AR. Uncovering the Hidden Dangers and Molecular Mechanisms of Excess Folate: A Narrative Review. Nutrients 2023; 15:4699. [PMID: 37960352 PMCID: PMC10648405 DOI: 10.3390/nu15214699] [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: 09/29/2023] [Revised: 10/26/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
This review delves into the intricate relationship between excess folate (vitamin B9) intake, especially its synthetic form, namely, folic acid, and its implications on health and disease. While folate plays a pivotal role in the one-carbon cycle, which is essential for DNA synthesis, repair, and methylation, concerns arise about its excessive intake. The literature underscores potential deleterious effects, such as an increased risk of carcinogenesis; disruption in DNA methylation; and impacts on embryogenesis, pregnancy outcomes, neurodevelopment, and disease risk. Notably, these consequences stretch beyond the immediate effects, potentially influencing future generations through epigenetic reprogramming. The molecular mechanisms underlying these effects were examined, including altered one-carbon metabolism, the accumulation of unmetabolized folic acid, vitamin-B12-dependent mechanisms, altered methylation patterns, and interactions with critical receptors and signaling pathways. Furthermore, differences in the effects and mechanisms mediated by folic acid compared with natural folate are highlighted. Given the widespread folic acid supplementation, it is imperative to further research its optimal intake levels and the molecular pathways impacted by its excessive intake, ensuring the health and well-being of the global population.
Collapse
Affiliation(s)
- Ali M. Fardous
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA;
| | - Ahmad R. Heydari
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA;
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
6
|
EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA Panel), Turck D, Bohn T, Castenmiller J, de Henauw S, Hirsch‐Ernst K, Knutsen HK, Maciuk A, Mangelsdorf I, McArdle HJ, Pentieva K, Siani A, Thies F, Tsabouri S, Vinceti M, Crous‐Bou M, Molloy A, Ciccolallo L, de Sesmaisons Lecarré A, Fabiani L, Horvath Z, Karavasiloglou N, Naska A. Scientific opinion on the tolerable upper intake level for folate. EFSA J 2023; 21:e08353. [PMID: 37965303 PMCID: PMC10641704 DOI: 10.2903/j.efsa.2023.8353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023] Open
Abstract
Following a request from the European Commission (EC), the EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA) was asked to deliver a scientific opinion on the revision of the tolerable upper intake level (UL) for folic acid/folate. Systematic reviews of the literature were conducted to assess evidence on priority adverse health effects of excess intake of folate (including folic acid and the other authorised forms, (6S)-5-methyltetrahydrofolic acid glucosamine and l-5-methyltetrahydrofolic acid calcium salts), namely risk of cobalamin-dependent neuropathy, cognitive decline among people with low cobalamin status, and colorectal cancer and prostate cancer. The evidence is insufficient to conclude on a positive and causal relationship between the dietary intake of folate and impaired cognitive function, risk of colorectal and prostate cancer. The risk of progression of neurological symptoms in cobalamin-deficient patients is considered as the critical effect to establish an UL for folic acid. No new evidence has been published that could improve the characterisation of the dose-response between folic acid intake and resolution of megaloblastic anaemia in cobalamin-deficient individuals. The ULs for folic acid previously established by the Scientific Committee on Food are retained for all population groups, i.e. 1000 μg/day for adults, including pregnant and lactating women, 200 μg/day for children aged 1-3 years, 300 μg/day for 4-6 years, 400 μg/day for 7-10 years, 600 μg/day for 11-14 years and 800 μg/day for 15-17 years. A UL of 200 μg/day is established for infants aged 4-11 months. The ULs apply to the combined intake of folic acid, (6S)-5-methyltetrahydrofolic acid glucosamine and l-5-methyltetrahydrofolic acid calcium salts, under their authorised conditions of use. It is unlikely that the ULs for supplemental folate are exceeded in European populations, except for regular users of food supplements containing high doses of folic acid/5-methyl-tetrahydrofolic acid salts.
Collapse
|
7
|
Xu C, Du L, Guo Y, Liang Y. TCN1 Expression Is Increased in Asthma. Int Arch Allergy Immunol 2023; 184:1135-1142. [PMID: 37586352 DOI: 10.1159/000531073] [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: 11/06/2022] [Accepted: 04/19/2023] [Indexed: 08/18/2023] Open
Abstract
INTRODUCTION Asthma is a chronic disease that affects populations worldwide. The purpose of this study was to investigate the expression of TCN1 in sputum and its correlation with inflammation and lung function in asthma. METHODS We recruited 141 subjects, detected TCN1 mRNA level by quantitative reverse transcription polymerase chain reaction, detected TCN1 protein expression by Western blot, detected TCN1 protein level by enzyme-linked immunosorbent assay, and analyzed the correlation between TCN1 and fraction of exhaled nitric oxide (FeNO), IgE, EOS%, lung functions, and some Th2 cytokines. The diagnostic value of TCN1 was evaluated by receiver operating characteristics curve. The expression of TCN1 was further confirmed by human bronchial epithelial cell in vitro. RESULTS Compared with the health group, the expression of TCN1 in induced sputum cells increased in asthma group and was correlated with FeNO, IgE, and EOS%. TCN1 level was also elevated in the induced sputum supernatant of asthma patients. The protein level of TCN1 in induced sputum supernatant was correlated with FeNO, IgE and PC-20, forced expiratory volume in the first second (FEV1)%pred, FEV1/FVC, and some cytokines (IL-4, IL-5, IL-10, IL-13, MUC5AC). TCN1 was also differentially expressed in patients with different severity of asthma. Four weeks after ICS treatment, the expression of TCN1 in induced sputum supernatant increased. In vitro, the protein level of TCN1 in human bronchial epithelial cells' supernatant increased after stimulated with IL-4 and IL-13. CONCLUSION The expression of TCN1 was increased in asthma patients' sputum, and was positively correlated with some inflammatory markers, negatively correlated with lung function. TCN1 may be used as a potential biomarker for the diagnosis and treatment of asthma.
Collapse
Affiliation(s)
- Changyi Xu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangdong, China
- Institute of Respiratory Diseases of Sun Yat-Sen University, Guangdong, China
- Department of Clinical Laboratory, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lijuan Du
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangdong, China
- Institute of Respiratory Diseases of Sun Yat-Sen University, Guangdong, China
| | - Yubiao Guo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangdong, China
- Institute of Respiratory Diseases of Sun Yat-Sen University, Guangdong, China
| | - Yuxia Liang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangdong, China
- Institute of Respiratory Diseases of Sun Yat-Sen University, Guangdong, China
| |
Collapse
|
8
|
Ter Borg S, Koopman N, Verkaik-Kloosterman J. An Evaluation of Food and Nutrient Intake among Pregnant Women in The Netherlands: A Systematic Review. Nutrients 2023; 15:3071. [PMID: 37447397 DOI: 10.3390/nu15133071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Nutritional deficiencies during pregnancy can have serious consequences for the health of the (unborn) child. This systematic review provides an updated overview of the available food and nutrient intake data for pregnant women in The Netherlands and an evaluation based on the current recommendations. Embase, MEDLINE, and national institute databases were used. Articles were selected if they had been published since 2008 and contained data on food consumption, nutrient intake, or the status of healthy pregnant women. A qualitative comparison was made with the 2021 Dutch Health Council recommendations and reference values. A total of 218 reports were included, representing 54 individual studies. Dietary assessments were primarily performed via food frequency questionnaires. Protein, vitamin A, thiamin, riboflavin, vitamin B6, folate, vitamin B12, vitamin C, iron, calcium, and magnesium intakes seemed to be adequate. For folate and vitamin D, supplements were needed to reach the recommended intake. The reasons for concern are the low intakes of fruits, vegetables, and (fatty) fish, and the intakes of alcohol, sugary drinks, and salt. For several foods and nutrients, no or limited intake data were found. High-quality, representative, and recent data are needed to evaluate the nutrient intake of pregnant women in order to make accurate assessments and evaluations, supporting scientific-based advice and national nutritional policies.
Collapse
Affiliation(s)
- Sovianne Ter Borg
- National Institute for Public Health and the Environment, 3721 BA Bilthoven, The Netherlands
| | - Nynke Koopman
- National Institute for Public Health and the Environment, 3721 BA Bilthoven, The Netherlands
| | | |
Collapse
|
9
|
Zajac D, Wojciechowski P. The Role of Vitamins in the Pathogenesis of Asthma. Int J Mol Sci 2023; 24:ijms24108574. [PMID: 37239921 DOI: 10.3390/ijms24108574] [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: 02/28/2023] [Revised: 04/27/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Vitamins play a crucial role in the proper functioning of organisms. Disturbances of their levels, seen as deficiency or excess, enhance the development of various diseases, including those of the cardiovascular, immune, or respiratory systems. The present paper aims to summarize the role of vitamins in one of the most common diseases of the respiratory system, asthma. This narrative review describes the influence of vitamins on asthma and its main symptoms such as bronchial hyperreactivity, airway inflammation, oxidative stress, and airway remodeling, as well as the correlation between vitamin intake and levels and the risk of asthma in both pre- and postnatal life.
Collapse
Affiliation(s)
- Dominika Zajac
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warszawa, Poland
| | - Piotr Wojciechowski
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warszawa, Poland
| |
Collapse
|
10
|
Schedel M, Leach SM, Strand MJ, Danhorn T, MacBeth M, Faino AV, Lynch AM, Winn VD, Munoz LL, Forsberg SM, Schwartz DA, Gelfand EW, Hauk PJ. Molecular networks in atopic mothers impact the risk of infant atopy. Allergy 2023; 78:244-257. [PMID: 35993851 DOI: 10.1111/all.15490] [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: 03/07/2022] [Revised: 06/28/2022] [Accepted: 07/26/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND The prevalence of atopic diseases has increased with atopic dermatitis (AD) as the earliest manifestation. We assessed if molecular risk factors in atopic mothers influence their infants' susceptibility to an atopic disease. METHODS Pregnant women and their infants with (n = 174, high-risk) or without (n = 126, low-risk) parental atopy were enrolled in a prospective birth cohort. Global differentially methylated regions (DMRs) were determined in atopic (n = 92) and non-atopic (n = 82) mothers. Principal component analysis was used to predict atopy risk in children dependent on maternal atopy. Genome-wide transcriptomic analyses were performed in paired atopic (n = 20) and non-atopic (n = 15) mothers and cord blood. Integrative genomic analyses were conducted to define methylation-gene expression relationships. RESULTS Atopic dermatitis was more prevalent in high-risk compared to low-risk children by age 2. Differential methylation analyses identified 165 DMRs distinguishing atopic from non-atopic mothers. Inclusion of DMRs in addition to maternal atopy significantly increased the odds ratio to develop AD in children from 2.56 to 4.26. In atopic compared to non-atopic mothers, 139 differentially expressed genes (DEGs) were identified significantly enriched of genes within the interferon signaling pathway. Expression quantitative trait methylation analyses dependent on maternal atopy identified 29 DEGs controlled by 136 trans-acting methylation marks, some located near transcription factors. Differential expression for the same nine genes, including MX1 and IFI6 within the interferon pathway, was identified in atopic and non-atopic mothers and high-risk and low-risk children. CONCLUSION These data suggest that in utero epigenetic and transcriptomic mechanisms predominantly involving the interferon pathway may impact and predict the development of infant atopy.
Collapse
Affiliation(s)
- Michaela Schedel
- Divisions of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA.,Department of Pulmonary Medicine, University Medicine Essen-Ruhrlandklinik, Essen, Germany.,Department of Pulmonary Medicine, University Medicine Essen, University Hospital, Essen, Germany
| | - Sonia M Leach
- Department of Biomedical Research, National Jewish Health, Denver, Colorado, USA.,Center for Genes, Environment & Health, National Jewish Health, Denver, Colorado, USA
| | - Matthew J Strand
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, Colorado, USA
| | - Thomas Danhorn
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, Colorado, USA.,Department of Pharmacology, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Morgan MacBeth
- Divisions of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA.,Department of Medical Oncology, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Anna V Faino
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, Colorado, USA.,Biostatistics, Epidemiology and Research Core, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Anne M Lynch
- Department of Ophthalmology, School of Medicine, University of Colorado, Aurora, Colorado, USA.,Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Aurora, Colorado, USA.,Department of Obstetrics and Gynecology, Stanford University, Stanford, California, USA
| | - Lindsay L Munoz
- Divisions of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA.,Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Shannon M Forsberg
- Divisions of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA.,Department of Thoracic Oncology, University of Colorado Cancer Center, University of Colorado, Aurora, Colorado, USA
| | - David A Schwartz
- Department of Medicine, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Erwin W Gelfand
- Divisions of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Pia J Hauk
- Divisions of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA.,Section Allergy/Immunology, Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
11
|
Adgent MA, Vereen S, McCullough A, Jones SH, Torstenson E, Velez Edwards DR, Hartmann KE, Carroll KN. Periconceptional folic acid supplementation and child asthma: a Right From the Start follow-up study. J Matern Fetal Neonatal Med 2022; 35:10232-10238. [PMID: 36117404 PMCID: PMC9810277 DOI: 10.1080/14767058.2022.2122795] [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: 12/13/2021] [Revised: 07/08/2022] [Accepted: 07/13/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVE High maternal folic acid exposure has been studied as a risk factor for child asthma with inconclusive results. Folic acid supplementation that begins before pregnancy may propagate high exposures during pregnancy, particularly in regions with fortified food supplies. We investigated whether folic acid supplementation initiated periconceptionally is associated with childhood asthma in a US cohort. MATERIALS AND METHODS We re-contacted mother-child dyads previously enrolled in a prospective pregnancy cohort and included children age 4 to 8 years at follow-up (n = 540). Using first trimester interviews, we assessed whether initial folic acid-containing supplement (FACS) use occurred near/before estimated conception ("periconceptional") or after (during the "first trimester"). Follow-up questionnaires were used to determine if a child ever had an asthma diagnosis ("ever asthma") or asthma diagnosis with prevalent symptoms or medication use ("current asthma"). We examined associations between FACS initiation and asthma outcomes using logistic regression, excluding preterm births and adjusting for child age, sex, maternal race, maternal education, and parental asthma. RESULTS Approximately half of women initiated FACS use periconceptionally (49%). Nine percent of children had "ever asthma" and 6% had "current asthma." Periconceptional initiation was associated with elevated odds of ever asthma [adjusted odds ratio (95% Confidence Interval): 1.65 (0.87, 3.14)] and current asthma [1.87 (0.88, 4.01)], relative to first trimester initiation. CONCLUSION We observed positive, but imprecisely estimated associations between periconceptional FACS initiation and child asthma. Folic acid prevents birth defects and is recommended. However, larger studies of folic acid dosing and timing, with consideration for childhood asthma, are needed.
