351
|
Bailey LB, Stover PJ, McNulty H, Fenech MF, Gregory JF, Mills JL, Pfeiffer CM, Fazili Z, Zhang M, Ueland PM, Molloy AM, Caudill MA, Shane B, Berry RJ, Bailey RL, Hausman DB, Raghavan R, Raiten DJ. Biomarkers of Nutrition for Development-Folate Review. J Nutr 2015; 145:1636S-1680S. [PMID: 26451605 PMCID: PMC4478945 DOI: 10.3945/jn.114.206599] [Citation(s) in RCA: 352] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/11/2014] [Accepted: 04/14/2015] [Indexed: 12/13/2022] Open
Abstract
The Biomarkers of Nutrition for Development (BOND) project is designed to provide evidence-based advice to anyone with an interest in the role of nutrition in health. Specifically, the BOND program provides state-of-the-art information and service with regard to selection, use, and interpretation of biomarkers of nutrient exposure, status, function, and effect. To accomplish this objective, expert panels are recruited to evaluate the literature and to draft comprehensive reports on the current state of the art with regard to specific nutrient biology and available biomarkers for assessing nutrients in body tissues at the individual and population level. Phase I of the BOND project includes the evaluation of biomarkers for 6 nutrients: iodine, iron, zinc, folate, vitamin A, and vitamin B-12. This review represents the second in the series of reviews and covers all relevant aspects of folate biology and biomarkers. The article is organized to provide the reader with a full appreciation of folate's history as a public health issue, its biology, and an overview of available biomarkers (serum folate, RBC folate, and plasma homocysteine concentrations) and their interpretation across a range of clinical and population-based uses. The article also includes a list of priority research needs for advancing the area of folate biomarkers related to nutritional health status and development.
Collapse
Affiliation(s)
- Lynn B Bailey
- Department of Foods and Nutrition, University of Georgia, Athens, GA;
| | - Patrick J Stover
- Division of Nutritional Sciences, Cornell University, Ithaca, NY
| | - Helene McNulty
- Northern Ireland Centre for Food and Health, Biomedical Sciences Research Institute, University of Ulster, Londonderry, United Kingdom
| | - Michael F Fenech
- Genome Health Nutrigenomics Laboratory, Food, Nutrition, and Bioproducts Flagship, Commonwealth Scientific and Industrial Research Organization, Adelaide, Australia
| | - Jesse F Gregory
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL
| | - James L Mills
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD
| | | | - Zia Fazili
- National Center for Environmental Health, CDC, Atlanta, GA
| | - Mindy Zhang
- National Center for Environmental Health, CDC, Atlanta, GA
| | - Per M Ueland
- Department of Clinical Science, Univeristy of Bergen, Bergen, Norway
| | - Anne M Molloy
- Institute of Molecular Medicine, Trinity College, Dublin, Ireland
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, NY
| | - Barry Shane
- Department of Nutritional Sciences and Toxicology, University of California-Berkeley, Berkeley, CA
| | - Robert J Berry
- National Center on Birth Defects and Developmental Disabilities, CDC, Atlanta, GA; and
| | | | - Dorothy B Hausman
- Department of Foods and Nutrition, University of Georgia, Athens, GA
| | - Ramkripa Raghavan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD
| | - Daniel J Raiten
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD;
| |
Collapse
|
352
|
Al Rakaf MS, Kurdi AM, Ammari AN, Al Hashem AM, Shoukri MM, Garne E, Majeed-Saidan MA. Patterns of folic acid use in pregnant Saudi women and prevalence of neural tube defects - Results from a nested case-control study. Prev Med Rep 2015; 2:572-6. [PMID: 26844119 PMCID: PMC4721389 DOI: 10.1016/j.pmedr.2015.06.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Although the role of folic acid (FA) in preventing neural tube defects (NTDs) is well documented, its optimal intake in pregnant women is still low in many countries. Here, we prospectively studied the prevalence of NTDs in the newborns and the patterns of FA intake in pregnant Saudi mothers. METHODS This case-control study was nested within a 3-year project (July 2010 to June 2013) to study the patterns of birth defects in the offspring of Saudi women who received their antenatal care and delivered at Prince Sultan Military Medical City, Riyadh-Saudi Arabia. Enrolled mothers were divided into 4 groups: group 1 (FA taken before pregnancy and continued regularly after conception), group 2 (FA taken post-conception), group 3 (no FA intake), and group 4 (did not remember or were unsure of taking FA). Control mothers were randomly selected from those with normal first obstetrical ultrasound scan at 18-22 weeks of gestation. RESULTS The cohort included 30,531 mothers giving birth to 28,646 infants. We studied 1179 mothers of babies with birth defects (BDs) and 1262 control mothers. There were 237 (9.7%) mothers in-group 1; 2001 (82%) in-group 2; 154 (6.3%) in-group 3; and 49 (2%) in-group 4. There were 49 babies with NTDs, a prevalence of 1.7/1000 total births. Among the studied mothers 2274 (93%) took FA either full or partial course. CONCLUSION The high prevalence of NTDs and the low optimal FA intake highlight the need for a strict implementation of staple food fortification and health education program for Saudi women.
Collapse
Affiliation(s)
- Maha S. Al Rakaf
- Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Ahmed M. Kurdi
- Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Amer N. Ammari
- Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Amal M. Al Hashem
- Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia
| | | | - Ester Garne
- Hospital Lillebaelt-Kolding, Kolding, Denmark
| | | |
Collapse
|
353
|
Functional variant in methionine synthase reductase intron-1 is associated with pleiotropic congenital malformations. Mol Cell Biochem 2015; 407:51-6. [DOI: 10.1007/s11010-015-2453-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/16/2015] [Indexed: 01/03/2023]
|
354
|
Manolio TA, Abramowicz M, Al-Mulla F, Anderson W, Balling R, Berger AC, Bleyl S, Chakravarti A, Chantratita W, Chisholm RL, Dissanayake VHW, Dunn M, Dzau VJ, Han BG, Hubbard T, Kolbe A, Korf B, Kubo M, Lasko P, Leego E, Mahasirimongkol S, Majumdar PP, Matthijs G, McLeod HL, Metspalu A, Meulien P, Miyano S, Naparstek Y, O'Rourke PP, Patrinos GP, Rehm HL, Relling MV, Rennert G, Rodriguez LL, Roden DM, Shuldiner AR, Sinha S, Tan P, Ulfendahl M, Ward R, Williams MS, Wong JEL, Green ED, Ginsburg GS. Global implementation of genomic medicine: We are not alone. Sci Transl Med 2015; 7:290ps13. [PMID: 26041702 PMCID: PMC4898888 DOI: 10.1126/scitranslmed.aab0194] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Around the world, innovative genomic-medicine programs capitalize on singular capabilities arising from local health care systems, cultural or political milieus, and unusual selected risk alleles or disease burdens. Such individual efforts might benefit from the sharing of approaches and lessons learned in other locales. The U.S. National Human Genome Research Institute and the National Academy of Medicine recently brought together 25 of these groups to compare projects, to examine the current state of implementation and desired near-term capabilities, and to identify opportunities for collaboration that promote the responsible practice of genomic medicine. Efforts to coalesce these groups around concrete but compelling signature projects should accelerate the responsible implementation of genomic medicine in efforts to improve clinical care worldwide.
