1
|
Wang G, Song S, Shen WB, Reece EA, Yang P. MicroRNA-322 overexpression reduces neural tube defects in diabetic pregnancies. Am J Obstet Gynecol 2024; 230:254.e1-254.e13. [PMID: 37531989 PMCID: PMC10828117 DOI: 10.1016/j.ajog.2023.07.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/14/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND Hyperglycemia from pregestational diabetes mellitus induces neural tube defects in the developing fetus. Folate supplementation is the only effective way to prevent neural tube defects; however, some cases of neural tube defects are resistant to folate. Excess folate has been linked to higher maternal cancer risk and infant allergy. Therefore, additional interventions are needed. Understanding the mechanisms underlying maternal diabetes mellitus-induced neural tube defects can identify potential targets for preventing such defects. Despite not yet being in clinical use, growing evidence suggests that microRNAs are important intermediates in embryonic development and can serve as both biomarkers and drug targets for disease intervention. Our previous studies showed that maternal diabetes mellitus in vivo activates the inositol-requiring transmembrane kinase/endoribonuclease 1α (IRE1α) in the developing embryo and that a high glucose condition in vitro reduces microRNA-322 (miR-322) levels. IRE1α is an RNA endonuclease; however, it is unknown whether IRE1α targets and degrades miR-322 specifically or whether miR-322 degradation leads to neural tube defects via apoptosis. We hypothesize that IRE1α can inhibit miR-322 in maternal diabetes mellitus-induced neural tube defects and that restoring miR-322 expression in developing neuroepithelium ameliorates neural tube defects. OBJECTIVE This study aimed to identify potential targets for preventing maternal diabetes mellitus-induced neural tube defects and to investigate the roles and relationship of a microRNA and an RNA endonuclease in mouse embryos exposed to maternal diabetes mellitus. STUDY DESIGN To determine whether miR-322 reduction is necessary for neural tube defect formation in pregnancies complicated by diabetes mellitus, male mice carrying a transgene expressing miR-322 were mated with nondiabetic or diabetic wide-type female mice to generate embryos with or without miR-322 overexpression. At embryonic day 8.5 when the neural tube is not yet closed, embryos were harvested for the assessment of 3 miR-322 transcripts (primary, precursor, and mature miR-322), tumor necrosis factor receptor-associated factor 3 (TRAF3), and neuroepithelium cell survival. Neural tube defect incidences were determined in embryonic day 10.5 embryos when the neural tube should be closed if there is no neural tube defect formation. To identify which miR-322 transcript is affected by maternal diabetes mellitus and high glucose conditions, 3 miR-322 transcripts were assessed in embryos from dams with or without diabetes mellitus and in C17.2 mouse neural stem cells treated with different concentrations of glucose and at different time points. To determine whether the endonuclease IRE1α targets miR-322, small interfering RNA knockdown of IRE1α or overexpression of inositol-requiring transmembrane kinase/endoribonuclease 1α by DNA plasmid transfection was used to determine the effect of IRE1α deficiency or overexpression on miR-322 expression. RNA immunoprecipitation was performed to reveal the direct targets of inositol-requiring transmembrane kinase/endoribonuclease 1α. RESULTS Maternal diabetes mellitus suppressed miR-322 expression in the developing neuroepithelium. Restoring miR-322 expression in the neuroepithelium blocked maternal diabetes mellitus-induced caspase-3 and caspase-8 cleavage and cell apoptosis, leading to a neural tube defect reduction. Reversal of maternal diabetes mellitus-inhibited miR-322 via transgenic overexpression prevented TRAF3 up-regulation in embryos exposed to maternal diabetes mellitus. Activated IRE1α acted as an endonuclease and degraded precursor miR-322, resulting in mature miR-322 reduction. CONCLUSION This study supports the crucial role of the IRE1α-microRNA-TRAF3 circuit in the induction of neuroepithelial cell apoptosis and neural tube defect formation in pregnancies complicated by diabetes mellitus and identifies IRE1α and miR-322 as potential targets for preventing maternal diabetes mellitus-induced neural tube defects.
Collapse
Affiliation(s)
- Guanglei Wang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - Shicong Song
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - Wei-Bin Shen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - E Albert Reece
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD
| | - Peixin Yang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD.
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Perera N, Rudland VL, Simmons D, Price SAL. Folate Supplementation in Women with Pre-Existing Diabetes. Nutrients 2023; 15:nu15081879. [PMID: 37111098 PMCID: PMC10145371 DOI: 10.3390/nu15081879] [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: 03/20/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Folate supplementation in the periconceptual period is the standard of care for the prevention of neural tube defects. To support dietary folate intake, some countries have introduced mandatory folic acid fortification of food products. Robust evidence supports the additional use of a low-dose folic acid supplement (0.4 mg/day) in all women from 2-3 months preconception until the end of the 12th week of gestation. For women with pre-existing diabetes, high-dose folic acid supplementation (5 mg/day) is recommended in some, but not all international guidelines. The recommendation is made based on consensus opinion and reflects the increased risk of neural tube defects in pregnant women with pre-existing diabetes. However, there is limited evidence to clarify the high-risk groups that benefit from high-dose folic acid versus those that do not. There are also some data to suggest that high-dose folic acid may be harmful to mothers and offspring, although this issue remains controversial. This narrative review explores the evidence that supports the recommendation for women with pre-existing diabetes to take high-dose folic acid in the periconceptual period. It explores the potential benefits of high-dose supplemental folate beyond the prevention of neural tube defects, and also the potential adverse impacts of high-dose folate use. These topics are considered with a specific focus on the issues that are pertinent to women with pre-existing diabetes. Based on the available evidence, a pragmatic approach to the use of folic acid supplements in women with pre-existing diabetes during the periconception period is suggested. The need for comprehensive preconception care that optimises glycaemic control and addresses other modifiable risk factors before pregnancy is emphasized.
Collapse
Affiliation(s)
- Nayomi Perera
- Department of Obstetric Medicine, Royal Women's Hospital, Flemington Rd, North Melbourne, VIC 3051, Australia
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Grattan St, Parkville, VIC 3010, Australia
| | - Victoria L Rudland
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - David Simmons
- Macarthur Clinical School, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Sarah A L Price
- Department of Obstetric Medicine, Royal Women's Hospital, Flemington Rd, North Melbourne, VIC 3051, Australia
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Grattan St, Parkville, VIC 3010, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Grattan St, Parkville, VIC 3010, Australia
| |
Collapse
|
5
|
Karaçil Ermumcu MŞ, Acar Tek N. Effects of High-dose Folic Acid Supplementation on Maternal/Child Health Outcomes: Gestational Diabetes Mellitus in Pregnancy and Insulin Resistance in Offspring. Can J Diabetes 2023; 47:133-142. [PMID: 36411183 DOI: 10.1016/j.jcjd.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVES The purpose of this study was to evaluate the effects of maternal high folic acid (FA) supplementation during pregnancy on glucose intolerance in dams and insulin resistance in offspring. METHODS Wistar female rats (n=18) were mated and randomly divided into 3 groups: a control group and 2 experimental groups. Three different feeding protocols were administered during pregnancy: control group, 2 mg/kg FA (recommended level FA supplementation); experimental 1 group, 5 mg/kg FA (tolerable upper intake level of FA supplementation [ULFolS]); and experimental 2 group, 40 mg/kg FA (high FA supplementation [HfolS]). All dams were fed the same FA content diet (2 mg/kg FA) during the lactation period. An oral glucose tolerance test was performed on day 16 of pregnancy. After the lactation period, body weight and food intake of 36 pups were monitored. Dams were euthanized at the end of the lactation period and half of the pups were euthanized at the end of week 7 and the others at the end of week 12. Serum FA, homocysteine, vitamin B12, insulin, glucose, interleukin-6, tumor necrosis factor-alpha, glycated hemoglobin (A1C), and adiponectin levels of mothers and pups were evaluated. The homeostatic model of insulin resistance (HOMA-IR) was used to determine insulin resistance in dams and offspring. RESULTS According to glucose tolerance test results of dams, blood glucose values at minutes 0, 60, 90, and 120 for the HFolS group were significantly higher compared with the control group (p<0.05). The A1C level in HFolS dams was significantly higher than in the control group (p<0.05). The mean birthweight of the pups in the HFolS group was significantly higher than that of control pups (p<0.05). HOMA-IR values for control and HFolS offspring were similar at weeks 7 and 12 and higher than in ULFolS offspring (p>0.05). CONCLUSIONS It was determined that high doses of FA exposure during pregnancy might be effective in the development of glucose intolerance in dams and insulin resistance in offspring in this study.