Collapse
Affiliation(s)
- Margaret A. Adgent
- Division of General Pediatrics, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Epidemiology Center, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, TN
| | - Shanda Vereen
- Division of General Pediatrics, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
- College of Public Health, University of South Florida, Tampa, FL
| | - Alexis McCullough
- Division of General Pediatrics, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Sarah H. Jones
- Vanderbilt Epidemiology Center, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN
- Department of Obstetrics and Gynecology; Vanderbilt University Medical Center, Nashville, TN
- Women’s Health Research at Vanderbilt, Vanderbilt University Medical Center, Nashville, TN
| | - Eric Torstenson
- Department of Epidemiology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Digna R. Velez Edwards
- Vanderbilt Epidemiology Center, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN
- Department of Obstetrics and Gynecology; Vanderbilt University Medical Center, Nashville, TN
- Women’s Health Research at Vanderbilt, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Katherine E. Hartmann
- Vanderbilt Epidemiology Center, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN
- Department of Obstetrics and Gynecology; Vanderbilt University Medical Center, Nashville, TN
- Women’s Health Research at Vanderbilt, Vanderbilt University Medical Center, Nashville, TN
| | - Kecia N. Carroll
- Division of General Pediatrics, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pediatrics, Kravis Children’s Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
12
|
Masuda H, Kobayashi S, Miyashita C, Itoh S, Bamai YA, Saijo Y, Ito Y, Kishi R, Ikeda-Araki A, Japan Environment and Children’s Study (JECS) Group. Maternal dietary folate intake with folic acid supplements and wheeze and eczema in children aged 2 years in the Japan Environment and Children’s Study. PLoS One 2022; 17:e0272968. [PMID: 35994490 PMCID: PMC9394831 DOI: 10.1371/journal.pone.0272968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/28/2022] [Indexed: 11/18/2022] Open
Abstract
Maternal intake of folic acid supplements is reportedly associated with the risk of early-onset allergies in offspring. However, only a few studies have considered the intake of both folic acid supplements and dietary folate. Here, the relationship between maternal intake of folic acid supplements and allergic symptoms such as wheeze and eczema in offspring was analyzed while considering dietary folate intake. We examined 84,361 mothers and 85,114 children in the Japan Environment and Children’s Study. The participants were divided into three groups depending on maternal folic acid supplementation (“no use,” “occasional use,” and “daily use”). Each group was then subdivided into three groups based on total folic acid and dietary folate intake. Outcomes were determined considering the wheeze and eczema status of each child at the age of 2 years. The status was based on the International Study of Asthma and Allergies in Childhood. It was found that 22.1% of the mothers took folic acid supplements daily. In contrast, 56.3% of the mothers did not take these supplements. Maternal intake of folic acid supplements was not associated with wheeze and eczema in the offspring. In contrast, only dietary folate intake was positively associated with wheeze at the age of 2 (adjusted odds ratio, 1.103; 95% confidence interval, 1.003–1.212). However, there is no scientific evidence of a biological mechanism that clarifies this result. Potential confounders such as other nutrition, outdoor/indoor air pollution, and genetic factors may have affected the results. Therefore, further studies on the association between maternal intake of folic acid and allergic symptoms at the age of 3 or above are needed to confirm the results of this study. Trial registration UMIN Clinical Trials Registry (number: UMIN000030786)
Collapse
Affiliation(s)
- Hideyuki Masuda
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Sumitaka Kobayashi
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Chihiro Miyashita
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Sachiko Itoh
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Yu Ait Bamai
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Yasuaki Saijo
- Department of Social Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Yoshiya Ito
- Faculty of Nursing, Japanese Red Cross Hokkaido College of Nursing, Kitami, Japan
| | - Reiko Kishi
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Atsuko Ikeda-Araki
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
- * E-mail:
| | | |
Collapse
|
13
|
Yang F, Zhu J, Wang Z, Wang L, Tan T, Sun L. Relationship between maternal folic acid supplementation during pregnancy and risk of childhood asthma: Systematic review and dose-response meta-analysis. Front Pediatr 2022; 10:1000532. [PMID: 36467483 PMCID: PMC9714269 DOI: 10.3389/fped.2022.1000532] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/31/2022] [Indexed: 11/18/2022] Open
Abstract
Growing evidence suggests that maternal folic acid supplementation during pregnancy may be associated with the risk of childhood asthma, but these findings remain controversial. Therefore, the purpose of this systematic review and meta-analysis was to assess the association between maternal folic acid supplementation during pregnancy and the risk of childhood asthma, and to determine the safe dose of folic acid supplementation during pregnancy based on a dose-response analysis to lower the risk of childhood asthma. The PubMed, Embase, Cochrane Library, and Web of Science databases were searched for relevant studies published before April 2022. The Newcastle-Ottawa Scale (NOS) was used to evaluate the quality of eligible studies, and a fixed-effect model was employed to calculate the odds ratio (OR) of asthma with 95% confidence intervals (CI). In addition, the generalized least-squares trend (GLST) was used to explore a nonlinear dose-response relationship. Stata 15.0 was used for the statistical analysis mentioned above. This systematic review included 18 studies (13 cohort studies, 5 case-control studies) with a total of 252,770 participants, 50,248 of whom were children with asthma. The meta-analysis showed that maternal folic acid supplementation during pregnancy was significantly associated with the risk of childhood asthma (OR = 1.07; 95% CI = 1.04-1.11). The subgroup analysis revealed a significant correlation between the risk of childhood asthma and the folic acid supplementation in the first Trimester (OR = 1.09; 95% CI = 1.05-1.12), the third Trimester (OR = 1.15; 95% CI = 1.04-1.26) and the whole pregnancy (OR = 1.13; 95% CI = 1.10-1.16). At the same time, the dose-response analysis showed a nonlinear relationship between maternal folic acid intake during pregnancy and the risk of childhood asthma. The risk of asthma in children significantly increased when maternal folic acid intake reached 581 μg/day. This meta-analysis showed that maternal folic acid supplementation during pregnancy increased the risk of asthma in children. Based on the results of the dose-response analysis, less than 580 μg folic acid per day is advised in order to effectively prevent birth defects without increasing the risk of childhood asthma. Systematic Review Registration: https://www.crd.york.ac.uk/prospero/display_record.php?, identifier: CRD42022332140.
Collapse
Affiliation(s)
- Fushuang Yang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jinpu Zhu
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Zhongtian Wang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Lei Wang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Tianhui Tan
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Liping Sun
- Center of Children's Clinic, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
14
|
Kang CM, Chiang BL, Wang LC. Maternal Nutritional Status and Development of Atopic Dermatitis in Their Offspring. Clin Rev Allergy Immunol 2021; 61:128-155. [PMID: 32157654 DOI: 10.1007/s12016-020-08780-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Atopic dermatitis (AD) is the leading chronic skin inflammatory disease and the initial manifestation of atopic march. Available evidence supports the notion that primary prevention early in life leads to a decreased incidence of AD, thus possibly decreasing the subsequent occurrence of atopic march. Nutritional status is essential to a proper functioning immune system and is valued for its important role in AD. Essential nutrients, which include carbohydrates, proteins, lipids, vitamins, and minerals, are transferred from the mother to the fetus through the placenta during gestation. Various nutrients, such as polyunsaturated fatty acids (PUFAs) and vitamin D, were studied in relation to maternal status and offspring allergy. However, no strong evidence indicates that a single nutrient or food in mothers' diet significantly affects the risk of childhood AD. In the light of current evidence, mothers should not either increase nor avoid consuming these nutrients to prevent or ameliorate allergic diseases in their offspring. Each essential nutrient has an important role in fetal development, and current government recommendations suggest specific intake amounts for pregnant women. This review discusses evidence on how various nutrients, including lipids (monounsaturated fatty acids, PUFAs, saturated fatty acids, and short-chain fatty acids), carbohydrates (oligosaccharides and polysaccharides), proteins, vitamins (A, B, C, D, and E), and trace minerals (magnesium, iron, zinc, copper, selenium, and strontium) in maternal status are associated with the development of AD and their possible mechanisms.
Collapse
Affiliation(s)
- Chun-Min Kang
- Department of Pediatrics, National Taiwan University Hospital, No. 7, Chung Shan South Road, Taipei, 10002, Taiwan, Republic of China
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Bor-Luen Chiang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Li-Chieh Wang
- Department of Pediatrics, National Taiwan University Hospital, No. 7, Chung Shan South Road, Taipei, 10002, Taiwan, Republic of China.
| |
Collapse
|
15
|
Best KP, Green TJ, Sulistyoningrum DC, Sullivan TR, Aufreiter S, Prescott SL, Makrides M, Skubisz M, O'Connor DL, Palmer DJ. Maternal Late-Pregnancy Serum Unmetabolized Folic Acid Concentrations Are Not Associated with Infant Allergic Disease: A Prospective Cohort Study. J Nutr 2021; 151:1553-1560. [PMID: 33851208 DOI: 10.1093/jn/nxab040] [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: 12/15/2020] [Revised: 01/14/2021] [Accepted: 02/02/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The increase in childhood allergic disease in recent decades has coincided with increased folic acid intakes during pregnancy. Circulating unmetabolized folic acid (UMFA) has been proposed as a biomarker of excessive folic acid intake. OBJECTIVE We aimed to determine if late-pregnancy serum UMFA and total folate concentrations were associated with allergic disease risk in the offspring at 1 y of age in a population at high risk of allergy. METHODS The cohort consisted of 561 mother-infant pairs from Western Australia. To be eligible the infant had to have a first-degree relative (mother, father, or sibling) with a history of medically diagnosed allergic disease. Maternal venous blood was collected between 36 and 40 wk of gestation. Serum UMFA was measured by LC-tandem MS. Serum total folate was determined using a microbiological method with chloramphenicol-resistant Lactobacillus rhamnosus as the test organism, and was collected between 36 and 40 wk of gestation. UMFA concentrations were measured by tandem MS using stable isotope dilution; folate concentrations were determined using the microbiological method with standardized kits. Infant allergic disease outcomes of medically diagnosed eczema, steroid-treated eczema, atopic eczema, IgE-mediated food allergy, allergen sensitization, and medically diagnosed wheeze were assessed at 1 y of age. RESULTS Median (IQR) concentrations for UMFA and serum folate were 1.6 (0.6-4.7) and 53.2 (32.6-74.5) nmol/L, respectively. Of the infants, 34.6% had medically diagnosed eczema, 26.4% allergen sensitization, and 14.9% had an IgE-mediated food allergy. In both adjusted and unadjusted models there was little evidence of association between UMFA or serum folate and any of the infant allergy outcomes. CONCLUSIONS In this cohort of children at high risk of allergic disease there was no association between maternal UMFA or serum folate concentrations measured in late pregnancy and allergic disease outcomes at 1 y of age.