Collapse
Affiliation(s)
- Teri A Manolio
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-9305, USA.
| | | | - Fahd Al-Mulla
- Genatak-Global Med Clinic, Kuwait University, Kuwait 46300, Kuwait
| | - Warwick Anderson
- National Health and Medical Research Council, Canberra, ACT 2601, Australia
| | - Rudi Balling
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, L-4362 Luxembourg
| | - Adam C Berger
- Board on Health Sciences Policy, Institute of Medicine, Washington, DC 20001, USA
| | - Steven Bleyl
- Intermountain Healthcare, Salt Lake City, UT 84111, USA
| | - Aravinda Chakravarti
- McKusick-Nathans Institute of Genetic Medicine, John Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | - Rex L Chisholm
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Vajira H W Dissanayake
- Human Genetics Unit, Faculty of Medicine, University of Colombo, Colombo 00800, Sri Lanka
| | - Michael Dunn
- Genetic and Molecular Sciences, The Wellcome Trust, London NW1 2BE, UK
| | - Victor J Dzau
- National Academy of Medicine, Washington, DC 20001, USA
| | - Bok-Ghee Han
- Center for Genome Science, Korea National Institute of Health, Chungcheongbuk-do 363-951 Korea
| | - Tim Hubbard
- Department of Medical and Molecular Genetics, King's College, London SE1 9RT, and Genomics England, London EC1M 6BQ, UK
| | - Anne Kolbe
- National Health Committee, Auckland 1050, New Zealand
| | - Bruce Korf
- Center for Genomic Science, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michiaki Kubo
- Center for Integrative Medical Science (IMS), RIKEN, Yokohama 230-0045, Japan
| | - Paul Lasko
- Institute of Genetics, Canadian Institutes of Health Research, and McGill University, Montreal, Quebec, H3A 0G4 Canada
| | - Erkki Leego
- Estonian Genome Center, University of Tartu, Tartu 51010 Estonia
| | | | - Partha P Majumdar
- National Institute of Biomedical Genomics and Indian Statistical Institute, Kalyani 741251 India
| | - Gert Matthijs
- Center for Human Genetics, University of Leuven (KU Leuven), B-3000 Leuven, Belgium
| | - Howard L McLeod
- DeBartolo Family Personalized Medicine Institute, Moffitt Cancer Center, Tampa, FL 33612 USA
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu 51010 Estonia
| | | | - Satoru Miyano
- Institute of Medical Science, University of Tokyo, 108-8639 Tokyo, Japan
| | - Yaakov Naparstek
- Research and Academic Affairs, Hadassah Hebrew University Hospital, Jerusalem 91120, Israel
| | - P Pearl O'Rourke
- Office of Human Research Affairs, Partners HealthCare, Boston, MA 02199, USA
| | - George P Patrinos
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras, 26504 Greece
| | - Heidi L Rehm
- Laboratory for Molecular Medicine, Partners Healthcare Systems, Boston, MA 02139, USA
| | - Mary V Relling
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Gad Rennert
- Carmel Medical Center Department of Community Medicine and Epidemiology, Clalit National Personalized Medicine Program, Haifa 34362, Israel
| | - Laura Lyman Rodriguez
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-9305, USA
| | - Dan M Roden
- Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Alan R Shuldiner
- Program in Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sukdeb Sinha
- Department of Biotechnology, Ministry of Science and Technology, Govt., New Delhi 110 003 India
| | - Patrick Tan
- Duke-National University of Singapore Graduate Medical School, Singapore 169857, Singapore
| | | | - Robyn Ward
- University of Queensland, St. Lucia, QLD 4067 Australia
| | - Marc S Williams
- Genomic Medicine Institute, Geisinger Health System, Danville, PA 18510, USA
| | - John E L Wong
- National University of Singapore, Singapore 119228, Singapore
| | - Eric D Green
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-9305, USA
| | - Geoffrey S Ginsburg
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
355
|
Brito A, Mujica-Coopman MF, Olivares M, López de Romaña D, Cori H, Allen LH. Folate and Vitamin B12 Status in Latin America and the Caribbean. Food Nutr Bull 2015; 36:S109-18. [DOI: 10.1177/0379572115585772] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background: The current magnitude of folate and vitamin B12 deficiency in Latin America and the Caribbean is uncertain. Objective: To summarize data on plasma or serum vitamin B12 and folate concentrations in Latin America and the Caribbean reported since 1990, a period that covers the era before and after the introduction of folic acid fortification. Methods: A systematic review was conducted in 2012 and updated in 2014. Studies and surveys using biochemical biomarkers and conducted in apparently healthy individuals were identified. Results: Folate deficiency in Latin America and the Caribbean appears not to be a public health problem (prevalence < 5%) after the introduction of folic acid fortification. However, there is some indication that high rates of low or marginal vitamin B12 status remain in most locations and across population groups. Conclusions: Adding vitamin B12 as a fortificant with folic acid may be the best strategy in areas where vitamin B12 deficiency is an established concern.
Collapse
Affiliation(s)
- Alex Brito
- US Department of Agriculture Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, USA
| | | | - Manuel Olivares
- Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | | | | | - Lindsay H. Allen
- US Department of Agriculture Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, USA
| |
Collapse
|
356
|
Tinker SC, Hamner HC, Qi YP, Crider KS. U.S. women of childbearing age who are at possible increased risk of a neural tube defect-affected pregnancy due to suboptimal red blood cell folate concentrations, National Health and Nutrition Examination Survey 2007 to 2012. BIRTH DEFECTS RESEARCH. PART A, CLINICAL AND MOLECULAR TERATOLOGY 2015; 103:517-26. [PMID: 25884850 PMCID: PMC4515959 DOI: 10.1002/bdra.23378] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/10/2015] [Indexed: 01/21/2023]
Abstract
BACKGROUND Red blood cell (RBC) folate concentrations are a potential biomarker of folate-sensitive neural tube defect (NTD) risk in the population. The purpose of this analysis was to describe women in the U.S. population with RBC folate concentrations below those associated with optimal NTD prevention. METHODS We used data from the 2007 to 2012 National Health and Nutrition Examination Survey (NHANES) to assess the RBC folate status of U.S. women of childbearing age relative to risk categories for NTD risk based on RBC folate concentrations. We defined suboptimal RBC folate concentrations as those associated with a prevalence of ≥9 NTDs per 10,000 live births. RESULTS Among nonpregnant women age 12 to 49 years, 22.8% (95% Confidence Interval: 21.1, 24.6) had suboptimal RBC folate concentrations. Women had greater odds of having a suboptimal RBC folate concentration if they did not use dietary supplements containing folic acid; had mandatorily fortified enriched cereal grain products as their only source of folic acid; were non-Hispanic black or Hispanic; or were current smokers. CONCLUSION Based on RBC folate concentrations, we would predict that the majority of U.S. women of reproductive age are not at increased risk for folate sensitive NTDs in the presence of mandatory folic acid fortification. Prevention policies and programs can be aimed at population subgroups identified as having higher predicted risk for folate-sensitive NTDs based on RBC folate concentrations.