Collapse
Affiliation(s)
| | - Nilüfer Acar Tek
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Gazi University, Ankara, Turkey
| |
Collapse
|
6
|
Stumpf K, Mirpuri J. Maternal Macro- and Micronutrient Intake During Pregnancy: Does It Affect Allergic Predisposition in Offspring? Immunol Allergy Clin North Am 2023; 43:27-42. [PMID: 36411006 DOI: 10.1016/j.iac.2022.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
This review article explores the available literature on the association of maternal nutrient intake with development of allergies in offspring. It examines the mechanisms for maternal diet-mediated effects on offspring immunity and dissects recent human and animal studies that evaluate the role of both maternal macro- and micronutrient intake on offspring susceptibility to asthma, eczema, food allergy, and atopy.
Collapse
Affiliation(s)
- Katherine Stumpf
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Boulevard- Suite F3.302, Dallas, TX 75390-9063, USA.
| | - Julie Mirpuri
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Boulevard- Suite F3.302, Dallas, TX 75390-9063, USA.
| |
Collapse
|
7
|
Kong X, Yan Q, Niu Y, Liu L. The metabolic adaptation of the adult offspring after maternal high-dosed folic acid supplementation based on the proteomics and metabolomics in rats. Biomed Chromatogr 2022; 36:e5490. [PMID: 36005806 DOI: 10.1002/bmc.5490] [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: 06/28/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/06/2022]
Abstract
The onset of complex diseases at a later stage of life has been linked with maternal folic acid (FA) ingestion. However, little is known regarding the underlying molecular fingerprints of the offspring. We integrated proteomics-metabolomics profiles and analyzed the influence of maternal FA supplementation on the metabolism of adult offspring rats. Twenty pregnant female rats were randomly assigned to a FA supplementation (FolS group, 10 mg/kg FA) or control group (2 mg/kg FA respectively). Such an omics approach revealed that the dopaminergic synapse pathway, tricarboxylic acid cycle and neural development-related metabolites such as glutamic acid and γ-aminobutyric acid were significantly up-regulated in the FolS group, whereas pyruvic acid, oxalic acid and adipic acid were reduced. Maternal FA supplementation can cause alterations of metabolites and protein in the offspring rats.
Collapse
Affiliation(s)
- Xiangju Kong
- Department of Gynaecology, First Affiliated Hospital of Harbin Medical University, Harbin, P. R. China
| | - Qingna Yan
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, P. R. China
| | - Yucun Niu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, P. R. China
| | - Liyan Liu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, P. R. China
| |
Collapse
|
8
|
Dwyer ER, Filion KB, MacFarlane AJ, Platt RW, Mehrabadi A. Who should consume high-dose folic acid supplements before and during early pregnancy for the prevention of neural tube defects? BMJ 2022; 377:e067728. [PMID: 35672044 DOI: 10.1136/bmj-2021-067728] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Erin Rose Dwyer
- Departments of Obstetrics and Gynecology and Urology, University of Washington, Seattle, Washington, USA
| | - Kristian B Filion
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | | | - Robert W Platt
- Nutrition Research Division, Health Canada, Ottawa, Ontario, Canada
| | - Azar Mehrabadi
- Departments of Obstetrics & Gynaecology and Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
Molloy AM, Mills JL. Folic Acid and Infant Allergy: Avoiding Rash Judgments. J Nutr 2021; 151:1367-1368. [PMID: 33877314 PMCID: PMC8169806 DOI: 10.1093/jn/nxab084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 11/14/2022] Open
Affiliation(s)
| | - James L Mills
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute for Child Health and Human Development, Bethesda, MD, USA
| |
Collapse
|
13
|
Liu L, Liu Z, Li Y, Sun C. Integration of metabolomics and proteomics to highlight altered neural development related pathways in the adult offspring after maternal folic acid supplement. Clin Nutr 2021; 40:476-487. [PMID: 32571678 DOI: 10.1016/j.clnu.2020.05.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/14/2020] [Accepted: 05/23/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND & AIMS Maternal folic acid (FA) supplement (FolS) programs the early development of an offspring. The onset of complex diseases at a later stage of life has been evidently linked with maternal FA ingestion. However, little is known regarding the underlying molecule fingerprints of the offspring. Here, we analyze the influence of maternal FolS on the metabolism of the adult offspring rats using the integrated metabolomics-proteomics. METHODS Twenty pregnant female rats were randomly assigned to a FA supplement (FolS group) or control group which were fed AIN93G diet with 2 or 5 mg/kg FA, respectively. The blood samples from the offspring at 0, 3 and 7 weeks after birth were collected. The brain samples were obtained from the offspring at 7 weeks after birth. Serum and brain metabolite profiles were performed by UPLC-MS/MS and the brain proteomics analysis was obtained using iTRAQ-based quantitative proteomics. RESULTS The metabolic change of the offspring for the maternal FA supplement is characterized by the phospholipids, fatty acid and amino acids, which are involved in linoleic acid, docosahexaenoic acid, glycerophosphocholine, lysophosphatidylcholine, tryptophan, glycine, arachidonic acid, γ-aminobutyric acid, and so on. Using iTRAQ-based quantitative proteomics analysis, 51 differential proteins in the brain are identified, which provides valuable insight into the underlying mechanisms of the offspring after the maternal FolS. These results demonstrate neural development related metabolites and proteins, such as docosahexaenoic acid, glycine, tryptophan, γ-aminobutyric acid, dopaminergic synapse related proteins including G protein, PPP1R1B and CAMK2G, are significantly altered, which suggests that the active neural conduction occurs in the offspring after maternal FA supplement. The behavioral testing demonstrates that the high level of memory is observed in rats with FA supplement. CONCLUSIONS We conceive that the alterations of metabolites and protein in the offspring are associated with the maternal FA supplement and these alterations are involved in the neural development, although such animal data are limited in their ability to mimic metabolic outcomes in humans.
Collapse
Affiliation(s)
- Liyan Liu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, PR China
| | - Zhipeng Liu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, PR China
| | - Ying Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, PR China.
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, PR China.
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
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
|
17
|
Venter C, Agostoni C, Arshad SH, Ben-Abdallah M, Du Toit G, Fleischer DM, Greenhawt M, Glueck DH, Groetch M, Lunjani N, Maslin K, Maiorella A, Meyer R, Antonella M, Netting MJ, Nwaru BI, Palmer DJ, Palumbo MP, Roberts G, Roduit C, Smith P, Untersmayr E, Vanderlinden LA, O’Mahony L. Dietary factors during pregnancy and atopic outcomes in childhood: A systematic review from the European Academy of Allergy and Clinical Immunology. Pediatr Allergy Immunol 2020; 31:889-912. [PMID: 32524677 PMCID: PMC9588404 DOI: 10.1111/pai.13303] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/06/2020] [Accepted: 05/20/2020] [Indexed: 12/17/2022]
Abstract
RATIONALE Allergic diseases are an increasing public health concern, and early life environment is critical to immune development. Maternal diet during pregnancy has been linked to offspring allergy risk. In turn, maternal diet is a potentially modifiable factor, which could be targeted as an allergy prevention strategy. In this systematic review, we focused on non-allergen-specific modifying factors of the maternal diet in pregnancy on allergy outcomes in their offspring. METHODS We undertook a systematic review of studies investigating the association between maternal diet during pregnancy and allergic outcomes (asthma/wheeze, hay fever/allergic rhinitis/seasonal allergies, eczema/atopic dermatitis (AD), food allergies, and allergic sensitization) in offspring. Studies evaluating the effect of food allergen intake were excluded. We searched three bibliographic databases (MEDLINE, EMBASE, and Web of Science) through February 26, 2019. Evidence was critically appraised using modified versions of the Cochrane Collaboration Risk of Bias tool for intervention trials and the National Institute for Clinical Excellence methodological checklist for cohort and case-control studies and meta-analysis performed from RCTs. RESULTS We identified 95 papers: 17 RCTs and 78 observational (case-control, cross-sectional, and cohort) studies. Observational studies varied in design and dietary intakes and often had contradictory findings. Based on our meta-analysis, RCTs showed that vitamin D supplementation (OR: 0.72; 95% CI: 0.56-0.92) is associated with a reduced risk of wheeze/asthma. A positive trend for omega-3 fatty acids was observed for asthma/wheeze, but this did not reach statistical significance (OR: 0.70; 95% CI: 0.45-1.08). Omega-3 supplementation was also associated with a non-significant decreased risk of allergic rhinitis (OR: 0.76; 95% CI: 0.56-1.04). Neither vitamin D nor omega-3 fatty acids were associated with an altered risk of AD or food allergy. CONCLUSIONS Prenatal supplementation with vitamin D may have beneficial effects for prevention of asthma. Additional nutritional factors seem to be required for modulating the risk of skin and gastrointestinal outcomes. We found no consistent evidence regarding other dietary factors, perhaps due to differences in study design and host features that were not considered. While confirmatory studies are required, there is also a need for performing RCTs beyond single nutrients/foods.