Collapse
Affiliation(s)
- Karen P Best
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Tim J Green
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Dian C Sulistyoningrum
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Thomas R Sullivan
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, Australia.,School of Public Health, University of Adelaide, Adelaide, Australia
| | - Susanne Aufreiter
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Susan L Prescott
- The ORIGINS Project, Telethon Kids, Nedlands, Australia.,Department of Immunology, Perth Children's Hospital, Nedlands, Australia.,School of Medicine, The University of Western Australia, Crawley, Australia
| | - Maria Makrides
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Monika Skubisz
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Deborah L O'Connor
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Debra J Palmer
- School of Medicine, The University of Western Australia, Crawley, Australia.,Telethon Kids Institute, The University of Western Australia, Nedlands, Australia
| |
Collapse
|
16
|
Mida LA, della Zazzera V, Fontaine-Bisson B. Knowledge, attitude and practice of physicians regarding periconceptional folic acid for women at low risk of a neural tube defect affected pregnancy. Prev Med Rep 2021; 22:101327. [PMID: 33665065 PMCID: PMC7903458 DOI: 10.1016/j.pmedr.2021.101327] [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: 02/26/2020] [Revised: 12/18/2020] [Accepted: 01/23/2021] [Indexed: 11/19/2022] Open
Abstract
Canadian expert guidelines recommend low-risk women to consume a daily multivitamin supplement containing 400 µg of folic acid (FA) to prevent neural tube defects. Mandatory food fortification coupled with intake of prenatal vitamin/mineral supplements (PVS), most of which contain ≥ 1000 µg-FA, has resulted in an unprecedented shift in Canadian pregnant women folate status. This study assessed the knowledge, attitude and practice (KAP) of physicians regarding periconceptional FA recommendations, intake and health related outcomes, since they play an essential role in promoting appropriate FA intake. Seventy-seven physicians answered the self-administered KAP survey. Only half of physicians knew the correct dose and duration of FA for low-risk women. Approximately 70% were unsure of, or unfamiliar with the most recent guidelines and 60% of physicians most often recommend a ≥ 1000 µg-FA supplement. Knowledge score 1 (KS1), which related to low-risk women, was associated with physicians' attitude toward believing that most PVS contain the recommended amount of FA (p = 0.004). Significant correlations were also found between KS1 and the total practice score (TPS) (r = 0.45, p < 0.0001) as well as between the total knowledge score and TPS (r = 0.38, p = 0.0007). Our findings show that physicians lacking knowledge regarding periconceptional FA is associated with their attitude and practice. Despite a vast majority of physicians being unsure or uncomfortable recommending PVS that are not in line with recommendations, a lack of knowledge and a widely accessible 400 µg-FA PVS, enables a contradictory practice in reality.
Collapse
Affiliation(s)
- Liana Arielle Mida
- Interdisciplinary School of Health Sciences, University of Ottawa, 25 University Private, Ottawa, Canada
| | | | - Bénédicte Fontaine-Bisson
- School of Nutrition Sciences, University of Ottawa, 25 University Private, Ottawa, Canada
- Institut du savoir Montfort, Hôpital Montfort, 713 Montreal Road, Ottawa, Canada
| |
Collapse
|
17
|
Acevedo N, Alashkar Alhamwe B, Caraballo L, Ding M, Ferrante A, Garn H, Garssen J, Hii CS, Irvine J, Llinás-Caballero K, López JF, Miethe S, Perveen K, Pogge von Strandmann E, Sokolowska M, Potaczek DP, van Esch BCAM. Perinatal and Early-Life Nutrition, Epigenetics, and Allergy. Nutrients 2021; 13:724. [PMID: 33668787 PMCID: PMC7996340 DOI: 10.3390/nu13030724] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 02/08/2023] Open
Abstract
Epidemiological studies have shown a dramatic increase in the incidence and the prevalence of allergic diseases over the last several decades. Environmental triggers including risk factors (e.g., pollution), the loss of rural living conditions (e.g., farming conditions), and nutritional status (e.g., maternal, breastfeeding) are considered major contributors to this increase. The influences of these environmental factors are thought to be mediated by epigenetic mechanisms which are heritable, reversible, and biologically relevant biochemical modifications of the chromatin carrying the genetic information without changing the nucleotide sequence of the genome. An important feature characterizing epigenetically-mediated processes is the existence of a time frame where the induced effects are the strongest and therefore most crucial. This period between conception, pregnancy, and the first years of life (e.g., first 1000 days) is considered the optimal time for environmental factors, such as nutrition, to exert their beneficial epigenetic effects. In the current review, we discussed the impact of the exposure to bacteria, viruses, parasites, fungal components, microbiome metabolites, and specific nutritional components (e.g., polyunsaturated fatty acids (PUFA), vitamins, plant- and animal-derived microRNAs, breast milk) on the epigenetic patterns related to allergic manifestations. We gave insight into the epigenetic signature of bioactive milk components and the effects of specific nutrition on neonatal T cell development. Several lines of evidence suggest that atypical metabolic reprogramming induced by extrinsic factors such as allergens, viruses, pollutants, diet, or microbiome might drive cellular metabolic dysfunctions and defective immune responses in allergic disease. Therefore, we described the current knowledge on the relationship between immunometabolism and allergy mediated by epigenetic mechanisms. The knowledge as presented will give insight into epigenetic changes and the potential of maternal and post-natal nutrition on the development of allergic disease.
Collapse
Affiliation(s)
- Nathalie Acevedo
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Bilal Alashkar Alhamwe
- Institute of Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, 35043 Marburg, Germany; (B.A.A.); (E.P.v.S.)
- College of Pharmacy, International University for Science and Technology (IUST), Daraa 15, Syria
| | - Luis Caraballo
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Mei Ding
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos, Switzerland; (M.D.); (M.S.)
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos, Switzerland
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Antonio Ferrante
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
- School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Holger Garn
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (H.G.); (S.M.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands;
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands
| | - Charles S. Hii
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - James Irvine
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Kevin Llinás-Caballero
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Juan Felipe López
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Sarah Miethe
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (H.G.); (S.M.)
| | - Khalida Perveen
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Elke Pogge von Strandmann
- Institute of Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, 35043 Marburg, Germany; (B.A.A.); (E.P.v.S.)
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos, Switzerland; (M.D.); (M.S.)
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos, Switzerland
| | - Daniel P. Potaczek
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (H.G.); (S.M.)
| | - Betty C. A. M. van Esch
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands;
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
18
|
ŞİMŞEK H, KARAAĞAÇ Y, TUNÇER E, YARDIMCI H. Gebelikte Folik Asit, B12 Vitamini, D Vitamini ve İyot Destekleri Kullanmak Gerekli Midir?: Olası Riskler. KAHRAMANMARAŞ SÜTÇÜ İMAM ÜNIVERSITESI TIP FAKÜLTESI DERGISI 2021. [DOI: 10.17517/ksutfd.832401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
19
|
Ye Y, Dou LM, Zhang Y, Dou YL, Zhao PP, Jiang Y, Gao W, Ji M, He LF, Niu DY, Zhang L, Gao XH, Li Y, Xiao LP, Huang J, Zhang XH, Wang LH, Yan WL. Maternal periconceptional folate status and infant atopic dermatitis: A prospective cohort study. Pediatr Allergy Immunol 2021; 32:137-145. [PMID: 32663346 DOI: 10.1111/pai.13321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 06/28/2020] [Accepted: 07/06/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Maternal folate status is linked with the risk of allergic disorders including atopic dermatitis (AD) in children, but findings remain inconclusive. We aim to assess the relationship between maternal folate status in early gestation and early-onset infant AD, based on a prospective mother-child cohort study. METHODS Pregnant women were recruited at 12-14 weeks of gestation. Red blood cell folate (RBC folate) and serum folate concentrations were examined at enrollment. Periconceptional folic acid supplementation was investigated through a self-administered questionnaire. The primary outcome was AD incidence before 6 months of age, diagnosed according to Williams' criteria. Multivariate logistic regression was used to evaluate associations of maternal folate status with infant AD by adjusting parental and child covariates. RESULTS In total, 107 (23.4%) of 458 infants developed AD before 6 months, with more male infants affected (P = .002). Higher maternal RBC folate levels (per 100 ng/mL) were associated with an increased risk of AD (adjusted odds ratio [aOR] 1.16, 95% confidence interval [CI] 1.04-1.31). An RBC folate level ≥620 ng/mL was associated with increased infant AD by 91% (aOR 1.91, 95% CI 1.09-3.36). However, associations were not observed for maternal serum folate at early gestation or periconceptional folic acid supplement intakes. CONCLUSIONS We provide the first evidence that higher maternal RBC folate concentrations during early gestation are associated with increased early-onset infant AD. Our findings support the importance of maintaining appropriate folate levels during the periconceptional period to reduce the risk of AD in infants.
Collapse
Affiliation(s)
- Ying Ye
- Department of Clinical Epidemiology, Children's Hospital of Fudan University, Shanghai, China.,Department of Dermatology, Children's Hospital of Fudan University, Shanghai, China
| | - Li-Min Dou
- Department of Dermatology, Children's Hospital of Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Clinical Epidemiology, Children's Hospital of Fudan University, Shanghai, China
| | - Ya-Lan Dou
- Department of Clinical Epidemiology, Children's Hospital of Fudan University, Shanghai, China
| | - Piao-Ping Zhao
- Department of Dermatology, Children's Hospital of Fudan University, Shanghai, China
| | - Yuan Jiang
- Department of Clinical Epidemiology, Children's Hospital of Fudan University, Shanghai, China
| | - Wei Gao
- Department of Dermatology, Children's Hospital of Fudan University, Shanghai, China
| | - Mi Ji
- Department of Clinical Epidemiology, Children's Hospital of Fudan University, Shanghai, China
| | - Lin-Feng He
- Department of Dermatology, Children's Hospital of Fudan University, Shanghai, China
| | - Da-Yan Niu
- Department of Clinical Epidemiology, Children's Hospital of Fudan University, Shanghai, China
| | - Lei Zhang
- Shanghai Minhang Maternal and Children Health Care Hospital, Shanghai, China
| | - Xiao-Hua Gao
- Shanghai Minhang Maternal and Children Health Care Hospital, Shanghai, China
| | - Yun Li
- Shanghai Minhang Maternal and Children Health Care Hospital, Shanghai, China
| | - Li-Ping Xiao
- Shanghai Minhang Maternal and Children Health Care Hospital, Shanghai, China
| | - Jun Huang
- Shanghai Minhang Maternal and Children Health Care Hospital, Shanghai, China
| | - Xiao-Hua Zhang
- Shanghai Minhang Maternal and Children Health Care Hospital, Shanghai, China
| | - Liu-Hui Wang
- Department of Dermatology, Children's Hospital of Fudan University, Shanghai, China
| | - Wei-Li Yan
- Department of Clinical Epidemiology, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
20
|
Sulistyoningrum D, Green T, Palmer D, Sullivan T, Wood S, Makrides M, Skubisz M, Best KP. Study protocol for a randomised controlled trial evaluating the effect of folic acid supplementation beyond the first trimester on maternal plasma unmetabolised folic acid in late gestation. BMJ Open 2020; 10:e040416. [PMID: 33199423 PMCID: PMC7670954 DOI: 10.1136/bmjopen-2020-040416] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/21/2020] [Accepted: 10/13/2020] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Taking folic acid containing supplements prior to and during early pregnancy reduces the risk of neural tube defects. Neural tube defects occur prior to 28 days postconception, after which, there is no proven benefit of continuing to take folic acid. However, many women continue to take folic acid containing supplements throughout the pregnancy. At higher intakes, folic acid is not converted to its active form and accumulates in circulation as unmetabolised folic acid (UMFA). Recently, concerns have been raised about possible links between late gestation folic acid supplementation and childhood allergy, metabolic disease and autism spectrum disorders. We aim to determine if removing folic acid from prenatal micronutrient supplements after 12 weeks gestation reduces circulating levels of maternal UMFA at 36 weeks gestation. METHODS AND ANALYSIS This is a parallel-design, double-blinded randomised controlled trial. Women ≥12 and <16 weeks' gestation with a singleton pregnancy and able to give informed consent are eligible to participate. Women (n=100; 50 per group) will be randomised to receive either a micronutrient supplement containing 0.8 mg of folic acid or a micronutrient supplement without folic acid daily from enrolment until delivery. The primary outcome is plasma UMFA concentration at 36 weeks gestation. Secondary outcomes include red blood cell folate and total plasma folate concentration. We will assess whether there is a difference in mean UMFA levels at 36 weeks gestation between groups using linear regression with adjustment for baseline UMFA levels and gestational age at trial entry. The treatment effect will be described as a mean difference with 95% CI. ETHICS AND DISSEMINATION Ethical approval has been granted from the Women's and Children's Health Network Research Ethics Committee (HREC/19/WCHN/018). The results of this trial will be presented at scientific conferences and published in peer-reviewed journals. TRIAL REGISTRATION NUMBER ACTRN12619001511123.