Collapse
Affiliation(s)
- Sarah C. Tinker
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia
| | - Heather C. Hamner
- National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Yan Ping Qi
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia
| | - Krista S. Crider
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia
| |
Collapse
|
357
|
Tsang BL, Devine OJ, Cordero AM, Marchetta CM, Mulinare J, Mersereau P, Guo J, Qi YP, Berry RJ, Rosenthal J, Crider KS, Hamner HC. Assessing the association between the methylenetetrahydrofolate reductase (MTHFR) 677C>T polymorphism and blood folate concentrations: a systematic review and meta-analysis of trials and observational studies. Am J Clin Nutr 2015; 101:1286-94. [PMID: 25788000 DOI: 10.3945/ajcn.114.099994] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 02/09/2015] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND The methylenetetrahydrofolate reductase (MTHFR) 677C>T polymorphism is a risk factor for neural tube defects. The T allele produces an enzyme with reduced folate-processing capacity, which has been associated with lower blood folate concentrations. OBJECTIVE We assessed the association between MTHFR C677T genotypes and blood folate concentrations among healthy women aged 12-49 y. DESIGN We conducted a systematic review of the literature published from January 1992 to March 2014 to identify trials and observational studies that reported serum, plasma, or red blood cell (RBC) folate concentrations and MTHFR C677T genotype. We conducted a meta-analysis for estimates of percentage differences in blood folate concentrations between genotypes. RESULTS Forty studies met the inclusion criteria. Of the 6 studies that used the microbiologic assay (MA) to measure serum or plasma (S/P) and RBC folate concentrations, the percentage difference between genotypes showed a clear pattern of CC > CT > TT. The percentage difference was greatest for CC > TT [S/P: 13%; 95% credible interval (CrI): 7%, 18%; RBC: 16%; 95% CrI: 12%, 20%] followed by CC > CT (S/P: 7%; 95% CrI: 1%, 12%; RBC: 8%; 95% CrI: 4%, 12%) and CT > TT (S/P: 6%; 95% CrI: 1%, 11%; RBC: 9%; 95% CrI: 5%, 13%). S/P folate concentrations measured by using protein-binding assays (PBAs) also showed this pattern but to a greater extent (e.g., CC > TT: 20%; 95% CrI: 17%, 22%). In contrast, RBC folate concentrations measured by using PBAs did not show the same pattern and are presented in the Supplemental Material only. CONCLUSIONS Meta-analysis results (limited to the MA, the recommended population assessment method) indicated a consistent percentage difference in S/P and RBC folate concentrations across MTHFR C677T genotypes. Lower blood folate concentrations associated with this polymorphism could have implications for a population-level risk of neural tube defects.
Collapse
Affiliation(s)
- Becky L Tsang
- From the Division of Birth Defects and Developmental Disabilities, National Center for Birth Defects and Developmental Disabilities (NCBDDD), CDC, Atlanta, GA (AMC, RJB, JR, and KSC); the Division of Nutrition, Physical Activity, and Obesity, National Center on Chronic Disease Prevention and Health Promotion, CDC, Atlanta, GA (HCH); Acentia (contractor for CDC, NCBDDD), Falls Church, VA (JG); Carter Consulting Inc. (contractor for CDC, NCBDDD), Atlanta, GA (OJD, JM, and YPQ); the Oak Ridge Institute for Science and Education, Oak Ridge, TN (BLT, CMM, and YPQ); SciMetrika LLC (contractor for CDC, NCBDDD), Atlanta, GA (PM)
| | - Owen J Devine
- From the Division of Birth Defects and Developmental Disabilities, National Center for Birth Defects and Developmental Disabilities (NCBDDD), CDC, Atlanta, GA (AMC, RJB, JR, and KSC); the Division of Nutrition, Physical Activity, and Obesity, National Center on Chronic Disease Prevention and Health Promotion, CDC, Atlanta, GA (HCH); Acentia (contractor for CDC, NCBDDD), Falls Church, VA (JG); Carter Consulting Inc. (contractor for CDC, NCBDDD), Atlanta, GA (OJD, JM, and YPQ); the Oak Ridge Institute for Science and Education, Oak Ridge, TN (BLT, CMM, and YPQ); SciMetrika LLC (contractor for CDC, NCBDDD), Atlanta, GA (PM)
| | - Amy M Cordero
- From the Division of Birth Defects and Developmental Disabilities, National Center for Birth Defects and Developmental Disabilities (NCBDDD), CDC, Atlanta, GA (AMC, RJB, JR, and KSC); the Division of Nutrition, Physical Activity, and Obesity, National Center on Chronic Disease Prevention and Health Promotion, CDC, Atlanta, GA (HCH); Acentia (contractor for CDC, NCBDDD), Falls Church, VA (JG); Carter Consulting Inc. (contractor for CDC, NCBDDD), Atlanta, GA (OJD, JM, and YPQ); the Oak Ridge Institute for Science and Education, Oak Ridge, TN (BLT, CMM, and YPQ); SciMetrika LLC (contractor for CDC, NCBDDD), Atlanta, GA (PM)
| | - Claire M Marchetta
- From the Division of Birth Defects and Developmental Disabilities, National Center for Birth Defects and Developmental Disabilities (NCBDDD), CDC, Atlanta, GA (AMC, RJB, JR, and KSC); the Division of Nutrition, Physical Activity, and Obesity, National Center on Chronic Disease Prevention and Health Promotion, CDC, Atlanta, GA (HCH); Acentia (contractor for CDC, NCBDDD), Falls Church, VA (JG); Carter Consulting Inc. (contractor for CDC, NCBDDD), Atlanta, GA (OJD, JM, and YPQ); the Oak Ridge Institute for Science and Education, Oak Ridge, TN (BLT, CMM, and YPQ); SciMetrika LLC (contractor for CDC, NCBDDD), Atlanta, GA (PM)
| | - Joseph Mulinare
- From the Division of Birth Defects and Developmental Disabilities, National Center for Birth Defects and Developmental Disabilities (NCBDDD), CDC, Atlanta, GA (AMC, RJB, JR, and KSC); the Division of Nutrition, Physical Activity, and Obesity, National Center on Chronic Disease Prevention and Health Promotion, CDC, Atlanta, GA (HCH); Acentia (contractor for CDC, NCBDDD), Falls Church, VA (JG); Carter Consulting Inc. (contractor for CDC, NCBDDD), Atlanta, GA (OJD, JM, and YPQ); the Oak Ridge Institute for Science and Education, Oak Ridge, TN (BLT, CMM, and YPQ); SciMetrika LLC (contractor for CDC, NCBDDD), Atlanta, GA (PM)
| | - Patricia Mersereau
- From the Division of Birth Defects and Developmental Disabilities, National Center for Birth Defects and Developmental Disabilities (NCBDDD), CDC, Atlanta, GA (AMC, RJB, JR, and KSC); the Division of Nutrition, Physical Activity, and Obesity, National Center on Chronic Disease Prevention and Health Promotion, CDC, Atlanta, GA (HCH); Acentia (contractor for CDC, NCBDDD), Falls Church, VA (JG); Carter Consulting Inc. (contractor for CDC, NCBDDD), Atlanta, GA (OJD, JM, and YPQ); the Oak Ridge Institute for Science and Education, Oak Ridge, TN (BLT, CMM, and YPQ); SciMetrika LLC (contractor for CDC, NCBDDD), Atlanta, GA (PM)