Collapse
Affiliation(s)
- Carina Venter
- Section of Allergy and Immunology, University of Colorado School of Medicine, Denver, CO, USA
- Children’s Hospital Colorado, Aurora, CO, USA
| | - Carlo Agostoni
- Pediatria Media Intensità di Cura Fondazione IRCCS Ca’ Granda - Ospedale Maggiore Policlinic, Milan, Italy
| | - S. Hasan Arshad
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- The David Hide Asthma and Allergy Centre, Isle of Wight, UK
| | | | - George Du Toit
- Department of Paediatric Allergy, Division of Asthma, Allergy and Lung Biology, King’s College London, London, UK
- Evelina London, Guy’s & St Thomas’ Hospital, London, UK
| | - David M. Fleischer
- Section of Allergy and Immunology, University of Colorado School of Medicine, Denver, CO, USA
- Children’s Hospital Colorado, Aurora, CO, USA
| | - Matthew Greenhawt
- Section of Allergy and Immunology, University of Colorado School of Medicine, Denver, CO, USA
- Children’s Hospital Colorado, Aurora, CO, USA
| | - Deborah H. Glueck
- Department of Pediatrics, University of Colorado School of Medicine, Denver, CO, USA
| | - Marion Groetch
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nonhlanhla Lunjani
- University of Zurich, Davos, Switzerland
- University of Cape Town, Cape Town, South Africa
| | | | | | | | - Muraro Antonella
- Centro di Specializzazione Regionale per lo Studio e la Cura delle Allergie e delle Intolleranze Alimentari presso l’Azienda Ospedaliera, Università di Padova, Padova, Italy
| | - Merryn J. Netting
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Discipline of Pediatrics, University of Adelaide, Adelaide, SA, Australia
| | | | - Debra J. Palmer
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Micheala P. Palumbo
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Colorado School of Public Health, Colorado, USA
| | - Graham Roberts
- The David Hide Asthma and Allergy Centre, Isle of Wight, UK
- Department of Paediatric Allergy, Division of Asthma, Allergy and Lung Biology, King’s College London, London, UK
- NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Faculty of Medicine, Human Development in Health Academic Units, University of Southampton, Southampton, UK
| | - Caroline Roduit
- University Children’s Hospital Zurich, Switzerland
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Pete Smith
- School of Medicine, Griffith University, Southport, Australia
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Lauren A. Vanderlinden
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - Liam O’Mahony
- Departments of Medicine and Microbiology, APC Microbiome Ireland, National University of Ireland, Cork, Ireland
| |
Collapse
|
18
|
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
|
19
|
Egorova O, Myte R, Schneede J, Hägglöf B, Bölte S, Domellöf E, Ivars A'roch B, Elgh F, Ueland PM, Silfverdal SA. Maternal blood folate status during early pregnancy and occurrence of autism spectrum disorder in offspring: a study of 62 serum biomarkers. Mol Autism 2020; 11:7. [PMID: 32131900 PMCID: PMC6964211 DOI: 10.1186/s13229-020-0315-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 01/02/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) evolves from an interplay between genetic and environmental factors during prenatal development. Since identifying maternal biomarkers associated with ASD risk in offspring during early pregnancy might result in new strategies for intervention, we investigated maternal metabolic biomarkers in relation to occurrence of ASD in offspring using both univariate logistic regression and multivariate network analysis. METHODS Serum samples from 100 women with an offspring diagnosed with ASD and 100 matched control women with typically developing offspring were collected at week 14 of pregnancy. Concentrations of 62 metabolic biomarkers were determined, including amino acids, vitamins (A, B, D, E, and K), and biomarkers related to folate (vitamin B9) metabolism, lifestyle factors, as well as C-reactive protein (CRP), the kynurenine-tryptophan ratio (KTR), and neopterin as markers of inflammation and immune activation. RESULTS We found weak evidence for a positive association between higher maternal serum concentrations of folate and increased occurrence of ASD (OR per 1 SD increase: 1.70, 95% CI 1.22-2.37, FDR adjusted P = 0.07). Multivariate network analysis confirmed expected internal biochemical relations between the biomarkers. Neither inflammation markers nor vitamin D3 levels, all hypothesized to be involved in ASD etiology, displayed associations with ASD occurrence in the offspring. CONCLUSIONS Our findings suggest that high maternal serum folate status during early pregnancy may be associated with the occurrence of ASD in offspring. No inference about physiological mechanisms behind this observation can be made at the present time because blood folate levels may have complex relations with nutritional intake, the cellular folate status and status of other B-vitamins. Therefore, further investigations, which may clarify the potential role and mechanisms of maternal blood folate status in ASD risk and the interplay with other potential risk factors, in larger materials are warranted.
Collapse
Affiliation(s)
- Olga Egorova
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden.
| | - Robin Myte
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Jörn Schneede
- Department of Clinical Pharmacology, Pharmacology and Clinical Neurosciences, Umeå University, Umeå, Sweden
| | - Bruno Hägglöf
- Department of Child and Adolescent Psychiatry, Umea University, Umeå, Sweden
| | - Sven Bölte
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Stockholm, Sweden.,Department of Women's and Children's Health, Karolinska Institutet & Child and Adolescent Psychiatry, Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden.,Curtin Autism Research Group, School of Occupational Therapy, Social Work and Speech Pathology, Curtin University, Perth, WA, Australia
| | - Erik Domellöf
- Department of Psychology, Umeå University, Umeå, Sweden
| | - Barbro Ivars A'roch
- Department of Child and Adolescent Psychiatry, Umea University, Umeå, Sweden
| | - Fredrik Elgh
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Per Magne Ueland
- Bevital AS, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | | |
Collapse
|
20
|
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
|
21
|
McGowan EC, Hong X, Selhub J, Paul L, Wood RA, Matsui EC, Keet CA, Wang X. Association Between Folate Metabolites and the Development of Food Allergy in Children. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2020; 8:132-140.e5. [PMID: 31252026 PMCID: PMC6930362 DOI: 10.1016/j.jaip.2019.06.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 05/10/2019] [Accepted: 06/07/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Studies on the association between folate/folic acid exposure and the development of allergic disease have yielded inconsistent results, which may be due, in part, to lack of data distinguishing folate from folic acid exposure. OBJECTIVE To examine the association between total folate, 5-methyltetrahydrofolate (5-MTHF), and unmetabolized folic acid (UMFA) concentrations at birth and in early childhood and the development of food sensitization (FS) and food allergy (FA). METHODS A nested case control study was performed in the Boston Birth Cohort (BBC). Total folate, 5-MTHF, and UMFA were measured at birth and in early childhood. Based on food-specific IgE (sIgE) levels, diet, and clinical history, children were classified as FS (sIgE ≥0.35 kU/L), FA, or non-FS/FA (controls). Folate concentrations were divided into quartiles, and multiple logistic regression was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS Of a total of 1394 children, 507 children with FS and 78 with FA were identified. Although mean total folate concentrations at birth were lower among those who developed FA (30.2 vs 35.3 nmol/L; P = .02), mean concentrations of the synthetic folic acid derivative, UMFA, were higher (1.7 vs 1.3 nmol/L, P = .001). Higher quartiles of UMFA at birth were associated more strongly with FA (OR 8.50; 95% CI 1.7-42.8; test for trend P = .001). Neither early childhood concentrations of 5-MTHF nor UMFA were associated with the development of FS or FA. CONCLUSION Among children in the BBC, higher concentrations of UMFA at birth were associated with the development of FA, which may be due to increased exposure to synthetic folic acid in utero or underlying genetic differences in synthetic folic acid metabolism.