Collapse
Affiliation(s)
- Dian Sulistyoningrum
- SAHMRI Women and Kids Theme, South Australia Health and Medical Research Institute, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Tim Green
- SAHMRI Women and Kids Theme, South Australia Health and Medical Research Institute, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Debbie Palmer
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Thomas Sullivan
- SAHMRI Women and Kids Theme, South Australia Health and Medical Research Institute, Adelaide, South Australia, Australia
- Faculty of Health and Medical Sciences, School of Public Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Simon Wood
- Faculty of Science and Engineering, Curtin University, Perth, West Australia, Australia
- Faculty of Land and Food Systems, University of British Columbia, Vancouver, British Columbia, Canada
| | - Maria Makrides
- SAHMRI Women and Kids Theme, South Australia Health and Medical Research Institute, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Monika Skubisz
- SAHMRI Women and Kids Theme, South Australia Health and Medical Research Institute, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Karen P Best
- SAHMRI Women and Kids Theme, South Australia Health and Medical Research Institute, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
21
|
Moore R, Anturaniemi J, Velagapudi V, Nandania J, Barrouin-Melo SM, Hielm-Björkman A. Targeted Metabolomics With Ultraperformance Liquid Chromatography-Mass Spectrometry (UPLC-MS) Highlights Metabolic Differences in Healthy and Atopic Staffordshire Bull Terriers Fed Two Different Diets, A Pilot Study. Front Vet Sci 2020; 7:554296. [PMID: 33195525 PMCID: PMC7653775 DOI: 10.3389/fvets.2020.554296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/22/2020] [Indexed: 01/09/2023] Open
Abstract
Background: While anecdotal evidence has long claimed that a raw meat-based diet (RMBD) improves the metabolic health of canines, no rigorous scientific study has clarified this issue. Canine atopic dermatitis (CAD) has also been linked to metabolic health, but its relation to diet remains poorly understood. This study investigates whether dietary choice is linked to metabolic health in healthy and CAD-diagnosed canines via targeted serum and urine metabolomic analysis of polar, non-ionic metabolites, as well as whether the underlying CAD condition modulates the response to nutritional intake. Materials and Methods: Serum metabolites of client-owned Staffordshire bull terriers, divided into CAD-diagnosed (n = 14) and healthy (n = 6) cohorts, were studied. Urine metabolites of a subset of the CAD-diagnosed canines (n = 8) were also studied. The canines were split into two cohorts based on diet. The first cohort were fed a commercially available high-fat, moderate-protein, low-carbohydrate RMBD (n = 11, CAD diagnosed n = 8, healthy n = 3). Those in the second cohort were fed a commercially available moderate-fat, moderate-protein, high-carbohydrate kibble diet (KD) (n = 9: CAD diagnosed n = 6, healthy n = 3). The diet intervention period lasted approximately 4.5 months (median 135 days). Statistical analyses of the serum profiles across all dogs (n = 20) and the urine profiles of the CAD-diagnosed subset (n = 8) were performed. Results and Discussion: The KD cohort was found to have higher concentrations of methionine than the RMBD cohort, both in serum (all dogs, p < 0.0001) and in urine (CAD-only cohort, p < 0.0002), as well as cystathionine and 4-pyridoxic acid. Methionine plays important roles in homocysteine metabolism, and elevated levels have been implicated in various pathologies. The CAD (n = 14) cohort dogs showed starker metabolic changes in response to diet regarding these pathways compared to the healthy (n = 6) cohort. However, there was no significant change in CAD severity as a result of either diet. Likely due to the higher meat content of the RMBD, higher concentrations of several carnitines and creatine were found in the RMBD cohort. Citrulline was found in higher concentrations in the KD cohort. Our findings provide insight into the relationship between diet and the serum and urine metabolite profiles of canines. They also suggest that neither diet significantly affected CAD severity.
Collapse
Affiliation(s)
- Robin Moore
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Johanna Anturaniemi
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Vidya Velagapudi
- Metabolomics Unit, Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jatin Nandania
- Metabolomics Unit, Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Stella Maria Barrouin-Melo
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Veterinary Anatomy, Pathology and Clinics, School of Veterinary Medicine and Zootechny, Federal University of Bahia, Salvador, Brazil
| | - Anna Hielm-Björkman
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
22
|
Maternal Folic Acid Supplementation Mediates Offspring Health via DNA Methylation. Reprod Sci 2020; 27:963-976. [PMID: 32124397 DOI: 10.1007/s43032-020-00161-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/09/2019] [Indexed: 10/24/2022]
Abstract
The clinical significance of periconceptional folic acid supplementation (FAS) in the prevention of neonatal neural tube defects (NTDs) has been recognized for decades. Epidemiological data and experimental findings have consistently been indicating an association between folate deficiency in the first trimester of pregnancy and poor fetal development as well as offspring health (i.e., NTDs, isolated orofacial clefts, neurodevelopmental disorders). Moreover, compelling evidence has suggested adverse effects of folate overload during perinatal period on offspring health (i.e., immune diseases, autism, lipid disorders). In addition to several single-nucleotide polymorphisms (SNPs) in genes related to folate one-carbon metabolism (FOCM), folate concentrations in maternal serum/plasma/red blood cells must be considered when counseling FAS. Epigenetic information encoded by 5-methylcytosines (5mC) plays a critical role in fetal development and offspring health. S-adenosylmethionine (SAM), a methyl donor for 5mC, could be derived from FOCM. As such, folic acid plays a double-edged sword role in offspring health via mediating DNA methylation. However, the underlying epigenetic mechanism is still largely unclear. In this review, we summarized the link across DNA methylation, maternal FAS, and offspring health to provide more evidence for clinical guidance in terms of precise FAS dosage and time point. Future studies are, therefore, required to set up the reference intervals of folate concentrations at different trimesters of pregnancy for different populations and to clarify the epigenetic mechanism for specific offspring diseases.
Collapse
|
23
|
Huang KT, Shen YL, Lee CN, Chu KY, Ku WC, Liu CY, Huang RFS. Using Differential Threshold Effects of Individual and Combined Periconceptional Methyl Donor Status on Maternal Genomic LINE-1 and Imprinted H19 DNA Methylation to Predict Birth Weight Variance in the Taiwan Pregnancy-Newborn Epigenetics (TPNE) Cohort Study. J Nutr 2020; 150:108-117. [PMID: 31504733 DOI: 10.1093/jn/nxz204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Few studies have comprehensively examined the effect of methyl donor status on maternal DNA methylation and birth outcomes. OBJECTIVES This study examined associations between periconceptional methyl donor status and genome-wide and specific imprinted gene methylation and fetal growth indices in the Taiwan Pregnancy-Newborn Epigenetics cohort. METHODS Plasma folate, choline (free form), and betaine concentrations of the participants enrolled at 7-10 weeks of gestation were analyzed. DNA methylation at regulatory sequences of the imprinted H19 gene and genomic long interspersed nuclear element 1 (LINE-1) were measured in maternal lymphocytes using bisulfite/high-resolution melt polymerase chain reaction. Associations with birth weight (BW) were estimated through multiple regressions from 112 mother-newborn pairs. RESULTS A nonlinear "L-shaped" relation and an inverse association between maternal plasma folate in T1 (mean ± SE: 17.6 ± 5.1 nmol/L) and lymphocytic LINE-1 methylation (β: -0.49, P = 0.027) were characterized. After adjusting for LINE-1 methylation, individual maternal folate concentrations were positively associated with BW variance (β = 0.24, P = 0.035), and the association was more pronounced in mothers with choline in T1 (mean ± SE: 5.4 ± 0.6 μmol/L; β: 0.40, P = 0.039). Choline status of the mothers in T2 (mean ± SE: 7.2 ± 0.6 μmol/L) was inversely associated with LINE-1 methylation (β: -0.43, P = 0.035), and a positive association was evident between T1 choline and H19 methylation (β: 0.48, P = 0.011). After adjusting for epigenetic modification, maternal choline status predicted a positive association with BW (β: 0.56, P = 0.005), but the effect was limited to mothers with high betaine concentrations in T3 (mean ± SE: 36.4 ± 8.8 μmol/L), depending on folate status. CONCLUSIONS Our data highlight the differential threshold effects of periconceptional folate, choline, and betaine status on genomic LINE-1 and H19 DNA methylation and how their interplay has a long-term effect on BW variance.
Collapse
Affiliation(s)
- Kuang-Ta Huang
- PhD Program in Nutrition and Food Science, Fu Jen Catholic University, New Taipei City, Taiwan.,Loving Care Maternity and Children's Health Centers, New Taipei City, Taiwan
| | - Yu-Li Shen
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chien-Nan Lee
- Department of Gynecology and Obstetrics, National Taiwan University Hospital, Taipei City, Taiwan
| | - Kuan-Yu Chu
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Wei-Chi Ku
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chieh-Yu Liu
- Biostatistical Consultant Lab, Department of Speech Language Pathology and Audiology, National Taipei University of Nursing and Health Sciences, Taipei City, Taiwan
| | - Rwei-Fen S Huang
- PhD Program in Nutrition and Food Science, Fu Jen Catholic University, New Taipei City, Taiwan.,Department of Nutritional Science, Fu Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
24
|
Molloy J, Collier F, Saffery R, Allen KJ, Koplin JJ, Louise Ponsonby A, Tang MLK, Ward AC, Martino D, Burgner D, Carlin JB, Ranganathan S, Symeonedies C, Dwyer T, Vuillermin P. Folate levels in pregnancy and offspring food allergy and eczema. Pediatr Allergy Immunol 2020; 31:38-46. [PMID: 31566807 DOI: 10.1111/pai.13128] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 09/09/2019] [Accepted: 09/16/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND High folate status in pregnancy has been implicated in the increased prevalence of allergic disease, but there are no published data relating directly measured folate status in pregnancy to challenge-proven food allergy among offspring. The study aim was to examine the association between red blood cell (RBC) folate status in trimester three of pregnancy and allergic disease among offspring. METHODS Red blood cell folate levels were measured at 28-32 weeks' gestation in a prospective birth cohort (n = 1074). Food allergy outcomes were assessed in 1-year-old infants by skin prick testing and subsequent food challenge. Eczema was assessed by questionnaire and clinical review. High trimester three RBC folate was defined as greater than (>) 1360 nmol/L. Binomial regression was used to examine associations between trimester three RBC folate and allergic outcomes, adjusting for potential confounders. RESULTS Red blood cell folate levels were measured in 88% (894/1064) of pregnant women. The mean concentration was 1695.6 nmol/L (standard deviation 415.4) with 82% (731/894) >1360 nmol/L. There was no evidence of either linear or non-linear relationships between trimester three RBC folate and allergic outcomes, nor evidence of associations between high RBC folate and food allergy (adjusted risk ratio (aRR) 2.89, 95% CI 0.90-9.35), food sensitization (aRR 1.72, 95% CI 0.85-3.49), or eczema (aRR 0.97, 95% CI 0.67-1.38). CONCLUSION The majority of pregnant women in this study had high RBC folate levels. There was no evidence of associations between trimester three RBC folate and food allergy, food sensitization, or eczema among the offspring, although larger studies are required.
Collapse
Affiliation(s)
- John Molloy
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia.,Child Health Research Unit, Barwon Health, Geelong, Victoria, Australia.,Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Centre for Food and Allergy Research, Parkville, Victoria, Australia
| | - Fiona Collier
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia.,Child Health Research Unit, Barwon Health, Geelong, Victoria, Australia
| | - Richard Saffery
- Murdoch Childrens Research Institute, Parkville, Victoria, Australia
| | - Katrina J Allen
- Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Centre for Food and Allergy Research, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.,Department of Allergy and Immunology, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Jennifer J Koplin
- Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Centre for Food and Allergy Research, Parkville, Victoria, Australia.,Centre for Epidemiology and Biostatistics, The University of Melbourne, Carlton, Victoria, Australia
| | - Anne Louise Ponsonby
- Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Centre for Food and Allergy Research, Parkville, Victoria, Australia
| | - Mimi L K Tang
- Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Centre for Food and Allergy Research, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.,Department of Allergy and Immunology, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Alister C Ward
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - David Martino
- Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Centre for Food and Allergy Research, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - David Burgner
- Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.,Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - John B Carlin
- Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.,Centre for Epidemiology and Biostatistics, The University of Melbourne, Carlton, Victoria, Australia
| | - Sarath Ranganathan
- Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.,Department of Respiratory Medicine, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Christos Symeonedies
- Child Health Research Unit, Barwon Health, Geelong, Victoria, Australia.,Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Terence Dwyer
- The George Institute for Global Health, University of Oxford, Oxford, UK
| | | | - Peter Vuillermin
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia.,Child Health Research Unit, Barwon Health, Geelong, Victoria, Australia.,Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Centre for Food and Allergy Research, Parkville, Victoria, Australia
| |
Collapse
|
25
|
Chen Z, Xing Y, Yu X, Dou Y, Ma D. Effect of Folic Acid Intake on Infant and Child Allergic Diseases: Systematic Review and Meta-Analysis. Front Pediatr 2020; 8:615406. [PMID: 33537268 PMCID: PMC7848186 DOI: 10.3389/fped.2020.615406] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/14/2020] [Indexed: 11/17/2022] Open
Abstract
Objective: This study aimed to analyze the effect of folic acid supplements on infant and child allergic diseases through systematic review and meta-analysis. Design: PubMed, The Cochrane Library and references of related articles published before January 1, 2020 were searched. Setting: Meta-analysis was used to explore the influence of folic acid on skin allergies (eczema, and atopic dermatitis) and respiratory allergies (asthma, wheezing, and allergic rhinitis). Participants: Data were collected from 15 studies with 244,018 individual participants from five different countries for meta-analysis. Results: Folic acid was confirmed as a risk factor for allergic diseases in infant and child. The risk of allergic diseases dramatically increased when maternal folic acid intake <400 μg/day (RR = 1.050; 95% CI = 1.027-1.073) during pregnancy. Stratified analyses revealed that the association was significant only for respiratory allergy (RR = 1.067; 95% CI = 1.028-1.108) and pregnant women who only used folic acid supplements (RR = 1.070; 95% CI = 1.030-1.112) and that countries without folic acid fortification (RR = 1.046; 95% CI = 1.026-1.067). Conclusions: This study suggested that folic acid intake can be a risk factor for allergic diseases, especially respiratory tract allergies among infants and young children. Furthermore, pregnant women should pay attention to supplementation of folic acid from both folic acid supplements and fortified foods with folic acid during pregnancy.