| | - Jing Guo
- From the Division of Birth Defects and Developmental Disabilities, National Center for Birth Defects and Developmental Disabilities (NCBDDD), CDC, Atlanta, GA (AMC, RJB, JR, and KSC); the Division of Nutrition, Physical Activity, and Obesity, National Center on Chronic Disease Prevention and Health Promotion, CDC, Atlanta, GA (HCH); Acentia (contractor for CDC, NCBDDD), Falls Church, VA (JG); Carter Consulting Inc. (contractor for CDC, NCBDDD), Atlanta, GA (OJD, JM, and YPQ); the Oak Ridge Institute for Science and Education, Oak Ridge, TN (BLT, CMM, and YPQ); SciMetrika LLC (contractor for CDC, NCBDDD), Atlanta, GA (PM)
| | - Yan Ping Qi
- From the Division of Birth Defects and Developmental Disabilities, National Center for Birth Defects and Developmental Disabilities (NCBDDD), CDC, Atlanta, GA (AMC, RJB, JR, and KSC); the Division of Nutrition, Physical Activity, and Obesity, National Center on Chronic Disease Prevention and Health Promotion, CDC, Atlanta, GA (HCH); Acentia (contractor for CDC, NCBDDD), Falls Church, VA (JG); Carter Consulting Inc. (contractor for CDC, NCBDDD), Atlanta, GA (OJD, JM, and YPQ); the Oak Ridge Institute for Science and Education, Oak Ridge, TN (BLT, CMM, and YPQ); SciMetrika LLC (contractor for CDC, NCBDDD), Atlanta, GA (PM)
| | - Robert J Berry
- From the Division of Birth Defects and Developmental Disabilities, National Center for Birth Defects and Developmental Disabilities (NCBDDD), CDC, Atlanta, GA (AMC, RJB, JR, and KSC); the Division of Nutrition, Physical Activity, and Obesity, National Center on Chronic Disease Prevention and Health Promotion, CDC, Atlanta, GA (HCH); Acentia (contractor for CDC, NCBDDD), Falls Church, VA (JG); Carter Consulting Inc. (contractor for CDC, NCBDDD), Atlanta, GA (OJD, JM, and YPQ); the Oak Ridge Institute for Science and Education, Oak Ridge, TN (BLT, CMM, and YPQ); SciMetrika LLC (contractor for CDC, NCBDDD), Atlanta, GA (PM)
| | - Jorge Rosenthal
- From the Division of Birth Defects and Developmental Disabilities, National Center for Birth Defects and Developmental Disabilities (NCBDDD), CDC, Atlanta, GA (AMC, RJB, JR, and KSC); the Division of Nutrition, Physical Activity, and Obesity, National Center on Chronic Disease Prevention and Health Promotion, CDC, Atlanta, GA (HCH); Acentia (contractor for CDC, NCBDDD), Falls Church, VA (JG); Carter Consulting Inc. (contractor for CDC, NCBDDD), Atlanta, GA (OJD, JM, and YPQ); the Oak Ridge Institute for Science and Education, Oak Ridge, TN (BLT, CMM, and YPQ); SciMetrika LLC (contractor for CDC, NCBDDD), Atlanta, GA (PM)
| | - Krista S Crider
- From the Division of Birth Defects and Developmental Disabilities, National Center for Birth Defects and Developmental Disabilities (NCBDDD), CDC, Atlanta, GA (AMC, RJB, JR, and KSC); the Division of Nutrition, Physical Activity, and Obesity, National Center on Chronic Disease Prevention and Health Promotion, CDC, Atlanta, GA (HCH); Acentia (contractor for CDC, NCBDDD), Falls Church, VA (JG); Carter Consulting Inc. (contractor for CDC, NCBDDD), Atlanta, GA (OJD, JM, and YPQ); the Oak Ridge Institute for Science and Education, Oak Ridge, TN (BLT, CMM, and YPQ); SciMetrika LLC (contractor for CDC, NCBDDD), Atlanta, GA (PM)
| | - Heather C Hamner
- From the Division of Birth Defects and Developmental Disabilities, National Center for Birth Defects and Developmental Disabilities (NCBDDD), CDC, Atlanta, GA (AMC, RJB, JR, and KSC); the Division of Nutrition, Physical Activity, and Obesity, National Center on Chronic Disease Prevention and Health Promotion, CDC, Atlanta, GA (HCH); Acentia (contractor for CDC, NCBDDD), Falls Church, VA (JG); Carter Consulting Inc. (contractor for CDC, NCBDDD), Atlanta, GA (OJD, JM, and YPQ); the Oak Ridge Institute for Science and Education, Oak Ridge, TN (BLT, CMM, and YPQ); SciMetrika LLC (contractor for CDC, NCBDDD), Atlanta, GA (PM).
| |
Collapse
|
358
|
Bánhidy F, Ács N, Puhó EH, Czeizel AE. Paroxysmal supraventricular tachycardia in pregnant women and birth outcomes of their children: A population-based study. Am J Med Genet A 2015; 167A:1779-86. [DOI: 10.1002/ajmg.a.33759] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 09/12/2010] [Indexed: 11/12/2022]
Affiliation(s)
- Ferenc Bánhidy
- Second Department of Obstetrics and Gynecology; Semmelweis University; School of Medicine; Budapest Hungary
| | - Nándor Ács
- Second Department of Obstetrics and Gynecology; Semmelweis University; School of Medicine; Budapest Hungary
| | - Erzsébet H. Puhó
- Foundation for the Community Control of Hereditary Diseases; Budapest Hungary
| | - Andrew E. Czeizel
- Foundation for the Community Control of Hereditary Diseases; Budapest Hungary
| |
Collapse
|
359
|
Abstract
The majority of neural tube defects were believed to be folic acid (FA)-preventable in the 1990s. The Japanese government recommended women planning pregnancy to take FA supplements of 400 μg/d in 2000, but the incidence of spina bifida has not decreased. We aimed to evaluate the OR of having an infant with spina bifida for women who periconceptionally took FA supplements and the association between an increase in supplement use and possible promoters for the increase. This is a case-control study which used 360 case women who gave birth to newborns afflicted with spina bifida, and 2333 control women who gave birth to healthy newborns during the first 12 years of this century. They were divided into two 6-year periods; from 2001 to 2006 and from 2007 to 2012. Logistic regression analyses were conducted to compute OR between cases and controls. The adjusted OR of having an infant with spina bifida for supplement users was 0.48 in the first period, and 0.53 in the second period. The proportion of women who periconceptionally consumed supplements significantly increased from 10 % in the first period to 30 % in the second period. Awareness of the preventive role of FA was a promoter for an increase in supplement use, and thus an FA campaign in high school seems rational and effective. The failure of the current public health policy is responsible for an epidemic of spina bifida. Mandatory food fortification with FA is urgent and long overdue in Japan.