Collapse
Affiliation(s)
- Emily C McGowan
- Division of Allergy and Immunology, University of Virginia School of Medicine, Charlottesville, Va; Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md.
| | - Xiumei Hong
- Department of Population, Family, and Reproductive Health, Center on the Early Life Origins of Disease, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Md
| | - Jacob Selhub
- Tufts University, JM USDA HNRCA at Tufts University, Boston, Mass
| | - Ligi Paul
- Tufts University, JM USDA HNRCA at Tufts University, Boston, Mass
| | - Robert A Wood
- Division of Pediatric Allergy and Immunology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Elizabeth C Matsui
- Division of Pediatric Allergy and Immunology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Corinne A Keet
- Division of Pediatric Allergy and Immunology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Xiaobin Wang
- Department of Population, Family, and Reproductive Health, Center on the Early Life Origins of Disease, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Md
| |
Collapse
|
22
|
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
|
23
|
Maulik D, van Haandel L, Allsworth J, Chaisanguanthum KS, Yeast JD, Leeder JS. The effect of race and supplementation on maternal and umbilical cord plasma folates. J Matern Fetal Neonatal Med 2019; 34:3057-3065. [PMID: 31630592 DOI: 10.1080/14767058.2019.1677597] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE The purpose of this study is to test the hypothesis that race and supplementation affect the concentration and correlation of various folate species in maternal and umbilical cord blood. METHODS This is a single-center, prospective, cross-sectional cohort of cord blood samples obtained from 40 uncomplicated term pregnancies as a pilot study, following a protocol approved by the Institutional Review Board. High performance liquid chromatography mass spectrometry quantitated the following concentrations in extracted plasma samples: 5-methyltetrahydrofolate (5MTHF), 5,10-methenyl-tetrahydrofolate (5,10-MeTHF), tetrahydrofolate (THF), and unmetabolized folic acid. RESULTS Folate concentrations in the umbilical cord plasma were consistently higher than maternal samples for 5MTHF (p < .001), 5,10-MeTHF (p < .001), and THF (p < .001); cord blood folic acid levels, however, were lower than maternal samples (p < .03). While 5MTHF was the most prevalent folate, ratios comparing cord blood to maternal blood folates suggests a fourfold preponderance of THF in cord blood folate signature, a trend unchanged by supplementation. Prenatal supplementation increased the concentrations of 5MTHF, for both maternal (p < .01) and cord blood samples (p < .005). In comparison to the other two racial groups, African American 5MTHF concentration demonstrated a lower total folate concentration in both maternal samples and cord blood samples, in addition to a relatively blunted response to supplementation. A significantly positive correlation between maternal and cord blood 5MTHF concentration was noted in all three racial groups. Supplementation resulted in a positive correlation between maternal and cord blood 5MTHF concentrations (r = 0.85, p < .0001). CONCLUSIONS 5MTHF is the most prevalent folate in both cord and maternal plasma, and race and supplementation primarily affect variations in maternal and fetal 5MTHF concentrations and their correlation with each other. However, the greater concentration of THF in cord blood relative to maternal blood offers preliminary insight into the importance of how folate metabolism differs in the specific context of fetal development and physiology, with greater emphasis on DNA synthesis and stability. Furthermore, supplementation appeared to not have as great an impact on African American maternal or cord blood folates, suggesting a variable benefit of current repletion strategies to certain subsets of the population. Future studies that further elucidate these differences and their impact on birth outcomes may help inform supplementation protocols that are more personalized, with greater efficacy in promoting positive perinatal outcomes.
Collapse
Affiliation(s)
- Devika Maulik
- Children's Mercy Hospital/University of Missouri at Kansas City, Kansas City, MO, USA
| | - Leon van Haandel
- Children's Mercy Hospital/University of Missouri at Kansas City, Kansas City, MO, USA
| | - Jenifer Allsworth
- Department of Biomedical and Health Informatics, University of Missouri at Kansas City, Kansas City, MO, USA
| | - Kris S Chaisanguanthum
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
| | | | - J Steven Leeder
- Children's Mercy Hospital/University of Missouri at Kansas City, Kansas City, MO, USA
| |
Collapse
|
24
|
Pretorius RA, Bodinier M, Prescott SL, Palmer DJ. Maternal Fiber Dietary Intakes during Pregnancy and Infant Allergic Disease. Nutrients 2019; 11:nu11081767. [PMID: 31374861 PMCID: PMC6722741 DOI: 10.3390/nu11081767] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/24/2022] Open
Abstract
Maternal diet during pregnancy plays a likely role in infant immune development through both direct nutrient specific immunomodulatory effects and by modulating the composition and metabolic activity of the maternal gut microbiome. Dietary fibers, as major substrates for microbial fermentation, are of interest in this context. This is the first study to examine maternal intakes of different fiber sub-types and subsequent infant allergic disease. In an observational study of 639 mother–infant pairs (all infants had a family history of allergic disease) we examined maternal intakes of total fiber, soluble fiber, insoluble fiber, resistant starch, and prebiotic fiber, by a semi-quantitative food frequency questionnaire at 36–40 weeks’ gestation. Infants attended an allergy clinical assessment at 12 months of age, including skin prick testing to common allergens. Higher maternal dietary intakes of resistant starch were associated with reduced doctor diagnosed infant wheeze, adjusted odds ratio (aOR) 0.68 (95% CI 0.49, 0.95, p = 0.02). However, in contrast, higher maternal intakes of resistant starch were associated with higher risk of parent reported eczema aOR 1.27 (95% CI 1.09, 1.49, p < 0.01) and doctor diagnosed eczema aOR 1.19 (95% CI 1.01, 1.41, p = 0.04). In conclusion, maternal resistant starch consumption was differentially associated with infant phenotypes, with reduced risk of infant wheeze, but increased risk of eczema.
Collapse
Affiliation(s)
- Rachelle A Pretorius
- School of Medicine, University of Western Australia, 35 Stirling Highway, Crawley 6009, Western Australia, Australia
| | - Marie Bodinier
- INRA Pays de la Loire, UR 1268 Biopolymers Interactions Assemblies, rue de la géraudière, BP 71627, Cedex 3, 44316 Nantes, France
| | - Susan L Prescott
- School of Medicine, University of Western Australia, 35 Stirling Highway, Crawley 6009, Western Australia, Australia
- Telethon Kids Institute, University of Western Australia, 15 Hospital Ave, Nedlands 6009, Western Australia, Australia
| | - Debra J Palmer
- School of Medicine, University of Western Australia, 35 Stirling Highway, Crawley 6009, Western Australia, Australia.
- Telethon Kids Institute, University of Western Australia, 15 Hospital Ave, Nedlands 6009, Western Australia, Australia.
| |
Collapse
|
25
|
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
|
26
|
Pre-natal folic acid and iron supplementation and atopic dermatitis in the first 6 years of life. Arch Dermatol Res 2019; 311:361-367. [PMID: 30923900 DOI: 10.1007/s00403-019-01911-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 02/07/2019] [Accepted: 03/20/2019] [Indexed: 12/16/2022]
Abstract
Exposure in utero has been suggested to influence health later in life. The aim of this study was to investigate, if the use of prenatal food supplements was associated with atopic dermatitis in the offspring. Mothers who gave birth in the hospital G. B. Grassi were invited to participate in the study (n = 395). Information on socio-demographic characteristics, clinical data of the mothers and babies, vegetables and fruit intake, food avoidance, and food supplements use during pregnancy, depression status, and environmental exposure was obtained for all subjects in the hospital at the time of delivery. Data on breastfeeding practice, introduction of weaning foods, day care attendance, and atopic dermatitis were collected in the postnatal follow-ups. Logistic regression was applied to estimate odds ratio (OR) and 95% confidence intervals (CI). Children in which mothers used both iron and folic acid supplementation had a fourfold decreased risk of developing atopic dermatitis [OR = 0.22; 95% confidence interval (CI) 0.06-0.79, p = 0.02], after adjusting for possible confounding factors. Findings suggest an independent and protective effect of prenatal folic acid and iron supplementation for atopic dermatitis in children.