Collapse
Affiliation(s)
- Zekun Chen
- School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yan Xing
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Xue Yu
- School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yuqi Dou
- School of Public Health, Peking University Health Science Center, Beijing, China
| | - Defu Ma
- School of Public Health, Peking University Health Science Center, Beijing, China
| |
Collapse
|
26
|
Vereen S, Gebretsadik T, Johnson N, Hartman TJ, Veeranki SP, Piyathilake C, Mitchel EF, Kocak M, Cooper WO, Dupont WD, Tylavsky F, Carroll KN. Association Between Maternal 2nd Trimester Plasma Folate Levels and Infant Bronchiolitis. Matern Child Health J 2019; 23:164-172. [PMID: 30027465 DOI: 10.1007/s10995-018-2610-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Objectives Viral bronchiolitis is the most common cause of infant hospitalization. Folic acid supplementation is important during the periconceptional period to prevent neural tube defects. An area of investigation is whether higher prenatal folate is a risk factor for childhood respiratory illnesses. We investigated the association between maternal 2nd trimester plasma folate levels and infant bronchiolitis. Methods We conducted a retrospective cohort analysis in a subset of mother-infant dyads (n = 676) enrolled in the Conditions Affecting Neurocognitive Development and Learning in Early Childhood study and Tennessee Medicaid. Maternal folate status was determined using 2nd trimester (16-28 weeks) plasma samples. Bronchiolitis diagnosis in the first year of life was ascertained using International Classification of Diagnosis-9 codes from Medicaid administrative data. We used multivariable logistic regression to assess the adjusted association of prenatal folate levels and infant bronchiolitis outcome. Results Half of the women in this lower-income and predominately African-American (84%) study population had high levels of folate (median 2nd trimester level 19.2 ng/mL) and 21% of infants had at least one bronchiolitis healthcare visit. A relationship initially positive then reversing between maternal plasma folate and infant bronchiolitis was observed that did not reach statistical significance (poverall = .112, pnonlinear effect = .088). Additional adjustment for dietary methyl donor intake did not significantly alter the association. Conclusions for Practice Results did not confirm a statistically significant association between maternal 2nd trimester plasma folate levels and infant bronchiolitis. Further work is needed to investigate the role of folate, particularly higher levels, in association with early childhood respiratory illnesses.
Collapse
Affiliation(s)
- Shanda Vereen
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Divisions of General Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Asthma Health Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tebeb Gebretsadik
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Asthma Health Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Terryl J Hartman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Sreenivas P Veeranki
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Divisions of General Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Asthma Health Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chandrika Piyathilake
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Edward F Mitchel
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mehmet Kocak
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - William O Cooper
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Divisions of General Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - William D Dupont
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Asthma Health Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Frances Tylavsky
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kecia N Carroll
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA. .,Divisions of General Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA. .,Center for Asthma Health Research, Vanderbilt University Medical Center, Nashville, TN, USA. .,Vanderbilt University School of Medicine, 313 Oxford House, 1313 21st Avenue South, Nashville, TN, 37232-4313, USA.
| |
Collapse
|
27
|
Zhang Y, He X, Xiong X, Chuan J, Zhong L, Chen G, Yu D. The association between maternal methylenetetrahydrofolate reductase C677T and A1298C polymorphism and birth defects and adverse pregnancy outcomes. Prenat Diagn 2018; 39:3-9. [PMID: 30474229 DOI: 10.1002/pd.5396] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 11/18/2018] [Accepted: 11/21/2018] [Indexed: 01/03/2023]
Abstract
Published studies indicate the MTHFR C677T and A1298C polymorphisms are associated with abnormal homocysteine levels, which may cause various pregnancy complications and birth defects. However, the results obtained from different studies have been inconsistent. Therefore, this meta-analysis explores the association between MTHFR polymorphisms and birth defects and adverse pregnancy outcomes. The PubMed, ScienceDirect, Embase, and China Biology Medicine literature databases and ClinicalTrials were searched. Analyses of public bias, meta-regression, subgroups, and sensitivity were used to ensure the robustness of our results. MTHFR C677T was significantly associated with recurrent pregnancy loss in developing countries (odds ratio [OR], 1.34; 95% confidence interval [CI], 1.20-1.50) but not in developed countries (OR, 0.87; 95% CI, 0.68-1.11). No significant relationship was found between MTHFR A1298C and recurrent pregnancy loss (OR, 1.04; 95% CI, 0.93-1.18). MTHFR C677T and A1298C were not associated with preeclampsia (OR, 1.06; 95% CI, 0.97-1.16 and OR, 1.16; 95% CI, 0.97-1.39, respectively), and C677T was not associated with placental abruption (OR, 1.03; 95% CI, 0.87-1.21), intrauterine growth retardation (OR, 1.02; 95% CI, 0.90-1.15), or congenital heart disease (OR, 1.05; 95% CI, 0.89-1.25). MTHFR C677T, but not A1298C, was associated with neural tube defects (OR, 1.24; 95% CI, 1.08-1.42) and Down syndrome (OR, 1.65; 95% CI, 1.39-1.95). CONCLUSION: Although MTHFR C677T and A1298C are significantly associated with some types of congenital defects and adverse pregnancy outcomes, the impact of these polymorphisms is moderate.
Collapse
Affiliation(s)
- Yuan Zhang
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, PR China
| | - Xia He
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, PR China
| | - Xuan Xiong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, PR China
| | - Junlan Chuan
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, PR China
| | - Lei Zhong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, PR China
| | - Guo Chen
- Department of Geriatrics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, PR China
| | - Dongke Yu
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, PR China
| |
Collapse
|
28
|
Ryan DP, Henzel KS, Pearson BL, Siwek ME, Papazoglou A, Guo L, Paesler K, Yu M, Müller R, Xie K, Schröder S, Becker L, Garrett L, Hölter SM, Neff F, Rácz I, Rathkolb B, Rozman J, Ehninger G, Klingenspor M, Klopstock T, Wolf E, Wurst W, Zimmer A, Fuchs H, Gailus-Durner V, Hrabě de Angelis M, Sidiropoulou K, Weiergräber M, Zhou Y, Ehninger D. A paternal methyl donor-rich diet altered cognitive and neural functions in offspring mice. Mol Psychiatry 2018; 23:1345-1355. [PMID: 28373690 PMCID: PMC5984088 DOI: 10.1038/mp.2017.53] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 02/13/2017] [Accepted: 02/14/2017] [Indexed: 12/17/2022]
Abstract
Dietary intake of methyl donors, such as folic acid and methionine, shows considerable intra-individual variation in human populations. While it is recognized that maternal departures from the optimum of dietary methyl donor intake can increase the risk for mental health issues and neurological disorders in offspring, it has not been explored whether paternal dietary methyl donor intake influences behavioral and cognitive functions in the next generation. Here, we report that elevated paternal dietary methyl donor intake in a mouse model, transiently applied prior to mating, resulted in offspring animals (methyl donor-rich diet (MD) F1 mice) with deficits in hippocampus-dependent learning and memory, impaired hippocampal synaptic plasticity and reduced hippocampal theta oscillations. Gene expression analyses revealed altered expression of the methionine adenosyltransferase Mat2a and BK channel subunit Kcnmb2, which was associated with changes in Kcnmb2 promoter methylation in MD F1 mice. Hippocampal overexpression of Kcnmb2 in MD F1 mice ameliorated altered spatial learning and memory, supporting a role of this BK channel subunit in the MD F1 behavioral phenotype. Behavioral and gene expression changes did not extend into the F2 offspring generation. Together, our data indicate that paternal dietary factors influence cognitive and neural functions in the offspring generation.
Collapse
Affiliation(s)
- D P Ryan
- Molecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - K S Henzel
- Molecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - B L Pearson
- Molecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - M E Siwek
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany
| | - A Papazoglou
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany
| | - L Guo
- Department of Physiology, Medical College of Qingdao University, Qingdao, Shandong, China
| | - K Paesler
- Molecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - M Yu
- Department of Physiology, Medical College of Qingdao University, Qingdao, Shandong, China
| | - R Müller
- Department of Psychiatry and Psychotherapy, University of Cologne, Faculty of Medicine, Cologne, Germany
| | - K Xie
- Molecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - S Schröder
- Molecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - L Becker
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany,Friedrich-Baur-Institut, Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - L Garrett
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany,Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - S M Hölter
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany,Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - F Neff
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany,Institute of Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - I Rácz
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn, Germany
| | - B Rathkolb
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany,Chair of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany,Member of German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - J Rozman
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany,Member of German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - G Ehninger
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - M Klingenspor
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center, Technische Universität München, Freising-Weihenstephan, Germany
| | - T Klopstock
- Friedrich-Baur-Institut, Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany,German Center for Vertigo and Balance Disorders, University Hospital Munich, Campus Grosshadern, Munich, Germany,DZNE, German Center for Neurodegenerative Diseases, Munich, Germany,Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Munich, Germany
| | - E Wolf
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - W Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany,DZNE, German Center for Neurodegenerative Diseases, Munich, Germany,Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Munich, Germany,Chair of Developmental Genetics, Technische Universität München, c/o Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - A Zimmer
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn, Germany
| | - H Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - V Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - M Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany,Member of German Center for Diabetes Research (DZD), München-Neuherberg, Germany,Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
| | - K Sidiropoulou
- Department of Biology, University of Crete, Vassilika Vouton, Heraklio, Greece
| | - M Weiergräber
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany
| | - Y Zhou
- Department of Physiology, Medical College of Qingdao University, Qingdao, Shandong, China
| | - D Ehninger
- Molecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany,Molecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, Bonn 53127, Germany. E-mail:
| |
Collapse
|
29
|
Roy A, Kocak M, Hartman TJ, Vereen S, Adgent M, Piyathilake C, Tylavsky FA, Carroll KN. Association of prenatal folate status with early childhood wheeze and atopic dermatitis. Pediatr Allergy Immunol 2018; 29:144-150. [PMID: 29168294 PMCID: PMC6087709 DOI: 10.1111/pai.12834] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/17/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND Prenatal folic acid supplementation is recommended to prevent birth defects. Some foods are fortified in the USA to ensure sufficient intake among reproductive-aged women. However, high prenatal folate exposure may be a risk factor for childhood atopic diseases. We investigated associations between prenatal folate and early childhood wheeze and atopic dermatitis in a US cohort. METHODS We studied 858 mother-child dyads, enrolled prenatally. Folate was measured in 2nd and 3rd trimester maternal plasma. Parents reported current wheeze (previous 12 months) and healthcare provider diagnosis of atopic dermatitis at 3 years. We examined associations using logistic regression, modeling folate continuously and dichotomously (< or ≥20 ng/mL), a level often considered supraphysiologic. RESULTS Over half of women were African American and on Medicaid. Median (interquartile range) folate levels were 22.6 (15.9-30.0) and 23.1 (16.1-30.0) ng/mL for 2nd and 3rd trimesters, respectively. Current wheeze and atopic dermatitis were reported for 20.4% and 26.8% of children, respectively. Second trimester folate as a continuous exposure was not significantly associated with outcomes. Decreased odds of current wheeze were observed in children born to mothers who had 2nd trimester folate ≥20 ng/mL (adjusted odds ratios = 0.67, 95% confidence interval = 0.46, 0.97) compared to children with maternal levels <20 ng/mL. Third trimester folate was not associated with outcomes. CONCLUSIONS High plasma folate in mid-pregnancy was associated with decreased odds of current wheeze at age 3. Our findings do not support harmful effects of high prenatal folate levels on childhood atopic diseases in this setting.