Collapse
|
360
|
Zhou Y, Blustein J, Li H, Ye R, Zhu L, Liu J. Maternal obesity, caesarean delivery and caesarean delivery on maternal request: a cohort analysis from China. Paediatr Perinat Epidemiol 2015; 29:232-40. [PMID: 25827169 DOI: 10.1111/ppe.12191] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND To quantify the association between maternal obesity and caesarean delivery, particularly caesarean delivery on maternal request (CDMR), a fast-growing component of caesarean delivery in many nations. METHODS We followed 1,019,576 nulliparous women registered in the Perinatal Healthcare Surveillance System during 1993-2010. Maternal body mass index (BMI, kg/m(2) ), before pregnancy or during early pregnancy, was classified as underweight (<18.5), normal (18.5 to <23; reference), overweight (23 to <27.5), or obese (≥27.5), consistent with World Health Organization guidelines for Asian people. The association between maternal obesity and overall caesarean and its subtypes was modelled using log-binomial regression. RESULTS During the 18-year period, 404,971 (39.7%) caesareans and 93,927 (9.2%) CDMRs were identified. Maternal obesity was positively associated with overall caesarean and CDMR. Adjusted risk ratios for overall caesarean in the four ascending BMI categories were 0.96 [95% confidence interval (CI) 0.94, 0.97], 1.00 (Reference), 1.16 [95% CI 1.14, 1.18], 1.39 [95% CI 1.43, 1.54], and for CDMR were 0.95 [95% CI 0.94, 0.96], 1.00 (Reference), 1.20 [95% CI 1.18, 1.22], 1.48 [95% CI 1.433, 1.54]. Positive associations were consistently found in women residing in southern and northern provinces and in subgroups stratified by year of delivery, urban or rural residence, maternal age, education, level of delivering hospital, and birthweight. CONCLUSIONS In a large Chinese cohort study, maternal obesity was associated with an increased risk of caesarean delivery and its subtypes, including CDMR. Given the rising global prevalence of obesity, and in view of the growth of CDMR, it seems likely that caesarean births will increase, unless there are changes in obstetrical practice.
Collapse
Affiliation(s)
- Yubo Zhou
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | | | | | | | | | | |
Collapse
|
361
|
Mills JL. Preventing folate-related neural tube defects: Problem solved, or not? ACTA ACUST UNITED AC 2015; 103:469-70. [PMID: 25884960 DOI: 10.1002/bdra.23380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 03/24/2015] [Indexed: 11/10/2022]
Affiliation(s)
- James L Mills
- Epidemiology Branch, Division of Intramural Public Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| |
Collapse
|
362
|
Marchetta CM, Devine OJ, Crider KS, Tsang BL, Cordero AM, Qi YP, Guo J, Berry RJ, Rosenthal J, Mulinare J, Mersereau P, Hamner HC. Assessing the association between natural food folate intake and blood folate concentrations: a systematic review and Bayesian meta-analysis of trials and observational studies. Nutrients 2015; 7:2663-86. [PMID: 25867949 PMCID: PMC4425166 DOI: 10.3390/nu7042663] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 03/06/2015] [Accepted: 03/09/2015] [Indexed: 12/25/2022] Open
Abstract
Folate is found naturally in foods or as synthetic folic acid in dietary supplements and fortified foods. Adequate periconceptional folic acid intake can prevent neural tube defects. Folate intake impacts blood folate concentration; however, the dose-response between natural food folate and blood folate concentrations has not been well described. We estimated this association among healthy females. A systematic literature review identified studies (1 1992-3 2014) with both natural food folate intake alone and blood folate concentration among females aged 12-49 years. Bayesian methods were used to estimate regression model parameters describing the association between natural food folate intake and subsequent blood folate concentration. Seven controlled trials and 29 observational studies met the inclusion criteria. For the six studies using microbiologic assay (MA) included in the meta-analysis, we estimate that a 6% (95% Credible Interval (CrI): 4%, 9%) increase in red blood cell (RBC) folate concentration and a 7% (95% CrI: 1%, 12%) increase in serum/plasma folate concentration can occur for every 10% increase in natural food folate intake. Using modeled results, we estimate that a natural food folate intake of ≥ 450 μg dietary folate equivalents (DFE)/day could achieve the lower bound of an RBC folate concentration (~ 1050 nmol/L) associated with the lowest risk of a neural tube defect. Natural food folate intake affects blood folate concentration and adequate intakes could help women achieve a RBC folate concentration associated with a risk of 6 neural tube defects/10,000 live births.
Collapse
Affiliation(s)
- Claire M Marchetta
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN 37831, USA.
| | | | - Krista S Crider
- Division of Birth Defects and Developmental Disabilities (DBDDD), National Center on Birth Defects and Developmental Disabilities (NCBDDD), Centers for Disease Control and Prevention, Atlanta, GA 30329, USA.
| | - Becky L Tsang
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN 37831, USA.
| | - Amy M Cordero
- Division of Birth Defects and Developmental Disabilities (DBDDD), National Center on Birth Defects and Developmental Disabilities (NCBDDD), Centers for Disease Control and Prevention, Atlanta, GA 30329, USA.
| | - Yan Ping Qi
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN 37831, USA.
| | - Jing Guo
- Acentia, Falls Church, VA 22042, USA.
| | - Robert J Berry
- Division of Birth Defects and Developmental Disabilities (DBDDD), National Center on Birth Defects and Developmental Disabilities (NCBDDD), Centers for Disease Control and Prevention, Atlanta, GA 30329, USA.
| | - Jorge Rosenthal
- Division of Birth Defects and Developmental Disabilities (DBDDD), National Center on Birth Defects and Developmental Disabilities (NCBDDD), Centers for Disease Control and Prevention, Atlanta, GA 30329, USA.
| | | | | | - Heather C Hamner
- Division of Nutrition, Physical Activity, and Obesity (DNPAO), National Center for Chronic Disease Prevention and Health Promotion (NCCDPHP), Centers for Disease Control and Prevention, Atlanta, GA 30329, USA.
| |
Collapse
|
363
|
Sargiotto C, Bidondo MP, Liascovich R, Barbero P, Groisman B. Descriptive study on neural tube defects in Argentina. ACTA ACUST UNITED AC 2015; 103:509-16. [DOI: 10.1002/bdra.23372] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 02/09/2015] [Accepted: 02/23/2015] [Indexed: 11/07/2022]
Affiliation(s)
- Carla Sargiotto
- National Registry of Congenital Anomalies of Argentina (RENAC), National Center of Medical Genetics, National Ministry of Health; Buenos Aires Argentina
| | - María Paz Bidondo
- National Registry of Congenital Anomalies of Argentina (RENAC), National Center of Medical Genetics, National Ministry of Health; Buenos Aires Argentina
| | - Rosa Liascovich
- National Registry of Congenital Anomalies of Argentina (RENAC), National Center of Medical Genetics, National Ministry of Health; Buenos Aires Argentina
| | - Pablo Barbero
- National Registry of Congenital Anomalies of Argentina (RENAC), National Center of Medical Genetics, National Ministry of Health; Buenos Aires Argentina
| | - Boris Groisman
- National Registry of Congenital Anomalies of Argentina (RENAC), National Center of Medical Genetics, National Ministry of Health; Buenos Aires Argentina
| |
Collapse
|
364
|
Taylor CM, Atkinson C, Penfold C, Bhattacharya S, Campbell D, Davey Smith G, Leary S, Ness A. Folic acid in pregnancy and mortality from cancer and cardiovascular disease: further follow-up of the Aberdeen folic acid supplementation trial. J Epidemiol Community Health 2015; 69:789-94. [PMID: 25855124 PMCID: PMC4515996 DOI: 10.1136/jech-2014-205324] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/23/2015] [Indexed: 12/31/2022]
Abstract
Background Supplemental periconceptional folic acid is recommended to reduce the risk of fetal neural tube defects. A previous report indicated an elevated risk of breast cancer and all cancer deaths in later life among women randomised by alternate allocation to high-dose (5 mg/day) folic acid in pregnancy compared with placebo; however, findings were based on small numbers of cases. Our aim was to extend the previous analysis by including data from an additional 10 years of follow-up. Methods Records of participants in a large (n=2928) trial of folate supplementation (5 or 0.2 mg folic acid, or placebo) in pregnancy in the 1960s were linked to central registries in Scotland. Unadjusted and adjusted HRs were calculated for all-cause, cardiovascular, all cancer and breast cancer mortality, and all cancer and breast cancer morbidity. Analyses were done using (1) data from the time of the previous linkage (2002) to March 2013; and (2) data from 1980 to March 2013. Results There was no evidence to suggest an excess risk of morbidity or mortality in either supplementation group compared with placebo for 2002–2013 and no associations were seen for the full time period (1980–2013). Conclusions Findings from this extended follow-up do not support our previous observation of an elevated risk of mortality from breast cancer or all cancers in later life among women who had taken 5 mg folic acid/day during pregnancy. Furthermore, there were no associations with risk of mortality from all-causes, all cancers or cardiovascular disease.