Collapse
|
27
|
Abstract
Periconceptional folic acid (FA) is known to have a protective effect in the prevention of neural tube defects (NTD), leading to global recommendations for FA supplementation before and in early pregnancy. Maternal folate throughout pregnancy may have other roles in offspring health, including neurodevelopment and cognitive performance in childhood. Folate is essential for C1 metabolism, a network of pathways involved in several biological processes including nucleotide synthesis, DNA repair and methylation reactions. The evidence reviewed here shows a conclusive role for offspring health of maternal folate nutrition in early pregnancy and probable benefits in later pregnancy. Folate-mediated epigenetic changes in genes related to brain development and function offer a plausible biological basis to link maternal folate with effects in offspring brain, albeit this research is in its infancy. Mandatory FA fortification of food has proven to be highly effective in decreasing NTD cases in populations where it has been implemented, but this policy is controversial owing to concerns related to potential adverse effects of over-exposure to FA. In the absence of population-wide fortification, and given the generally poor compliance with current FA recommendations, optimising folate status of mothers in very early pregnancy for protection against NTD remains challenging. Thus, current policy in the UK, Ireland and elsewhere in Europe for the prevention of NTD (based on periconceptional FA supplementation only), has proven to be largely ineffective. This review addresses the evidence and the controversies that surround this area, as well as identifying the challenges in translating policy into practice.
Collapse
|
28
|
Pham MN, Bunyavanich S. Prenatal Diet and the Development of Childhood Allergic Diseases: Food for Thought. Curr Allergy Asthma Rep 2018; 18:58. [PMID: 30229317 DOI: 10.1007/s11882-018-0811-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The development of allergic disease is shaped by genetics and the environment, including diet. Many studies suggest a role for maternal diet during pregnancy. In this article, we discuss potential mechanisms by which specific nutrients, particular foods, and dietary patterns may influence allergic disease development and review studies examining the relationship between prenatal diet and the risk of childhood allergy. RECENT FINDINGS The combination of in utero exposures and genetic predisposition may contribute to the development of allergic disease by altering immune and organ development. Inflammation predominates in the first and third trimesters whereas the second trimester is characterized by anti-inflammatory and Th2 immune responses. Maternal dietary exposures during pregnancy may interact with inherited genetic risk factors influence immune system development. There are varied results regarding the impact of maternal prenatal diet on the development of childhood allergies. Well-designed randomized controlled studies are needed to clarify this area.
Collapse
Affiliation(s)
- Michele N Pham
- Department of Pediatrics, Division of Allergy and Immunology, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1498, New York, NY, 10029, USA
| | - Supinda Bunyavanich
- Department of Pediatrics, Division of Allergy and Immunology, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1498, New York, NY, 10029, USA.
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
29
|
Alaskhar Alhamwe B, Khalaila R, Wolf J, von Bülow V, Harb H, Alhamdan F, Hii CS, Prescott SL, Ferrante A, Renz H, Garn H, Potaczek DP. Histone modifications and their role in epigenetics of atopy and allergic diseases. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2018; 14:39. [PMID: 29796022 PMCID: PMC5966915 DOI: 10.1186/s13223-018-0259-4] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 04/24/2018] [Indexed: 12/16/2022]
Abstract
This review covers basic aspects of histone modification and the role of posttranslational histone modifications in the development of allergic diseases, including the immune mechanisms underlying this development. Together with DNA methylation, histone modifications (including histone acetylation, methylation, phosphorylation, ubiquitination, etc.) represent the classical epigenetic mechanisms. However, much less attention has been given to histone modifications than to DNA methylation in the context of allergy. A systematic review of the literature was undertaken to provide an unbiased and comprehensive update on the involvement of histone modifications in allergy and the mechanisms underlying this development. In addition to covering the growing interest in the contribution of histone modifications in regulating the development of allergic diseases, this review summarizes some of the evidence supporting this contribution. There are at least two levels at which the role of histone modifications is manifested. One is the regulation of cells that contribute to the allergic inflammation (T cells and macrophages) and those that participate in airway remodeling [(myo-) fibroblasts]. The other is the direct association between histone modifications and allergic phenotypes. Inhibitors of histone-modifying enzymes may potentially be used as anti-allergic drugs. Furthermore, epigenetic patterns may provide novel tools in the diagnosis of allergic disorders.
Collapse
Affiliation(s)
- Bilal Alaskhar Alhamwe
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
- inVIVO Planetary Health, Group of the Worldwide Universities Network (WUN), New York, NJ USA
| | - Razi Khalaila
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
| | - Johanna Wolf
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
| | - Verena von Bülow
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
| | - Hani Harb
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
- inVIVO Planetary Health, Group of the Worldwide Universities Network (WUN), New York, NJ USA
- German Center for Lung Research (DZL), Gießen, Germany
- Present Address: Boston Children’s Hospital, Harvard Medical School, Boston, MA USA
| | - Fahd Alhamdan
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
| | - Charles S. Hii
- Department of Immunopathology, SA Pathology, Women and Children’s Hospital Campus, North Adelaide, SA Australia
- Robinson Research Institute, School of Medicine and School of Biological Science, University of Adelaide, Adelaide, SA Australia
| | - Susan L. Prescott
- inVIVO Planetary Health, Group of the Worldwide Universities Network (WUN), New York, NJ USA
- School of Paediatrics and Child Health, University of Western Australia, Perth, WA Australia
| | - Antonio Ferrante
- inVIVO Planetary Health, Group of the Worldwide Universities Network (WUN), New York, NJ USA
- Department of Immunopathology, SA Pathology, Women and Children’s Hospital Campus, North Adelaide, SA Australia
- Robinson Research Institute, School of Medicine and School of Biological Science, University of Adelaide, Adelaide, SA Australia
| | - Harald Renz
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
- inVIVO Planetary Health, Group of the Worldwide Universities Network (WUN), New York, NJ USA
- German Center for Lung Research (DZL), Gießen, Germany
| | - Holger Garn
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
- German Center for Lung Research (DZL), Gießen, Germany
| | - Daniel P. Potaczek
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
- inVIVO Planetary Health, Group of the Worldwide Universities Network (WUN), New York, NJ USA
- German Center for Lung Research (DZL), Gießen, Germany
- John Paul II Hospital, Krakow, Poland
| |
Collapse
|
30
|
Trends in folic acid supplementation during pregnancy - the effect on allergy development in children. Postepy Dermatol Alergol 2018; 35:139-144. [PMID: 29760612 PMCID: PMC5949539 DOI: 10.5114/pdia.2017.68785] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 02/01/2017] [Indexed: 11/17/2022] Open
Abstract
Introduction The results of some previous studies suggested that maternal folate supplementation during pregnancy may contribute to allergy development in offspring. Aim This study was performed to examine the influence of maternal folic acid intake prior to and during pregnancy on the development of various types of allergy in children taking into account the timing and dosage of supplemented folate. Material and methods The retrospective study was performed between 2010 and 2014 in 307 child-mother pairs (203 allergic children and 104 children without allergy symptoms, aged 2-72 months). Allergy diagnosis was based on medical history, physical examination, positive results of allergic tests: specific IgE and/or skin prick tests and double-blind, placebo-controlled food challenge. The data concerning maternal folate supplementation prior to and during pregnancy were obtained based on a questionnaire. Results Mothers of allergic children used to take folic acid more frequently in the preconception period (42.9%), in the 1st (94.1%) and the 2nd/3rd (81.3%) trimester of pregnancy than mothers of the healthy ones (30.8%, 82.7% and 55.8%, respectively) (p < 0.05). Maternal intake of folate in a dosage higher than recommended (> 0.4 mg/day) was more often observed in the group of allergic subjects, especially in children with combined sensitization to food and inhalant allergens, than in the control group (p < 0.05). Conclusions Our results suggest an impact of maternal folic acid supplementation prior to and during pregnancy on allergy development in children. Further observations are required to establish the role of folate in fetal epigenetic modifications.