Collapse
Affiliation(s)
- A Roy
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of General Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Asthma and Environmental Health Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Kocak
- Department of Preventive Medicine, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - T J Hartman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - S Vereen
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of General Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Asthma and Environmental Health Research, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Community and Family Health, College of Public Health, University of South Florida, Tampa, Florida
| | - M Adgent
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of General Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Asthma and Environmental Health Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - C Piyathilake
- Department of Nutritional Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - F A Tylavsky
- Department of Preventive Medicine, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - K N Carroll
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of General Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Asthma and Environmental Health Research, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
30
|
den Dekker HT, Jaddoe VWV, Reiss IK, de Jongste JC, Duijts L. Maternal folic acid use during pregnancy, methylenetetrahydrofolate reductase gene polymorphism, and child's lung function and asthma. Clin Exp Allergy 2017; 48:175-185. [PMID: 29117460 DOI: 10.1111/cea.13056] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 09/18/2017] [Accepted: 10/14/2017] [Indexed: 11/27/2022]
Abstract
BACKGROUND Folic acid supplement use during pregnancy might affect childhood respiratory health, potentially mediated by methylenetetrahydrofolate reductase polymorphism C677T (MTHFR-C677T) carriership. OBJECTIVES We examined the associations of maternal folic acid supplement use and folate, vitamin B12 and homocysteine concentrations during pregnancy with childhood lung function and asthma. METHODS This study was embedded in a population-based prospective cohort study among 5653 children. Folic acid supplement use was assessed by questionnaires. Folate, vitamin B12 and homocysteine plasma concentrations were measured in early pregnancy and at birth. At age 10 years, forced expiratory volume in 1 second (FEV1 ), forced vital capacity (FVC), FEV1 /FVC, forced expiratory flow between 25% and 75% (FEF25-75 ), at 75% of FVC (FEF75 ), and asthma were examined. RESULTS Maternal folic acid supplement use during pregnancy was associated with higher childhood FEV1 and FVC and with a lower FEV1 /FVC, compared with no folic acid supplement use. Among mothers carrying MTHFR-C677T variants, preconceptional start of folic acid supplement use was associated with lower FEV1 /FVC (-0.17 [-0.32, -0.02]) and FEF25-75 (-0.24 [-0.40, -0.07]). Among children carrying MTHFR-C677T wild-type, a higher vitamin B12 level at birth was associated with a lower FEV1 (-0.07 [-0.12, -0.01]) and FVC (-0.09 [-0.15, -0.04]). Folate and homocysteine concentrations were not consistently associated with lower childhood lung function or asthma. CONCLUSIONS Preconceptional start of maternal folic acid supplement use and higher vitamin B12 concentrations at birth might adversely affect childhood lung function depending on MTHFR-C677T carriership. The clinical implications need to be evaluated.
Collapse
Affiliation(s)
- H T den Dekker
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - V W V Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - I K Reiss
- Department of Pediatrics, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - J C de Jongste
- Department of Pediatrics, Division of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - L Duijts
- Department of Pediatrics, Division of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
31
|
Silva C, Keating E, Pinto E. The impact of folic acid supplementation on gestational and long term health: Critical temporal windows, benefits and risks. Porto Biomed J 2017; 2:315-332. [PMID: 32258789 DOI: 10.1016/j.pbj.2017.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 05/10/2017] [Accepted: 05/31/2017] [Indexed: 01/23/2023] Open
Abstract
Highlights Associations between FA supplementation in pregnancy and effects on offspring's NTDs, allergy/respiratory problems, cancer and behaviour problems as been studied.There is growing concern about the effects of excessive FA supplementation, whether in terms of doses or times of exposure.FA supplementation in the periconceptional period is protective against NTDs while in later periods it could be deleterious.A daily dose of 0.4 mg FA in the periconceptional period seems to be effective and safe. Abstract Maternal folic acid (FA) supplementation is one of the most popular nutritional interventions during pregnancy for its protective effect against neural tube defects (NTDs).The purposes of this review are: (a) to gather the current evidence regarding supplementation of maternal diet with FA and (b) to problematize the available literature in terms of dosages, critical temporal windows, and its potential benefits and risks.The expression (pregnancy OR fetus OR offspring OR mother) AND ("folic acid" AND supplementation) was searched on PubMed database, filtering for articles published from 2005 to 2014. Publications referring to FA supplementation during the periconceptional period or pregnancy in which there was a conclusion about the effects of isolated FA supplementation on pregnant woman, pregnancy or offspring were included. Of the initial 1182 papers, 109 fulfilled the inclusion criteria.The majority of the publications reported FA supplementation outcomes on offspring's health, with emphasis in NTDs, allergy/respiratory problems, cancer and behaviour problems. Some inconsistency is observed on the impact of FA supplementation on different outcomes, except for NTDs. It is also visible an increased concern about the impact of excessive supplementation, either in terms of doses or exposure's duration.In conclusion, there is a growing interest in FA supplementation issues. The protective effect of FA supplementation over NTDs has been confirmed, being the periconceptional period a critical window, and it is frequently suggested that allergy/respiratory outcomes arise from (excessive) FA supplementation particularly later in pregnancy. Further research on critical doses and time of exposure should be conducted.
Collapse
Affiliation(s)
- Carla Silva
- Faculty of Medicine of University of Porto, Porto, Portugal
| | - Elisa Keating
- Department of Biomedicine - Biochemistry Unit, Faculty of Medicine of University of Porto, Porto, Portugal.,CINTESIS - Center for Health Technology and Services Research, Porto, Portugal
| | - Elisabete Pinto
- CBQF - Centre of Biotechnology and Fine Chemistry - School of Biotechnology, Portuguese Catholic University, Porto, Portugal.,Institute of Public Health of the University of Porto, Porto, Portugal
| |
Collapse
|
32
|
High-dose maternal folic acid supplementation before conception impairs reversal learning in offspring mice. Sci Rep 2017; 7:3098. [PMID: 28596566 PMCID: PMC5465191 DOI: 10.1038/s41598-017-03158-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 04/25/2017] [Indexed: 01/21/2023] Open
Abstract
Maternal folic acid (FA) supplementation prior to and during gestation is recommended for the prevention of neural tube closure defects in the developing embryo. Prior studies, however, suggested that excessive FA supplementation during gestation can be associated with toxic effects on the developing organism. Here, we address whether maternal dietary folic acid supplementation at 40 mg/kg chow (FD), restricted to a period prior to conception, affects neurobehavioural development in the offspring generation. Detailed behavioural analyses showed reversal learning impairments in the Morris water maze in offspring derived from dams exposed to FD prior to conceiving. Furthermore, offspring of FD dams showed minor and transient gene expression differences relative to controls. Our data suggest that temporary exposure of female germ cells to FD is sufficient to cause impaired cognitive flexibility in the subsequent generation.
Collapse
|
33
|
Pali-Schöll I, Namazy J, Jensen-Jarolim E. Allergic diseases and asthma in pregnancy, a secondary publication. World Allergy Organ J 2017; 10:10. [PMID: 28286601 PMCID: PMC5333384 DOI: 10.1186/s40413-017-0141-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 01/27/2017] [Indexed: 02/06/2023] Open
Abstract
Every fifth pregnant woman is affected by allergies, especially rhinitis and asthma. Allergic symptoms existing before pregnancy may be either attenuated, or equally often promoted through pregnancy. Optimal allergy and asthma diagnosis and management during pregnancy is vital to ensure the welfare of mother and baby. For allergy diagnosis in pregnancy, preferentially anamnestic investigation as well as in vitro testing should be applied, whereas skin testing or provocation tests should be postponed until after birth. Pregnant women with confirmed allergy should avoid exposure to, or consumption of the offending allergen. Allergen immunotherapy should not be initiated during pregnancy. In patients on immunotherapy since before pregnancy, maintenance treatment may be continued, but the allergen dose should not be increased further. Applicable medications for asthma, rhinitis or skin symptoms in pregnancy are discussed and listed. In conclusion, i) allergies in pregnancy should preferentially be diagnosed in vitro; ii) AIT may be continued, but not started, and symptomatic medications must be carefully selected; iii) management of asthma and allergic diseases is important during pregnancy for welfare of mother and child.
Collapse
Affiliation(s)
- Isabella Pali-Schöll
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Veterinärplatz 1, 1210 Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Physiology, Pathophysiology and Immunology, Medical University Vienna, Vienna, Austria
| | - Jennifer Namazy
- Scripps Clinic, 7565 Mission Valley Rd Ste 200, San Diego, CA 92108 USA
| | - Erika Jensen-Jarolim
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Veterinärplatz 1, 1210 Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Physiology, Pathophysiology and Immunology, Medical University Vienna, Vienna, Austria
- AllergyCare, Allergy Diagnosis and Study Center Vienna, Vienna, Austria
| |
Collapse
|
34
|
McStay CL, Prescott SL, Bower C, Palmer DJ. Maternal Folic Acid Supplementation during Pregnancy and Childhood Allergic Disease Outcomes: A Question of Timing? Nutrients 2017; 9:nu9020123. [PMID: 28208798 PMCID: PMC5331554 DOI: 10.3390/nu9020123] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 02/03/2017] [Indexed: 12/17/2022] Open
Abstract
Since the early 1990s, maternal folic acid supplementation has been recommended prior to and during the first trimester of pregnancy, to reduce the risk of infant neural tube defects. In addition, many countries have also implemented the folic acid fortification of staple foods, in order to promote sufficient intakes amongst women of a childbearing age, based on concerns surrounding variable dietary and supplementation practices. As many women continue to take folic acid supplements beyond the recommended first trimester, there has been an overall increase in folate intakes, particularly in countries with mandatory fortification. This has raised questions on the consequences for the developing fetus, given that folic acid, a methyl donor, has the potential to epigenetically modify gene expression. In animal studies, folic acid has been shown to promote an allergic phenotype in the offspring, through changes in DNA methylation. Human population studies have also described associations between folate status in pregnancy and the risk of subsequent childhood allergic disease. In this review, we address the question of whether ongoing maternal folic acid supplementation after neural tube closure, could be contributing to the rise in early life allergic diseases.
Collapse
Affiliation(s)
- Catrina L McStay
- Department of Health Western Australia, Perth 6004, Western Australia, Australia.
| | - Susan L Prescott
- School of Paediatrics and Child Health, The University of Western Australia, Subiaco 6008, Western Australia, Australia.
- Telethon Kids Institute, The University of Western Australia, Subiaco 6008, Western Australia, Australia.
- Members of the in-FLAME International Inflammation Network, Perth 6000, Western Australia, Australia.
| | - Carol Bower
- Telethon Kids Institute, The University of Western Australia, Subiaco 6008, Western Australia, Australia.
| | - Debra J Palmer
- School of Paediatrics and Child Health, The University of Western Australia, Subiaco 6008, Western Australia, Australia.
- Telethon Kids Institute, The University of Western Australia, Subiaco 6008, Western Australia, Australia.
- Members of the in-FLAME International Inflammation Network, Perth 6000, Western Australia, Australia.
| |
Collapse
|
35
|
Liang Y, Chang C, Lu Q. The Genetics and Epigenetics of Atopic Dermatitis-Filaggrin and Other Polymorphisms. Clin Rev Allergy Immunol 2017; 51:315-328. [PMID: 26385242 DOI: 10.1007/s12016-015-8508-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease caused by a combination of genetic and environmental factors. Genetic evidences depict a complex network comprising by epidermal barrier dysfunctions and dysregulation of innate and adaptive immunity in the pathogenesis of AD. Mutations in the human filaggrin gene (FLG) are the most significant and well-replicated genetic mutation associated with AD, and other mutations associated with epidermal barriers such as SPINK5, FLG-2, SPRR3, and CLDN1 have all been linked to AD. Gene variants may also contribute to the abnormal innate and adaptive responses found in AD, including mutations in PRRs and AMPs, TSLP and TSLPR, IL-1 family cytokines and receptors genes, vitamin D pathway genes, FCER1A, and Th2 and other cytokines genes. GWAS and Immunochip analysis have identified a total of 19 susceptibility loci for AD. Candidate genes at these susceptibility loci identified by GWAS and Immunochip analysis also suggest roles for epidermal barrier functions, innate and adaptive immunity, interleukin-1 family signaling, regulatory T cells, the vitamin D pathway, and the nerve growth factor pathway in the pathogenesis of AD. Increasing evidences show the modern lifestyle (i.e., the hygiene hypothesis, Western diet) and other environmental factors such as pollution and environmental tobacco smoke (ETS) lead to the increasing prevalence of AD with the development of industrialization. Epigenetic alterations in response to these environmental factors, including DNA methylation and microRNA related to immune system and skin barriers, have been found to contribute to the pathogenesis of AD. Genetic variants and epigenetic alteration might be the key tools for the molecular taxonomy of AD and provide the background for the personalized management.
Collapse
Affiliation(s)
- Yunsheng Liang
- Hunan Key Laboratory of Medical Epigenomics & Department of Dermatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Changsha, Hunan, 410011, China
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA, 95616, USA
| | - Qianjin Lu
- Hunan Key Laboratory of Medical Epigenomics & Department of Dermatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Changsha, Hunan, 410011, China.
| |
Collapse
|
36
|
Doğruel D, Bingöl G, Altıntaş DU, Yılmaz M, Kendirli SG. Prevalence of and risk factors for atopic dermatitis: A birth cohort study of infants in southeast Turkey. Allergol Immunopathol (Madr) 2016; 44:214-20. [PMID: 26589340 DOI: 10.1016/j.aller.2015.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 06/18/2015] [Accepted: 07/31/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is most common in the first year of life. The aim of this study was to determine the prevalence of and risk factors for AD in a birth cohort of infants from southeast Turkey. METHODS Adana Paediatric Allergy Research (ADAPAR) birth cohort study was derived from 1377 infants who were born in Cukurova University, Medical Hospital, Adana, Turkey between February 2010 and February 2011. At birth, a physical examination was performed, cord blood samples were taken, and the mother completed a baseline questionnaire that provided data on gestational conditions, family history of allergic diseases and environmental exposures. Follow-up visits scheduled at 3, 6, and 12 months included an infant physical examination and an extended questionnaire. Skin prick test was performed and food-specific IgE levels were measured at 6 and 12 months. Atopic dermatitis was diagnosed based on confirmatory examination by a physician. RESULTS Of the 1377 infants enrolled, 59 (4.3%) were diagnosed with AD as of 12 months. Maternal allergic disease (ORs 6.28, 95% CI 1.03-38.30; p=0.046), maternal infection during gestation (ORs 3.73, 95% CI 1.25-11.09; p=0.018), and presence of food allergy (ORs 13.7, 95% CI 3.07-61.0; p=0.001) were identified as risk factors for AD. Breastfeeding and cord blood IgE levels were not identified as risk factors. CONCLUSIONS In this cohort we found prevalence of AD as 4.3% during the first year of life. Positive family history of atopic diseases, prenatal infections and presence of food allergy are the risk factors for early presentation of AD.