Collapse
Affiliation(s)
- Caroline M Taylor
- NIHR Biomedical Research Unit in Nutrition, Diet and Lifestyle, University of Bristol, Bristol, UK
| | - Charlotte Atkinson
- NIHR Biomedical Research Unit in Nutrition, Diet and Lifestyle, University of Bristol, Bristol, UK
| | - Chris Penfold
- NIHR Biomedical Research Unit in Nutrition, Diet and Lifestyle, University of Bristol, Bristol, UK
| | - Sohinee Bhattacharya
- Dugald Baird Centre for Research on Women's Health, University of Aberdeen, Aberdeen Maternity Hospital, Aberdeen, UK
| | - Doris Campbell
- Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Sam Leary
- NIHR Biomedical Research Unit in Nutrition, Diet and Lifestyle, University of Bristol, Bristol, UK
| | - Andy Ness
- NIHR Biomedical Research Unit in Nutrition, Diet and Lifestyle, University of Bristol, Bristol, UK
| |
Collapse
|
365
|
Shorter KR, Felder MR, Vrana PB. Consequences of dietary methyl donor supplements: Is more always better? PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2015; 118:14-20. [PMID: 25841986 DOI: 10.1016/j.pbiomolbio.2015.03.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 03/11/2015] [Accepted: 03/13/2015] [Indexed: 11/16/2022]
Abstract
Epigenetic mechanisms are now recognized to play roles in disease etiology. Several diseases increasing in frequency are associated with altered DNA methylation. DNA methylation is accomplished through metabolism of methyl donors such as folate, vitamin B12, methionine, betaine (trimethylglycine), and choline. Increased intake of these compounds correlates with decreased neural tube defects, although this mechanism is not well understood. Consumption of these methyl donor pathway components has increased in recent years due to fortification of grains and high supplemental levels of these compounds (e.g. vitamins, energy drinks). Additionally, people with mutations in one of the enzymes that assists in the methyl donor pathway (5-MTHFR) are directed to consume higher amounts of methyl donors to compensate. Recent evidence suggests that high levels of methyl donor intake may also have detrimental effects. Individualized medicine may be necessary to determine the appropriate amounts of methyl donors to be consumed, particularly in women of child bearing age.
Collapse
Affiliation(s)
- Kimberly R Shorter
- University of Florida School of Medicine, Department of Psychiatry at the McKnight Brain Institute, 1149 Newell Drive, Gainesville, FL 32611, USA
| | - Michael R Felder
- University of South Carolina, Department of Biological Sciences, 715 Sumter Street, Columbia, SC 29208, USA; Peromyscus Genetic Stock Center, University of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA
| | - Paul B Vrana
- University of South Carolina, Department of Biological Sciences, 715 Sumter Street, Columbia, SC 29208, USA; Peromyscus Genetic Stock Center, University of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA.
| |
Collapse
|
366
|
Martiniova L, Field MS, Finkelstein JL, Perry CA, Stover PJ. Maternal dietary uridine causes, and deoxyuridine prevents, neural tube closure defects in a mouse model of folate-responsive neural tube defects. Am J Clin Nutr 2015; 101:860-9. [PMID: 25833982 PMCID: PMC4381776 DOI: 10.3945/ajcn.114.097279] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 12/22/2014] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Folic acid prevents neural tube closure defects (NTDs), but the causal metabolic pathways have not been established. Serine hydroxymethyltransferase 1 (SHMT1) is an essential scaffold protein in folate-dependent de novo thymidylate synthesis in the nucleus. SHMT1-deficient mice provide a model to investigate folic acid-responsive NTDs wherein disruption of de novo thymidylate synthesis impairs neural tube closure. OBJECTIVE We examined the effects of maternal supplementation with the pyrimidine nucleosides uridine, thymidine, or deoxyuridine with and without folate deficiency on NTD incidence in the Shmt1 mouse model. DESIGN Shmt1(+/+) and Shmt1(-/-) female mice fed folate-replete or folate-deficient diets and supplemented with uridine, thymidine, or deoxyuridine were bred, and litters (n = 10-23 per group) were examined for the presence of NTDs. Biomarkers of impaired folate status and metabolism were measured, including plasma nucleosides, hepatic uracil content, maternal plasma folate concentrations, and incorporation of nucleoside precursors into DNA. RESULTS Shmt1(+/-) and Shmt1(-/-) embryos from dams fed the folate-deficient diet were susceptible to NTDs. No NTDs were observed in litters from dams fed the folate-deficient diet supplemented with deoxyuridine. Surprisingly, uridine supplementation increased NTD incidence, independent of embryo genotype and dietary folic acid. These dietary nucleosides did not affect maternal hepatic uracil accumulation in DNA but did affect plasma folate concentrations. CONCLUSIONS Maternal deoxyuridine supplementation prevented NTDs in dams fed the folate-deficient diet, whereas maternal uridine supplementation increased NTD incidence, independent of folate and embryo genotype. These findings provide new insights into the metabolic impairments and mechanisms of folate-responsive NTDs resulting from decreased Shmt1 expression.
Collapse
Affiliation(s)
- Lucia Martiniova
- From the Division of Nutritional Sciences, Cornell University, Ithaca, NY
| | - Martha S Field
- From the Division of Nutritional Sciences, Cornell University, Ithaca, NY
| | | | - Cheryll A Perry
- From the Division of Nutritional Sciences, Cornell University, Ithaca, NY
| | - Patrick J Stover
- From the Division of Nutritional Sciences, Cornell University, Ithaca, NY
| |
Collapse
|
367
|
Liu J, Zhang Y, Jin L, Li G, Wang L, Bao Y, Fu Y, Li Z, Zhang L, Ye R, Ren A. Variants in maternal COMT and MTHFR genes and risk of neural tube defects in offspring. Metab Brain Dis 2015; 30:507-13. [PMID: 24990354 DOI: 10.1007/s11011-014-9582-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 06/23/2014] [Indexed: 11/30/2022]
Abstract
Methylenetetrahydrofolate reductase (MTHFR) C677T and catechol-O-Methyltransferase (COMT) G158A are associated with a risk of neural tube defects (NTDs) in offspring. This study examined the effect of a MTHFR × COMT interaction on the risk of NTDs in a Chinese population with a high prevalence of NTDs. A total of 576 fetuses or newborns with NTDs and 594 controls were genotyped for MTHFRrs1801133, MTHFRrs1801131, and COMTrs4680 and COMTrs737865. Information on maternal sociodemographic characteristics, reproductive history, and related behavior was collected through face-to-face interviews. Possible interactions between genetic variants of MTHFR and COMT were examined. MTHFR C677T homozygous TT was associated with an elevated risk of total NTDs (odds ratio [OR] = 1.37, 95 % confidence interval [CI] = 0.93-2.03) and of anencephaly (OR = 1.67, 95 % CI = 0.98-2.84) compared with the CC genotype. There was a COMT rs737865 CC × MTHFR rs1801133 TT interaction for total NTDs (OR = 3.02, 95 % CI = 1.00-9.14) and for anencephaly (OR = 3.39, 95 % CI = 0.94-12.18). No interaction was found between COMT rs4680 AA/AG and MTHFR CT/TT genotypes for total NTDs or any subtype of NTD. The interaction of COMT rs737865 and MTHFR C677T was associated with an increased risk of NTDs, especially anencephaly, in a Chinese population with a high prevalence of NTDs.