Collapse
|
31
|
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
|
32
|
McGee M, Bainbridge S, Fontaine-Bisson B. A crucial role for maternal dietary methyl donor intake in epigenetic programming and fetal growth outcomes. Nutr Rev 2018. [DOI: 10.1093/nutrit/nuy006] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Meghan McGee
- Department of Nutritional Sciences, University of Toronto, and Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Shannon Bainbridge
- Interdisciplinary School of Health Sciences and the Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Bénédicte Fontaine-Bisson
- School of Nutrition Sciences, University of Ottawa, and the Institut du savoir Montfort, Ottawa, Ontario, Canada
| |
Collapse
|
33
|
Wooldridge AL, Bischof RJ, Liu H, Heinemann GK, Hunter DS, Giles LC, Simmons RA, Lien YC, Lu W, Rabinowitz JD, Kind KL, Owens JA, Clifton VL, Gatford KL. Late-gestation maternal dietary methyl donor and cofactor supplementation in sheep partially reverses protection against allergic sensitization by IUGR. Am J Physiol Regul Integr Comp Physiol 2017; 314:R22-R33. [PMID: 28978515 DOI: 10.1152/ajpregu.00549.2016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Perinatal exposures are associated with altered risks of childhood allergy. Human studies and our previous work suggest that restricted growth in utero (IUGR) is protective against allergic disease. The mechanisms are not clearly defined, but reduced fetal abundance and altered metabolism of methyl donors are hypothesized as possible underlying mechanisms. Therefore, we examined whether late-gestation maternal dietary methyl donor and cofactor supplementation of the placentally restricted (PR) sheep pregnancy would reverse allergic protection in progeny. Allergic outcomes were compared between progeny from control pregnancies (CON; n = 49), from PR pregnancies without intervention (PR; n = 28), and from PR pregnancies where the dam was fed a methyl donor plus cofactor supplement from day 120 of pregnancy until delivery (PR + Methyl; n = 25). Both PR and PR + Methyl progeny were smaller than CON; supplementation did not alter birth size. PR was protective against cutaneous hypersensitivity responses to ovalbumin (OVA; P < 0.01 in singletons). Cutaneous hypersensitivity responses to OVA in PR + Methyl progeny were intermediate to and not different from the responses of CON and PR sheep. Cutaneous hypersensitivity responses to house dust mites did not differ between treatments. In singleton progeny, upper dermal mast cell density was greater in PR + Methyl than in PR or CON (each P < 0.05). The differences in the cutaneous allergic response were not explained by treatment effects on circulating immune cells or antibodies. Our results suggest that mechanisms underlying in utero programming of allergic susceptibility by IUGR and methyl donor availability may differ and imply that late-gestation methyl donor supplementation may increase allergy risk.
Collapse
Affiliation(s)
- Amy L Wooldridge
- Robinson Research Institute, University of Adelaide , Adelaide, South Australia , Australia.,Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia
| | - Robert J Bischof
- The Ritchie Centre, Hudson Institute of Medical Research , Clayton, Victoria , Australia.,Department of Physiology, Monash University , Melbourne, Victoria , Australia
| | - Hong Liu
- Robinson Research Institute, University of Adelaide , Adelaide, South Australia , Australia.,Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia
| | - Gary K Heinemann
- Robinson Research Institute, University of Adelaide , Adelaide, South Australia , Australia.,Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia
| | - Damien S Hunter
- Robinson Research Institute, University of Adelaide , Adelaide, South Australia , Australia.,Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia
| | - Lynne C Giles
- Robinson Research Institute, University of Adelaide , Adelaide, South Australia , Australia.,School of Population Health, University of Adelaide , Adelaide, South Australia , Australia
| | - Rebecca A Simmons
- Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Yu-Chin Lien
- Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Wenyun Lu
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University , Princeton, New Jersey
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University , Princeton, New Jersey
| | - Karen L Kind
- Robinson Research Institute, University of Adelaide , Adelaide, South Australia , Australia.,School of Animal and Veterinary Sciences, University of Adelaide , Adelaide, South Australia , Australia
| | - Julie A Owens
- Robinson Research Institute, University of Adelaide , Adelaide, South Australia , Australia.,Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia
| | - Vicki L Clifton
- Robinson Research Institute, University of Adelaide , Adelaide, South Australia , Australia.,Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia.,Mater Research Institute-University of Queensland and Translational Research Institute, South Brisbane, Queensland, Australia
| | - Kathryn L Gatford
- Robinson Research Institute, University of Adelaide , Adelaide, South Australia , Australia.,Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia
| |
Collapse
|
34
|
Berti C, Agostoni C, Davanzo R, Hyppönen E, Isolauri E, Meltzer HM, Steegers-Theunissen RPM, Cetin I. Early-life nutritional exposures and lifelong health: immediate and long-lasting impacts of probiotics, vitamin D, and breastfeeding. Nutr Rev 2017; 75:83-97. [PMID: 28130504 DOI: 10.1093/nutrit/nuw056] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/16/2016] [Indexed: 12/21/2022] Open
Abstract
Pregnancy and infancy comprise the most critical stages for conditioning an individual's health, with a number of implications for subsequent risks of morbidity, mortality, and reproductive health. Nutrition may influence both the overall pregnancy outcome and the growth trajectory and immune system of the fetus and infant, with short- and long-term effects on the health of the offspring. Within this context, leading experts at Expo Milano 2015 in Milan, Italy, discussed up-to-date knowledge while providing suggestions and challenges before, during, and after pregnancy. This narrative review summarizes the key issues raised by the experts concerning the interplay between the nutritional environment from conception to early infancy and the offspring's immediate and lifelong health, with a particular focus on epigenetic mechanisms, probiotics, vitamin D, and breastfeeding. Taken together, the findings strengthen the awareness that nutritional exposures occurring from preconception to the postnatal period may be strong determinants of the offspring's health and may provide supportive evidence for current nutritional recommendations and guidelines for pregnant women and infants. Critical topics to be addressed in future research and translated into recommendations of public health relevance are also highlighted.
Collapse
Affiliation(s)
- Cristiana Berti
- Department of Biomedical and Clinical Sciences, School of Medicine and Center for Fetal Research Giorgio Pardi, University of Milan, Milan, Italy
| | - Carlo Agostoni
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Riccardo Davanzo
- Division of Neonatology and NICU, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Elina Hyppönen
- Centre for Population Health Research, Sansom Institute of Health Research and School of Health Sciences, University of South Australia, Adelaide, Australia.,South Australian Health and Medical Research Institute, Adelaide, Australia.,Population, Policy and Practice Programme, University College London Institute of Child Health, London, UK
| | - Erika Isolauri
- Department of Paediatrics, Turku University Hospital and University of Turku, Turku, Finland
| | - Helle M Meltzer
- Norwegian Institute of Public Health, Domain of Infection Control and Environmental Health, Oslo, Norway
| | - Régine P M Steegers-Theunissen
- Department of Obstetrics and Department of Gynaecology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Irene Cetin
- Department of Biomedical and Clinical Sciences, School of Medicine and Center for Fetal Research Giorgio Pardi, University of Milan, Milan, Italy
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
Castaño E, Caviedes L, Hirsch S, Llanos M, Iñiguez G, Ronco AM. Folate Transporters in Placentas from Preterm Newborns and Their Relation to Cord Blood Folate and Vitamin B12 Levels. PLoS One 2017; 12:e0170389. [PMID: 28103309 PMCID: PMC5245900 DOI: 10.1371/journal.pone.0170389] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 01/04/2017] [Indexed: 02/01/2023] Open
Abstract
Folate deficiency during pregnancy has been related to low birth weight, preterm (PT) birth and other health risks in the offspring; however, it is unknown whether prematurity is related to low folate transport through the placenta due to altered expression of specific folate transporters. We determined placental expression (mRNA and protein concentrations by RT-qPCR and WB respectively) of specific folate transporters: RFC, PCFT/HCP1 and FOLR1 in chorionic (fetal) and basal (maternal) plates of placentas of PT pregnancies (PT, 32–36 weeks, n = 51). Term placentas were used as controls (T, 37–41 weeks, n = 47). Folates and vitamin B12 levels were measured by electrochemiluminescence in umbilical cord blood of newborns. FOLR1 mRNA expression was lower and protein concentration higher in PT placentas (both plates) relative to the control group (p <0.05). In addition, gestational age was positively correlated with mRNA expression (Rho = 0.7), and negatively with protein concentration (Rho = -0.7 for chorionic and -0.43 for basal plate). PCFT/HCP1 mRNA was lower in PT placentas, without changes in protein levels. RFC did not differ in PT placentas compared to controls. PT newborns presented higher cord blood folate level (p = 0.049) along with lower vitamin B12 concentration compared to controls (p = 0.037).In conclusion, placental FOLR1 mRNA was positively associated with gestational age. Conversely, FOLR1 protein concentrations along with folate/vitamin B12 ratio in cord blood were negatively associated with gestational age. Placental FOLR1 is likely the main placental folate transporter to the fetus in newborns.