Collapse
Affiliation(s)
- D Doğruel
- Department of Pediatric Allergy and Immunology, Çukurova University Faculty of Medicine, Adana, Turkey.
| | - G Bingöl
- Department of Pediatric Allergy and Immunology, Çukurova University Faculty of Medicine, Adana, Turkey
| | - D U Altıntaş
- Department of Pediatric Allergy and Immunology, Çukurova University Faculty of Medicine, Adana, Turkey
| | - M Yılmaz
- Department of Pediatric Allergy and Immunology, Çukurova University Faculty of Medicine, Adana, Turkey
| | - S G Kendirli
- Department of Pediatric Allergy and Immunology, Çukurova University Faculty of Medicine, Adana, Turkey
| |
Collapse
|
37
|
Huot PSP, Ly A, Szeto IMY, Reza-López SA, Cho D, Kim YI, Anderson GH. Maternal and postweaning folic acid supplementation interact to influence body weight, insulin resistance, and food intake regulatory gene expression in rat offspring in a sex-specific manner. Appl Physiol Nutr Metab 2015; 41:411-20. [PMID: 26989972 DOI: 10.1139/apnm-2015-0503] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Maternal intake of multivitamins or folic acid above the basal dietary requirement alters the growth and metabolic trajectory of rat offspring. We hypothesized that a modest increase in the folic acid content of maternal diets would alter the offspring's metabolic phenotype, and that these effects could be corrected by matching the folic acid content of the offspring's diet with that of the maternal diet. Female Sprague-Dawley rats were placed on a control or a 2.5× folic acid-supplemented diet prior to mating and during pregnancy and lactation. At weaning, pups from each maternal diet group were randomized to the control or to the 2.5× folic acid-supplemented diet for 25 weeks. Male pups from dams fed the folic acid-supplemented diet were 3.7% heavier than those from control-fed dams and had lower mRNA expression for leptin receptor Obrb isoform (Lepr) (11%) and Agouti-related protein (Agrp) (14%). In contrast, female pups from folic acid-supplemented dams were 5% lighter than those from control-fed dams and had lower proopiomelanocortin (Pomc) (42%), Lepr (32%), and Agrp (13%), but higher neuropeptide Y (Npy) (18%) mRNA expression. Folic acid supplementation ameliorated the alterations induced by maternal folic acid supplementation in male pups and led to the lowest insulin resistance, but the effects were smaller in female pups and led to the highest insulin resistance. In conclusion, maternal folic acid supplementation at 2.5× the control level was associated with alterations in body weight and hypothalamic gene expression in rat offspring in a sex-specific manner, and some of these effects were attenuated by postweaning folic acid supplementation.
Collapse
Affiliation(s)
- Pedro S P Huot
- a Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Anna Ly
- a Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Ignatius M Y Szeto
- a Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Sandra A Reza-López
- a Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Daniel Cho
- a Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Young-In Kim
- a Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 3E2, Canada.,b Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON M5G 2C4, Canada.,c Keenan Research Centre for Biomedical Sciences of St. Michael's Hospital, Toronto, ON M5B 1W8, Canada.,d Division of Gastroenterology, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada
| | - G Harvey Anderson
- a Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 3E2, Canada
| |
Collapse
|
38
|
Dogru M, Aydin H, Aktas A, Cırık A. Methylenetetrahydrofolate Reductase gene polymorphism in children with allergic rhinitis. Allergol Immunopathol (Madr) 2015; 43:579-83. [PMID: 25796308 DOI: 10.1016/j.aller.2014.11.001] [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: 09/04/2014] [Revised: 11/07/2014] [Accepted: 11/11/2014] [Indexed: 10/23/2022]
Abstract
BACKGROUND Methylenetetrahydrofolate Reductase (MTHFR) polymorphisms by impairing folate metabolism may influence the development of allergic diseases. The results of studies evaluating the relationship between MTHFR polymorphisms and atopic disease are controversial. The aim of this study was to investigate the association between the polymorphisms of C677T and A1298C for MTHFR gene and allergic rhinitis (AR) in children. METHODS Ninety patients followed up with diagnosis of allergic rhinitis in our clinic and 30 children with no allergic diseases were included in the study. All participants were genotyped for the MTHFR (C677T) and (A1298C) polymorphisms. Vitamin b12, folate and homocysteine levels were measured. RESULTS The mean age of patients was 9.2±2.9 years; 66.7% of the patients were male. There was no significant difference between patient and control groups regarding gender, age and atopy history of the family (p>0.05). The frequency of homozygotes for MTHFR C677T polymorphism in the patient and control groups was 3.3% and 10%, respectively. The frequency of homozygotes for MTHFR A1298C polymorphism among groups was 26.7% and 16.7%, respectively. The association between allergic rhinitis and polymorphisms of C677T and A1298C for MTHFR gene was not statistically significant in patients compared with controls (p>0.05). There were no statistically significant differences between the patients and the control group in terms of serum vitamin b12, folate and homocysteine levels (p>0.05). CONCLUSION We found no evidence for an association between allergic rhinitis and polymorphisms of C677T and A1298C for MTHFR gene in children. Further studies investigating the relationship between MTHFR polymorphism and AR are required.
Collapse
|
39
|
Chan YM, MacFarlane AJ, O'Connor DL. Modeling Demonstrates That Folic Acid Fortification of Whole-Wheat Flour Could Reduce the Prevalence of Folate Inadequacy in Canadian Whole-Wheat Consumers. J Nutr 2015; 145:2622-9. [PMID: 26423740 DOI: 10.3945/jn.115.217851] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 09/02/2015] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Mandatory folic acid fortification of white-wheat flour and selected other grain products has reduced the prevalence of neural tube defects in Canada; however, the fortification of whole-wheat flour is not permitted. OBJECTIVE The objective of this study was to model the impact of adding folic acid to whole-wheat flour on the folate intake distribution of Canadians. METHODS Twenty-four-hour dietary recall and supplement intake data (n = 35,107) collected in the 2004 Canadian Community Health Survey 2.2 were used to calculate the prevalence of folate inadequacy (POFI) and the proportion of folic acid intakes above the Tolerable Upper Intake Level (UL). In model 1, folic acid was added to whole-wheat flour-containing foods in amounts comparable to those that are mandatory for white-wheat flour-containing foods. In model 2, a 50% overage of folic acid fortification was considered. Models 3 and 4 included assessment of folate intake distributions in adult whole-wheat consumers with or without a fortification overage. SIDE (Software for Intake Distribution Estimation; Department of Statistics and Center for Agricultural and Rural Development, Iowa State University) was used to estimate usual folate intakes. RESULTS Mean folate intakes increased by ∼ 5% in all sex and age groups when whole-wheat foods were fortified (models 1 and 2; P < 0.0001). Folic acid fortification of whole-wheat flour-containing foods did not change the POFI or percentage of intakes above the UL in the general population, whether in supplement users or nonusers. Among whole-wheat consumers, the POFI was reduced by 10 percentage points after fortification of whole-wheat flour-containing foods (95% CIs did not overlap). The percentage of whole-wheat consumers with intakes above the UL did not change. CONCLUSION Although folic acid fortification of whole-wheat flour-containing foods is unlikely to change the POFI or proportion of folic acid intakes above the UL in the general Canadian population, this fortification strategy may reduce the POFI in adult whole-wheat consumers.
Collapse
Affiliation(s)
- Yen-Ming Chan
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada; The Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; and
| | | | - Deborah L O'Connor
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada; The Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; and
| |
Collapse
|
40
|
Miles EA, Calder PC. Maternal diet and its influence on the development of allergic disease. Clin Exp Allergy 2015; 45:63-74. [PMID: 25394813 DOI: 10.1111/cea.12453] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The early presentation of childhood allergies and the rise in their prevalence suggest that changes in early-life exposures may increase the predisposition. Very early-life exposures may act upon the developing foetal immune system and include infection, environmental tobacco smoke, other pollutants and nutrients provided via the mother. Three nutrients have come under close scrutiny: vitamin D, omega 3 polyunsaturated fatty acids (PUFAs) and folate (or the synthetic form, folic acid). Much of the data on these nutrients are observational although some randomised, placebo-controlled trials have been conducted with omega 3 PUFAs and one with vitamin D. Some studies with omega 3 PUFA supplements in pregnancy have demonstrated immunomodulatory effects on the neonate and a reduction in risk of early sensitisation to allergens. A few studies with omega 3 polyunsaturated fatty acid supplements in pregnancy have shown a reduction in proportion of children affected by allergic symptoms (food allergy) or in symptom severity (atopic dermatitis). Observational studies investigating the association of maternal vitamin D intake or maternal or neonatal vitamin D status have been inconsistent. One randomised, controlled trial of vitamin D supplementation during pregnancy did not show any significant effect on allergic outcome in the offspring. Studies investigating the association between maternal folic acid or folate intake or maternal or neonatal folate status and offspring risk of allergic disease have been equivocal. Further evidence is required to clarify whether increased intake of these nutrients during pregnancy influences allergic disease in the offspring. In the light of current evidence, mothers should not either increase or avoid consuming these nutrients to prevent or ameliorate allergic disease in their offspring. However, these essential nutrients each have important roles in foetal development. This is reflected in current government recommendations for intake of these nutrients by pregnant women.
Collapse
Affiliation(s)
- E A Miles
- Human Development & Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | | |
Collapse
|
41
|
Masih SP, Plumptre L, Ly A, Berger H, Lausman AY, Croxford R, Kim YI, O'Connor DL. Pregnant Canadian Women Achieve Recommended Intakes of One-Carbon Nutrients through Prenatal Supplementation but the Supplement Composition, Including Choline, Requires Reconsideration. J Nutr 2015; 145:1824-34. [PMID: 26063067 DOI: 10.3945/jn.115.211300] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 05/19/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Folate, vitamin B-6, vitamin B-12, and choline are involved in one-carbon metabolism and play critical roles in pregnancy including prevention of birth defects and promotion of neurodevelopment. However, excessive intakes may adversely affect disease susceptibility in offspring. Intakes of these nutrients during pregnancy are not well characterized. OBJECTIVE Our aim was to determine dietary and supplemental intakes and major dietary sources of one-carbon nutrients during pregnancy. METHODS In pregnant women (n = 368) at ≤16 wk postconception, supplement use >30 d before pregnancy was assessed by maternal recall and supplement and dietary intakes in early (0-16 wk) and late pregnancy (23-37 wk) were assessed by food-frequency questionnaire. RESULTS Preconception, 60.1% (95% CI: 55.8, 64.3) of women used B vitamin-containing supplements. This increased to 92.8% (95% CI: 89.6, 95.2) in early and 89.0% (95% CI: 85.0, 92.3) in late pregnancy. Median supplemental folic acid, vitamin B-12, and vitamin B-6 were 1000 μg/d, 2.6 μg/d, and 1.9 mg/d, respectively. Forty-one percent and 50% of women had dietary intakes of folate and vitamin B-6 less than the estimated average requirement (520 mg/d dietary folate equivalents and 1.6 mg/d, respectively). Eight-seven percent of women had choline intakes less than the Adequate Intake (450 mg/d). Dietary intakes did not change appreciably during pregnancy. Fruits and vegetables and fortified foods contributed ∼57% to total dietary folate intake. Fruits and vegetables contributed ∼32% to total dietary vitamin B-6 intake and dairy and egg products contributed ∼37% to total dietary vitamin B-12 intake. CONCLUSIONS Vitamin supplements were an important source of one-carbon nutrients during pregnancy in our sample. Without supplements, many women would not have consumed quantities of folate and vitamin B-6 consistent with recommendations. Given the importance of choline in pregnancy, further research to consider inclusion in prenatal supplements is warranted. This trial was registered at clinicaltrials.gov as NCT02244684.