Collapse
Affiliation(s)
- Jufen Liu
- Institute of Reproductive and Child Health/Ministry of Health Key Laboratory of Reproductive Health; Department of Epidemiology and Biostatistics, School of Public Health, Peking University, 38 College Rd, Haidian District, Beijing, 100191, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
368
|
Abstract
Neural tube defects (NTDs), including spina bifida and anencephaly, are severe birth defects of the central nervous system that originate during embryonic development when the neural tube fails to close completely. Human NTDs are multifactorial, with contributions from both genetic and environmental factors. The genetic basis is not yet well understood, but several nongenetic risk factors have been identified as have possibilities for prevention by maternal folic acid supplementation. Mechanisms underlying neural tube closure and NTDs may be informed by experimental models, which have revealed numerous genes whose abnormal function causes NTDs and have provided details of critical cellular and morphological events whose regulation is essential for closure. Such models also provide an opportunity to investigate potential risk factors and to develop novel preventive therapies.
Collapse
Affiliation(s)
- Nicholas D E Greene
- Newlife Birth Defects Research Center, Institute of Child Health, University College London, WC1N 1EH, United Kingdom;
| | | |
Collapse
|
369
|
Wang X, Guan Z, Chen Y, Dong Y, Niu Y, Wang J, Zhang T, Niu B. Genomic DNA hypomethylation is associated with neural tube defects induced by methotrexate inhibition of folate metabolism. PLoS One 2015; 10:e0121869. [PMID: 25822193 PMCID: PMC4379001 DOI: 10.1371/journal.pone.0121869] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 02/16/2015] [Indexed: 11/18/2022] Open
Abstract
DNA methylation is thought to be involved in the etiology of neural tube defects (NTDs). However, the exact mechanism between DNA methylation and NTDs remains unclear. Herein, we investigated the change of methylation in mouse model of NTDs associated with folate dysmetabolism by use of ultraperformance liquid chromatography tandem mass spectrometry (UPLC/MS/MS), liquid chromatography-electrospray ionization tandem mass spectrometry (LC-MS/MS), microarray, matrix-assisted laser desorption/ionization time-of-flight mass spectrometry and Real time quantitative PCR. Results showed that NTD neural tube tissues had lower concentrations of 5-methyltetrahydrofolate (5-MeTHF, P = 0.005), 5-formyltetrahydrofolate (5-FoTHF, P = 0.040), S-adenosylmethionine (SAM, P = 0.004) and higher concentrations of folic acid (P = 0.041), homocysteine (Hcy, P = 0.006) and S-adenosylhomocysteine (SAH, P = 0.045) compared to control. Methylation levels of genomic DNA decreased significantly in the embryonic neural tube tissue of NTD samples. 132 differentially methylated regions (35 low methylated regions and 97 high methylated regions) were selected by microarray. Two genes (Siah1b, Prkx) in Wnt signal pathway demonstrated lower methylated regions (peak) and higher expression in NTDs (P<0.05; P<0.05). Results suggest that DNA hypomethylation was one of the possible epigenetic variations correlated with the occurrence of NTDs induced by folate dysmetabolism and that Siah1b, Prkx in Wnt pathway may be candidate genes for NTDs.
Collapse
Affiliation(s)
- Xiuwei Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Zhen Guan
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Yan Chen
- Department of Respiratory, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yanting Dong
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Yuhu Niu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Jianhua Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Ting Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
- * E-mail: (BN); (TZ)
| | - Bo Niu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
- * E-mail: (BN); (TZ)
| |
Collapse
|
370
|
Yang Y, Chen J, Wang B, Ding C, Liu H. Association between MTHFR C677T polymorphism and neural tube defect risks: A comprehensive evaluation in three groups of NTD patients, mothers, and fathers. ACTA ACUST UNITED AC 2015; 103:488-500. [PMID: 25808073 DOI: 10.1002/bdra.23361] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND The C677T polymorphism in the methylenetetrahydrofolate reductase gene (MTHFR) gene has been reported to play a critical role in the pathogenesis of neural tube defects (NTDs). The association of the C677T polymorphism in the MTHFR gene and NTD susceptibility has been widely demonstrated, but the results are inconclusive. In this study, we performed a meta-analysis in three groups to investigate the association between the MTHFR C677T polymorphism and NTD risk. METHODS A computer retrieval of PubMed, Cochrane Library, CBM, and Embase for papers on the MTHFR C677T polymorphism and NTD risk was performed. All data were analyzed with STATA (Version 13.0). Odds ratios (ORs) with 95% confidence intervals (CIs) were estimated to assess the association. A test for heterogeneity, a sensitivity analysis, and an assessment of publication bias were performed in our meta-analysis. RESULTS Forty articles were included in this meta-analysis: 13 studies for Group A: 1329 NTD patients versus 2965 healthy controls; 34 studies for Group B: 3018 mothers with NTD progeny versus 8746 healthy controls; three studies for Group C: 157 fathers with NTD progeny versus 705 healthy controls. The analysis results show: allele contrast in NTD patients: OR = 1.445, 95% CI [1.186, 1.760]; allele contrast in mothers: OR = 1.342, 95% CI [1.166, 1.544]; allele contrast in fathers: OR = 1.062, 95% CI [0.821, 1.374]. CONCLUSION We found no association between any of the fathers' genotypes and NTDs, whereas a significant correlation between MTHFR C677T polymorphism and NTD risk was found in NTD patients and in their mother.
Collapse
Affiliation(s)
- Yi Yang
- West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jie Chen
- West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Beiyu Wang
- West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Chen Ding
- West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hao Liu
- West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
371
|
Tinker SC, Gilboa S, Reefhuis J, Jenkins MM, Schaeffer M, Moore CA. Challenges in Studying Modifiable Risk Factors for Birth Defects. CURR EPIDEMIOL REP 2015; 2:23-30. [PMID: 26236577 PMCID: PMC4516719 DOI: 10.1007/s40471-014-0028-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Conducting research to identify modifiable risk factors for birth defects is difficult for a variety of reasons. While some challenges are familiar to researchers across many disciplines, the confluence of issues affecting birth defects research may not be well understood by those outside of the field. This article describes several methodological challenges to the study of birth defects and ways these challenges might be addressed: (1) ascertainment, definition and classification of birth defects; (2) exposure assessment on modifiable risk factors; (3) analytical challenges related to small numbers and multiple statistical tests; (4) the role of genetics, including the collection of specimens and analysis of genetic data; and (5) challenges in translating research and demonstrating public health impact. Understanding these issues is important for researchers planning studies, reviewers evaluating the scientific merit of results from these studies, and consumers of the research, including fellow researchers, policy makers, health care providers, and families.