Collapse
Affiliation(s)
- Erika Castaño
- Laboratory of Nutrition and Metabolic Regulation, Human Nutrition Unit, Institute of Nutrition and Food Technology, Dr. Fernando Monckeberg Barros (INTA), University of Chile, Santiago, Chile
| | - Lorena Caviedes
- Laboratory of Nutrition and Metabolic Regulation, Human Nutrition Unit, Institute of Nutrition and Food Technology, Dr. Fernando Monckeberg Barros (INTA), University of Chile, Santiago, Chile
| | - Sandra Hirsch
- Laboratory of Nutrition and Metabolic Regulation, Human Nutrition Unit, Institute of Nutrition and Food Technology, Dr. Fernando Monckeberg Barros (INTA), University of Chile, Santiago, Chile
| | - Miguel Llanos
- Laboratory of Nutrition and Metabolic Regulation, Human Nutrition Unit, Institute of Nutrition and Food Technology, Dr. Fernando Monckeberg Barros (INTA), University of Chile, Santiago, Chile
| | - Germán Iñiguez
- Mother and Child Research Institute, Division of Medical Sciences, School of Medicine, University of Chile, Santiago, Chile
| | - Ana María Ronco
- Laboratory of Nutrition and Metabolic Regulation, Human Nutrition Unit, Institute of Nutrition and Food Technology, Dr. Fernando Monckeberg Barros (INTA), University of Chile, Santiago, Chile
- * E-mail:
| |
Collapse
|
38
|
Palmer D. Early Nutrition and its Effect on Allergy Development. EARLY NUTRITION AND LONG-TERM HEALTH 2017:175-201. [DOI: 10.1016/b978-0-08-100168-4.00007-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
39
|
Folate status, regulatory T cells and MTHFR C677T polymorphism study in allergic children. Adv Med Sci 2016; 61:300-305. [PMID: 27149557 DOI: 10.1016/j.advms.2016.03.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 03/01/2016] [Accepted: 03/30/2016] [Indexed: 12/22/2022]
Abstract
PURPOSE This study aimed to investigate early-life folate serum concentrations in children with food, inhalant or mixed type allergy. The influence of folate levels on the FoxP3 expression in Treg (regulatory T) cells in the studied children, taking into account the MTHFR (5,10-methylenetetrahydrofolate reductase) genotypes was also analyzed. MATERIAL AND METHODS The study was performed in 83 allergic children (study group) and 49 healthy children (control group), aged 2-72 months. Medical history of each child was obtained and laboratory tests (serum folic acid concentrations and MTHFR C677T polymorphism) were carried out. The percentage of Treg cells was evaluated in almost a half of the examined subjects (48.5%). RESULTS Significantly higher serum folate levels in the group of children with food allergy than in those with inhalant allergy was confirmed (P=0.037). In the study group the TT homozygotes were characterized by significantly lower folate concentrations than CC homozygotes (P=0.045). A negative correlation was demonstrated between the FoxP3 expression in CD4+CD25highFoxP3+ peripheral blood lymphocytes and serum folic acid concentrations. The correlation was more pronounced in the group of allergic children and it was statistically significant (r=-0.339, P<0.05). CONCLUSIONS The results of the study indicate a possibility of some effects of folate status on Treg cells, thus suggesting their potential role in the development and course of allergy in children.
Collapse
|
40
|
Maternal dietary folate, folic acid and vitamin D intakes during pregnancy and lactation and the risk of cows’ milk allergy in the offspring. Br J Nutr 2016; 116:710-8. [DOI: 10.1017/s0007114516002464] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
AbstractMaternal nutrient intake during pregnancy and lactation potentially influences the development of allergic diseases. Cows’ milk allergy (CMA) is often the first manifestation of atopic diseases, but the impact of early nutritional influences on CMA has not been explored. The associations between maternal intakes of folate, folic acid and vitamin D during pregnancy and lactation were addressed in a prospective, population-based birth cohort within the Finnish Type 1 Diabetes Prediction and Prevention Study. Mothers of 4921 children during pregnancy and 2940 children during lactation provided information on maternal dietary intake during the 8th month of pregnancy and the 3rd month of lactation using a detailed, validated FFQ. Information on diagnosed CMA in the offspring was obtained from a medical registry as well as queried from the parents. The Finnish food composition database was used to calculate nutrient intake. Logistic regression was applied for statistical analyses. Folate intake and folic acid and vitamin D supplement use were associated with an increased risk of CMA in the offspring, whereas vitamin D intake from foods during pregnancy was associated with a decreased risk of CMA. Thus, maternal nutrient intake during pregnancy and lactation may affect the development of CMA in offspring. Supplementation with folic acid may not be beneficial in terms of CMA development, especially in children of allergic mothers. The association between dietary supplement use and CMA risk can at least partly be explained by increased health-seeking behaviour among more educated mothers who also use more dietary supplements.
Collapse
|
41
|
Gazibara T, Elbert NJ, den Dekker HT, de Jongste JC, Reiss I, McGrath JJ, Eyles DW, Burne TH, Tiemeier H, Jaddoe VWV, Pasmans SGMA, Duijts L. Associations of maternal and fetal 25-hydroxyvitamin D levels with childhood eczema: The Generation R Study. Pediatr Allergy Immunol 2016; 27:283-9. [PMID: 26683760 DOI: 10.1111/pai.12530] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/14/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Exposure to low levels of vitamin D in fetal life might affect the developing immune system, and subsequently the risk of childhood eczema. We examined whether 25-hydroxyvitamin D levels in mid-gestation and at birth were associated with the risk of eczema until the age of 4 years. METHODS In a population-based prospective cohort study of 3019 mothers and their children, maternal blood samples in mid-gestation and umbilical cord blood samples at birth were used to determine 25-hydroxyvitamin D levels (severely deficient <25.0 nmol/l, deficient 25.0-49.9 nmol/l, sufficient 50.0-74.9 nmol/l, optimal ≥75.0 nmol/l). Eczema was prospectively assessed by annual questionnaires until the age of 4 years. Eczema patterns included never, early (age ≤1 year only), late (age >1 year only), and persistent eczema (age ≤ and >1 year). Data were assessed using the generalized estimating equations and multinomial regression models. RESULTS Compared with the optimal 25-hydroxyvitamin D group, sufficient, deficient, and severely deficient groups of 25-hydroxyvitamin D level in mid-gestation were not associated with the risk of overall eczema (odds ratios [95% confidence interval]: 1.09 [0.82, 1.43], 1.04 [0.87, 1.25], and 0.94 [0.81, 1.10], p-values for trend >0.05), nor with eczema per year or eczema patterns in children up to the age of 4 years. Similarly, we observed no associations of 25-hydroxyvitamin D groups at birth with any eczema outcome. CONCLUSION Our results suggest that levels of 25-hydroxyvitamin D in mid-gestation and at birth are not associated with the risk of overall eczema, eczema per year, or eczema patterns among children until the age of 4 years.
Collapse
Affiliation(s)
- Tatjana Gazibara
- 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.,Institute of Epidemiology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Niels J Elbert
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Dermatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Herman 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
| | - Johan C de Jongste
- Department of Pediatrics, Division of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Irwin Reiss
- Department of Pediatrics, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - John J McGrath
- Queensland Brain Institute, The University of Queensland, Brisbane, Qld, Australia.,Queensland Centre for Mental Health Research, Park Centre for Mental Health, Wacol, Qld, Australia
| | - Darryl W Eyles
- Queensland Brain Institute, The University of Queensland, Brisbane, Qld, Australia.,Queensland Centre for Mental Health Research, Park Centre for Mental Health, Wacol, Qld, Australia
| | - Thomas H Burne
- Queensland Brain Institute, The University of Queensland, Brisbane, Qld, Australia.,Queensland Centre for Mental Health Research, Park Centre for Mental Health, Wacol, Qld, Australia
| | - Henning Tiemeier
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Psychiatry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Vincent 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
| | - Suzanne G M A Pasmans
- Department of Dermatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Liesbeth 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
|
42
|
Folic acid supplementation: what is new? Fetal, obstetric, long-term benefits and risks. Future Sci OA 2016; 2:FSO116. [PMID: 28031963 PMCID: PMC5137972 DOI: 10.4155/fsoa-2015-0015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 02/24/2016] [Indexed: 01/08/2023] Open
Abstract
The association between folic acid supplementation, prior to conception and/or during pregnancy and pregnancy outcomes, has been the subject of numerous studies. The worldwide recommendation of folic acid is at least 0.4 mg daily for all women of reproductive age, and 4–5 mg in high-risk women. In addition, evidence shows that folic acid supplementation could modulate other adverse pregnancy outcomes, specifically, in pregnancies complicated by seizure disorders, preeclampsia, anemia, fetal growth restriction and autism. This review summarizes the available national and international guidelines, concerning the indications and dosage of folic acid supplementation during pregnancy. In addition, it describes the potential preventive benefits of folic acid supplementation on multiple maternal and fetal outcomes, as well as potential risks.