Collapse
Affiliation(s)
- Shannon P Masih
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Keenan Research Center for Biomedical Science
| | - Lesley Plumptre
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Keenan Research Center for Biomedical Science
| | - Anna Ly
- Keenan Research Center for Biomedical Science
| | - Howard Berger
- Keenan Research Center for Biomedical Science, Departments of Obstetrics and Gynecology and
| | - Andrea Y Lausman
- Keenan Research Center for Biomedical Science, Departments of Obstetrics and Gynecology and
| | - Ruth Croxford
- freelance statistics consultant, Toronto, Canada; and
| | - Young-In Kim
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Keenan Research Center for Biomedical Science, Medicine, St. Michael's Hospital, Toronto, Canada
| | - Deborah L O'Connor
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Research Institute, The Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
42
|
Wang T, Zhang HP, Zhang X, Liang ZA, Ji YL, Wang G. Is Folate Status a Risk Factor for Asthma or Other Allergic Diseases? ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2015; 7:538-46. [PMID: 26333700 PMCID: PMC4605926 DOI: 10.4168/aair.2015.7.6.538] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 05/03/2015] [Accepted: 05/12/2015] [Indexed: 02/05/2023]
Abstract
PURPOSE It is controversial whether folate status is a risk factor for the development of asthma or other allergic diseases. This study was conducted to investigate whether indirect or direct exposure to folate and impaired folate metabolism, reflected as methylene-tetrahydrofolate reductase (MTHFR) C677T polymorphism, would contribute to the development of asthma and other allergic diseases. METHODS Electronic databases were searched to identify all studies assessing the association between folate status and asthma or other allergic diseases. Two reviewers independently assessed the eligibility of studies and extracted data. The relative risk (RR) or odds ratio (OR) with 95% confidence intervals (CI) was calculated and pooled. RESULTS Twenty-six studies (16 cohort, 7 case-control, and 3 cross-sectional studies) were identified. Maternal folic acid supplementation was not associated with the development of asthma, atopic dermatitis (AD), eczema, and sensitization in the offspring, whereas exposure during early pregnancy was related to wheeze occurrence in the offspring (RR=1.06, 95% CI=[1.02-1.09]). The TT genotype of MTHFR C677T polymorphism was at high risk of asthma (OR=1.41, 95% CI=[1.07-1.86]). CONCLUSIONS It is indicated that maternal folic acid supplementation during early pregnancy may increase the risk of wheeze in early childhood and that the TT genotype of MTHFR C677T polymorphism impairing folic acid metabolism would be at high risk of asthma development. These results might provide additional information for recommendations regarding forced folate consumption or folic acid supplements during pregnancy based on its well-established benefits for the prevention of congenital malformations. However, currently available evidence is of low quality which is needed to further elucidate.
Collapse
Affiliation(s)
- Ting Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Hong Ping Zhang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy of China, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xin Zhang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy of China, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Zong An Liang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yu Lin Ji
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Gang Wang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy of China, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
43
|
Colapinto CK, O'Connor DL, Sampson M, Williams B, Tremblay MS. Systematic review of adverse health outcomes associated with high serum or red blood cell folate concentrations. J Public Health (Oxf) 2015; 38:e84-97. [PMID: 26160024 DOI: 10.1093/pubmed/fdv087] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND To examine the relationship between reported high serum or red blood cell (RBC) folate status and adverse health outcomes. METHODS We systematically searched PubMed/Medline and EMBASE (to May 2013), with no limits by study type, country or population, to identify studies reporting high folate concentrations in association with adverse health outcomes. Two reviewers screened studies and extracted data. Study quality was assessed. RESULTS We included 51 articles, representing 46 studies and 71 847 participants. Quantiles were used by 96% of studies to identify high folate concentrations. Eighty-three percent of serum folate and 50% of RBC folate studies reported a high folate cutoff that corresponded with a clinically normal concentration. Increasing values of reported high folate concentration did not demonstrate a consistent association with risk of adverse health outcomes. Overall, reported high folate concentrations appeared to be associated with a decreased risk of adverse health outcomes, though substantial methodological heterogeneity precluded complex analyses. CONCLUSIONS Our interpretation was complicated by methodological variability. High folate cutoffs varied and often corresponded with normal or desirable blood concentrations. In general, a negative association appeared to exist between reported high folate status and adverse health outcomes. Consistent methods and definitions are needed to examine high folate status and ultimately inform public health interventions.
Collapse
Affiliation(s)
- Cynthia K Colapinto
- Healthy Active Living and Obesity Research, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada University of Ottawa, Ottawa, Ontario, Canada
| | - Deborah L O'Connor
- The Hospital for Sick Children, Toronto, Ontario, Canada University of Toronto, Toronto, Ontario, Canada
| | - Margaret Sampson
- Healthy Active Living and Obesity Research, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
| | - Brock Williams
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mark S Tremblay
- Healthy Active Living and Obesity Research, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
44
|
Kim JH, Jeong KS, Ha EH, Park H, Ha M, Hong YC, Bhang SY, Lee SJ, Lee KY, Lee SH, Kim Y, Kim MH, Chang N. Relationship between prenatal and postnatal exposures to folate and risks of allergic and respiratory diseases in early childhood. Pediatr Pulmonol 2015; 50:155-63. [PMID: 24616290 DOI: 10.1002/ppul.23025] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 02/02/2014] [Accepted: 02/09/2014] [Indexed: 11/06/2022]
Abstract
BACKGROUND It is uncertain whether folate is risk or preventive factor for allergic and respiratory diseases. OBJECTIVE To determine the relationship between maternal or offspring folate status and subsequent development of allergic and respiratory diseases in early childhood. METHODS In total, 917 mother-child pairs from a prospective birth cohort in South Korea were studied. Data regarding the children's allergic and respiratory outcomes were obtained from standardized questionnaires completed by the mothers at postnatal months 6, 12, and 24. Serum folate levels were measured in the mothers at mid- and late-pregnancy, and in their children at 24 months of age. Atopic biomarkers were measured in the cord blood (CB) and at 24 months after birth. Biomarkers and clinical outcomes were analyzed and compared between the mother-child pairs divided into two groups according to median serum folate status at mid- and late-pregnancy. RESULTS Serum folate levels during mid-pregnancy were inversely associated with CB eosinophil count (adjusted odds ratio [OR] 0.72, 95% confidence interval [CI], 0.54-0.96) and positively associated with CB interleukin-10 levels (1.47, 1.11-1.94). Maternal folate level above the median value (≥9.5 ng/ml) during mid-pregnancy was associated with a decreased risk for the child of lower respiratory tract infections (LRTIs) at 6 months of age (adjusted OR 0.50, 95% CI 0.28-0.91) and atopic dermatitis (AD) at 24 months (adjusted OR 0.52, 95% CI 0.31-0.88), but not with LRTIs and AD at other ages. CONCLUSIONS A relatively high maternal serum folate level in mid-pregnancy was associated with a decreased risk of LRTIs and AD in early childhood.
Collapse
Affiliation(s)
- Ja Hyeong Kim
- Department of Pediatrics, Ulsan University Hospital, University of Ulsan Collage of Medicine, Ulsan, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
|
46
|
The Generation R Study: Biobank update 2015. Eur J Epidemiol 2014; 29:911-27. [PMID: 25527369 DOI: 10.1007/s10654-014-9980-6] [Citation(s) in RCA: 182] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Accepted: 12/06/2014] [Indexed: 12/14/2022]
Abstract
The Generation R Study is a population-based prospective cohort study from fetal life until adulthood. The study is designed to identify early environmental and genetic causes and causal pathways leading to normal and abnormal growth, development and health from fetal life, childhood and young adulthood. In total, 9,778 mothers were enrolled in the study. Data collection in children and their parents include questionnaires, interviews, detailed physical and ultrasound examinations, behavioural observations, Magnetic Resonance Imaging and biological samples. Efforts have been conducted for collecting biological samples including blood, hair, faeces, nasal swabs, saliva and urine samples and generating genomics data on DNA, RNA and microbiome. In this paper, we give an update of the collection, processing and storage of these biological samples and available measures. Together with detailed phenotype measurements, these biological samples provide a unique resource for epidemiological studies focused on environmental exposures, genetic and genomic determinants and their interactions in relation to growth, health and development from fetal life onwards.
Collapse
|
47
|
High dose of maternal folic acid supplementation is associated to infant asthma. Food Chem Toxicol 2014; 75:88-93. [PMID: 25449200 DOI: 10.1016/j.fct.2014.11.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 08/21/2014] [Accepted: 11/08/2014] [Indexed: 01/19/2023]
Abstract
Maternal folic acid supplementation had a positive effect on preventing neural tube defects (NTDs), but its effects in infant asthma remained unclear. A hospital-based case-control study was conducted with outpatients between March 2010 and March 2011 including 150 onset infant asthma cases and 212 controls, together with a meta-analysis involving 14,438 participants, was performed. The association between maternal folic acid supplementation and the risk of infant asthma was not significant either in the meta-analysis (OR = 1.06, 95% CI =0.99-1.14) or in the case-control study (OR = 0.72, 95% CI =0.37-1.39). However, quantitative analysis of the supplementation dose demonstrated that the risk of infant asthma significantly increased for the infants whose mother were with high-dose supplementation (>72,000 µg•d; OR = 3.16, 95% CI =1.15-8.71) after adjusting for confounding factors in the case-control study. Meanwhile, the risk of infant asthma significantly decreased for the infants whose mother were with low-dose supplementation (<36,000 µg•d; OR = 0.36, 95% CI =0.17-0.77). A high dose of folic acid supplementation for mother during pregnancy was associated with an increased risk of infant asthma, whereas supplementation with a relatively low-dose was associated with a decreased risk of infant asthma. These findings should be further investigated in a large population.
Collapse
|
48
|
Maslova E, Hansen S. Prenatal Dietary Determinants of Asthma and Related Allergic Disorders in Childhood. Curr Nutr Rep 2014. [DOI: 10.1007/s13668-014-0089-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
49
|
Barua S, Kuizon S, Junaid MA. Folic acid supplementation in pregnancy and implications in health and disease. J Biomed Sci 2014; 21:77. [PMID: 25135350 PMCID: PMC4237823 DOI: 10.1186/s12929-014-0077-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 08/11/2014] [Indexed: 12/31/2022] Open
Abstract
Maternal exposure to dietary factors during pregnancy can influence embryonic development and may modulate the phenotype of offspring through epigenetic programming. Folate is critical for nucleotide synthesis, and preconceptional intake of dietary folic acid (FA) is credited with reduced incidences of neural tube defects in infants. While fortification of grains with FA resulted in a positive public-health outcome, concern has been raised for the need for further investigation of unintended consequences and potential health hazards arising from excessive FA intakes, especially following reports that FA may exert epigenetic effects. The objective of this article is to discuss the role of FA in human health and to review the benefits, concerns and epigenetic effects of maternal FA on the basis of recent findings that are important to design future studies.
Collapse
Affiliation(s)
- Subit Barua
- Department of Developmental Biochemistry, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island 10314, NY, USA
| | - Salomon Kuizon
- Department of Developmental Biochemistry, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island 10314, NY, USA
| | - Mohammed A Junaid
- Department of Developmental Biochemistry, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island 10314, NY, USA
| |
Collapse
|
50
|
Amarasekera M, Martino D, Ashley S, Harb H, Kesper D, Strickland D, Saffery R, Prescott SL. Genome-wide DNA methylation profiling identifies a folate-sensitive region of differential methylation upstream of ZFP57-imprinting regulator in humans. FASEB J 2014; 28:4068-76. [PMID: 24891518 DOI: 10.1096/fj.13-249029] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 05/27/2014] [Indexed: 12/21/2022]
Abstract
Folate intake during pregnancy may affect the regulation of DNA methylation during fetal development. The genomic regions in the offspring that may be sensitive to folate exposure during in utero development have not been characterized. Using genome-scale profiling, we investigated DNA methylation in 2 immune cell types (CD4(+) and antigen-presenting cells) isolated from neonatal cord blood, selected on the basis of in utero folate exposure. High-folate (HF; n=11) and low-folate (LF; n=12) groups were selected from opposite extremes of maternal serum folate levels measured in the last trimester of pregnancy. A comparison of these groups revealed differential methylation at 7 regions across the genome. By far, the biggest effect observed was hypomethylation of a 923 bp region 3 kb upstream of the ZFP57 transcript, a regulator of DNA methylation during development, observed in both cell types. Levels of H3/H4 acetylation at ZFP57 promoter and ZFP57 mRNA expression were higher in CD4(+) cells in the HF group relative to the LF group. Hypomethylation at this region was replicated in an independent sample set. These data suggest that exposure to folate has effects on the regulation of DNA methylation during fetal development, and this may be important for health and disease.
Collapse
Affiliation(s)
- Manori Amarasekera
- School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia, Australia
| | - David Martino
- Cancer and Disease Epigenetics Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia; Centre for Food and Allergy Research, Melbourne, Victoria, Australia
| | - Sarah Ashley
- Cancer and Disease Epigenetics Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Institute for Medical Research, Monash University, Melbourne, Victoria, Australia
| | - Hani Harb
- Institute of Laboratory Medicine, Pathobiochemistry, and Molecular Diagnostics, Philipps University, Marburg, Germany; and
| | - Dörthe Kesper
- Institute of Laboratory Medicine, Pathobiochemistry, and Molecular Diagnostics, Philipps University, Marburg, Germany; and
| | - Deborah Strickland
- Telethon Institute for Child Health Research, Perth, Western Australia, Australia
| | - Richard Saffery
- Cancer and Disease Epigenetics Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Susan L Prescott
- School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia, Australia;
| |
Collapse
|