Collapse
|
43
|
Grieger JA, Clifton VL, Tuck AR, Wooldridge AL, Robertson SA, Gatford KL. In utero Programming of Allergic Susceptibility. Int Arch Allergy Immunol 2016; 169:80-92. [PMID: 27044002 DOI: 10.1159/000443961] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Around 30-40% of the world's population will experience allergy, the most common and earliest-onset noncommunicable disease. With a steady rise in the incidence of allergic disease over recent decades, up to 18% of children will suffer a respiratory, food or skin allergy before their 18th birthday. There is compelling evidence that the risk of developing allergy is influenced by early life events and particularly in utero exposures. METHODS A comprehensive literature review was undertaken which outlines prenatal risk factors and potential mechanisms underlying the development of allergy in childhood. RESULTS Exposures including maternal cigarette smoking, preterm birth and Caesarean delivery are implicated in predisposing infants to the later development of allergy. In contrast, restricted growth in utero, a healthy maternal diet and a larger family size are protective, but the mechanisms here are unclear and require further investigation. CONCLUSION To ameliorate the allergy pandemic in young children, we must define prenatal mechanisms that alter the programming of the fetal immune system and also identify specific targets for antenatal interventions.
Collapse
Affiliation(s)
- Jessica A Grieger
- Robinson Research Institute and School of Medicine, University ofAdelaide, Adelaide, S.A., Australia
| | | | | | | | | | | |
Collapse
|
44
|
Beckhaus AA, Garcia-Marcos L, Forno E, Pacheco-Gonzalez RM, Celedón JC, Castro-Rodriguez JA. Maternal nutrition during pregnancy and risk of asthma, wheeze, and atopic diseases during childhood: a systematic review and meta-analysis. Allergy 2015; 70:1588-604. [PMID: 26296633 DOI: 10.1111/all.12729] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2015] [Indexed: 02/02/2023]
Abstract
BACKGROUND Epidemiologic studies suggest a relationship between maternal nutrition during pregnancy and the occurrence of asthma and atopic conditions during childhood. However, individual study results are conflicting. The objective of this meta-analysis was to critically examine the current evidence for an association between nutrition (dietary patterns, food groups, vitamins, or oligo-elements) ingestion during pregnancy and asthma, wheeze, or atopic conditions in childhood. METHODS The inclusion criteria were as follows: (i) systematic recording of diet during the gestational period and (ii) documentation of asthma, wheezing, eczema, or other atopic disease in the offspring. The primary outcomes were prevalence of asthma or wheeze among the offspring during childhood; and secondary outcomes were prevalence of eczema, allergic rhinitis, or other atopic conditions. RESULTS We found 120 titles, abstracts, and citations, and 32 studies (29 cohorts) were included in this analysis. Data on vitamins, oligo-elements, food groups, and dietary patterns during pregnancy were collected. A meta-analysis revealed that higher maternal intake of vitamin D [odds ratio (OR) = 0.58, 95% confidence interval (CI) = 0.38-0.88], vitamin E (OR = 0.6, 95% CI = 0.46-0.78), and zinc (OR = 0.62, 95% CI = 0.40-0.97) was associated with lower odds of wheeze during childhood. However, none of these or other nutrients was consistently associated with asthma per se or other atopic conditions. CONCLUSIONS Current evidence suggests a protective effect of maternal intake of each of three vitamins or nutrients (vitamin D, vitamin E, and zinc) against childhood wheeze but is inconclusive for an effect on asthma or other atopic conditions.
Collapse
Affiliation(s)
- A. A. Beckhaus
- Division of Pediatrics; School of Medicine; Pontificia Universidad Católica de Chile; Santiago Chile
| | - L. Garcia-Marcos
- Respiratory and Allergy Units; Arrixaca University Children's Hospital; University of Murcia; Murcia Spain
- IMIB Bio-research Institute; Murcia Spain
| | - E. Forno
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology; Children's Hospital of Pittsburgh of UPMC; University of Pittsburgh; Pittsburgh PA USA
| | - R. M. Pacheco-Gonzalez
- Respiratory and Allergy Units; Arrixaca University Children's Hospital; University of Murcia; Murcia Spain
- IMIB Bio-research Institute; Murcia Spain
| | - J. C. Celedón
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology; Children's Hospital of Pittsburgh of UPMC; University of Pittsburgh; Pittsburgh PA USA
| | - J. A. Castro-Rodriguez
- Division of Pediatrics; School of Medicine; Pontificia Universidad Católica de Chile; Santiago Chile
| |
Collapse
|
45
|
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
|
46
|
Abstract
It has been recognized for centuries that allergic disease runs in families, implying a role for genetic factors in determining individual susceptibility. More recently, a range of evidence shows that many of these genetic factors, together with in utero environmental exposures, lead to the development of allergic disease through altered immune and organ development. Environmental exposures during pregnancy including diet, nutrient intake and toxin exposures can alter the epigenome and interact with inherited genetic and epigenetic risk factors to directly and indirectly influence organ development and immune programming. Understanding of these factors will be essential in identifying at-risk individuals and possible development of therapeutic interventions for the primary prevention of allergic disease. In this review, we summarize the evidence that suggests allergic disease begins in utero, together with possible mechanisms for the effect of environmental exposures during pregnancy on allergic disease risk, including epigenetics.
Collapse
Affiliation(s)
- Gabrielle A Lockett
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Johanna Huoman
- Department of Clinical and Experimental Medicine, Division of Neuro and Inflammation Sciences, Unit of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,International Inflammation network (in-FLAME) of the World Universities Network
| |
Collapse
|
47
|
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
|
48
|
Campbell DE, Boyle RJ, Thornton CA, Prescott SL. Mechanisms of allergic disease - environmental and genetic determinants for the development of allergy. Clin Exp Allergy 2015; 45:844-858. [DOI: 10.1111/cea.12531] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- D. E. Campbell
- Children's Hospital Westmead; Sydney NSW Australia
- Discipline of Paediatrics and Child Health; University of Sydney; Sydney NSW Australia
| | - R. J. Boyle
- Section of Paediatrics; Faculty of Medicine; Imperial College; London UK
| | - C. A. Thornton
- Institute of Life Science; College of Medicine; Swansea University; Swansea UK
| | - S. L. Prescott
- School of Paediatrics and Child Health and Telethon KIDS Institute; c/o Princess Margaret Hospital; University of Western Australia; Perth WA Australia
| |
Collapse
|
49
|
Wegienka G, Zoratti E, Johnson CC. The role of the early-life environment in the development of allergic disease. Immunol Allergy Clin North Am 2015; 35:1-17. [PMID: 25459574 PMCID: PMC4427897 DOI: 10.1016/j.iac.2014.09.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A consensus has been reached that the development of allergic disorders is strongly influenced by early life exposures. An overview of several prenatal and early life factors that have been investigated for their associations with development of childhood allergy is presented. Delivery mode, the gut microbiome, vitamin D, folate, breastfeeding, pets, antibiotics, environmental tobacco smoke, and airborne traffic pollutants are discussed. Although many studies suggest an effect, overall, no risk factors clearly increase or reduce the risk of allergic outcomes.
Collapse
Affiliation(s)
- Ganesa Wegienka
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, MI, USA.
| | - Edward Zoratti
- Division of Allergy and Immunology, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | | |
Collapse
|
50
|
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
|