Collapse
Affiliation(s)
- Sarah C. Tinker
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, 1600 Clifton Road NE, MS E86, Atlanta, GA 30333, Phone: 404-498-3509, Fax: 404-498-3040
| | - Suzanne Gilboa
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, 1600 Clifton Road NE, MS E86, Atlanta, GA 30333, Phone: 404-498-4425, Fax: 404-498-3040
| | - Jennita Reefhuis
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, 1600 Clifton Road NE, MS E86, Atlanta, GA 30333, Phone: 404-498-3917, Fax: 404-498-3040
| | - Mary M. Jenkins
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, 1600 Clifton Road NE, MS E86, Atlanta, GA 30333, Phone: 404-498-3889, Fax: 404-498-3550
| | - Marcy Schaeffer
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, 1600 Clifton Road NE, MS E86, Atlanta, GA 30333, Phone: 404-498-0265, Fax: 404-498-3040
| | - Cynthia A. Moore
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, 1600 Clifton Road NE, MS E86, Atlanta, GA 30333, Phone: 404-498-3927, Fax: 404-498-3040
| |
Collapse
|
372
|
Liu Y, Green TJ, Kitts DD. Stability of microencapsulated L-5-methyltetrahydrofolate in fortified noodles. Food Chem 2015; 171:206-11. [DOI: 10.1016/j.foodchem.2014.08.129] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 06/13/2014] [Accepted: 08/29/2014] [Indexed: 11/28/2022]
|
373
|
Hao N, Xia W, Tang Y, Wu M, Jiang H, Lin X, Liu J, Zhou D. Periconceptional folic acid supplementation among pregnant women with epilepsy in a developing country: a retroprospective survey in China. Epilepsy Behav 2015; 44:27-34. [PMID: 25618500 DOI: 10.1016/j.yebeh.2014.12.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 12/19/2014] [Accepted: 12/21/2014] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The aims of this study were to investigate the implementation of guidelines on periconceptional folic acid supplementation among pregnant women with epilepsy (WWE) in China and to identify its potential correlations with selected sociodemographic and clinical factors. METHODS A detailed investigation was conducted in China using a structured questionnaire from December 2013 to May 2014. Data on the awareness and use of folic acid supplementation, as well as sociodemographic and clinical characteristics, were collected from 153 pregnant WWE. Descriptive analysis, followed by univariate and multivariate logistic regression analyses, was applied to the data from this survey. RESULTS Among the enrolled subjects, 67.3% became pregnant after the promulgation of the relevant guidelines. Only 26.2% of them knew the exact effects of folic acid, and 73.8% had taken folic acid at some point during periconception. In addition, the folic acid intake of most of these women (67.1%) did not exceed that of the average pregnant woman. The prevalence of folic acid supplementation for pregnant WWE three months before pregnancy was only 15.5%. There has been almost no improvement in the level of additional awareness and use of folic acid supplementation for WWE since the relevant guidelines were established in China. Pregnant WWE with higher education levels, those with a planned pregnancy, or those who live in urban areas were more likely to know about and implement folic acid supplementation during periconception. Moreover, pregnant WWE with a planned pregnancy or those living in cities seemed to have a higher folic acid intake and started folic acid supplementation earlier before conception. CONCLUSION The extent of awareness and use of folic acid supplementation in pregnant WWE remains low in China. More efforts are needed to promote periconceptional folic acid supplementation for WWE, especially those with low education levels and those who live in rural areas. Planned pregnancy should be encouraged for WWE.
Collapse
Affiliation(s)
- Nanya Hao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Xia
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yingying Tang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mengqian Wu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Han Jiang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xu Lin
- Department of Neurology, 363 Hospital, Chengdu 610041, China
| | - Jie Liu
- Department of Neurology, Sichuan People's Hospital, Chengdu 610072, China
| | - Dong Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
374
|
Steel A, Lucke J, Adams J. The prevalence and nature of the use of preconception services by women with chronic health conditions: an integrative review. BMC WOMENS HEALTH 2015; 15:14. [PMID: 25783639 PMCID: PMC4338627 DOI: 10.1186/s12905-015-0165-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 01/20/2015] [Indexed: 12/28/2022]
Abstract
Background There is growing evidence that preconception care may have an important role in preventing short and long term adverse health consequences for women and their offspring. This is particularly the case for women with chronic health conditions due to the rising prevalence of chronic disease in global populations. With this in mind, this paper presents an integrative systematic review of contemporary research outlining the use of preconception services and practices by women with chronic health conditions. Methods A search was conducted through PubMed, CINAHL, AMED, and Maternity and Infant Care databases which identified 672 papers examining preconception care and preconception services for women with chronic health conditions. Fourteen papers which were written in English, presented original research, and reported on the prevalence or nature of use of preconception care by women with chronic health conditions were included in the review. Critical appraisal of study quality and thematic categorical grouping of identified papers was undertaken. Results Current research evidence, as identified through this review, examines three major topic areas: the prevalence of preconception care practices, use of services and characteristics of users; knowledge of the value and impact of preconception care and availability of preconception services for women with chronic health conditions; and women’s attitudes, approaches and experiences of preconception care and preconception services. Prevalence estimates of engagement with preconception care range between 18.1% and 45%, with most studies focusing on women with type 1 or 2 diabetes. Significant gaps in women’s knowledge of preconception care for women with chronic health conditions were also identified. Women with chronic health conditions reported experiencing emotional distress as a result of their engagement with preconception care services. They also commonly described feeling a need to employ discipline to comply with preconception care programs, and experiencing a fear of pregnancy complications. Conclusion Future research requires a broad and sophisticated approach to research design and analysis, improved consideration of temporal changes to women’s health behaviour, representative samples to more effectively inform health policy, and a deeper understanding of women’s motivations, attitudes and perceptions of preconception care to assist in the development of tailored preconception health services.
Collapse
Affiliation(s)
- Amie Steel
- Office of Research, Endeavour College of Natural Health, Brisbane, Queensland, Australia,
| | | | | |
Collapse
|
375
|
Yang B, Fan S, Zhi X, Wang Y, Wang Y, Zheng Q, Sun G. Prevalence of hyperhomocysteinemia in China: a systematic review and meta-analysis. Nutrients 2014; 7:74-90. [PMID: 25551247 PMCID: PMC4303827 DOI: 10.3390/nu7010074] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/12/2014] [Indexed: 12/31/2022] Open
Abstract
Hyperhomocysteinemia (HHcy, total homocysteine concentrations > 15 μmol/L) has been associated with increased risk of many diseases. A systematic review was performed to summarize the prevalence of HHcy in China. We searched multiple international and Chinese scientific databases for relevant literature, and further manually screened reference lists and corresponded with original authors. Pooled prevalence of HHcy was calculated using random effects model. Subgroup analysis, meta-regression and sensitivity analysis were also performed. A total of 36 studies consisting 60,754 subjects (57.3% male; age range, 3–97 years) were finally included. The overall pooled prevalence of HHcy was 27.5%. Geographically, the prevalence was high in north areas, intermediate in central areas, and low in south areas, and was higher in inland versus coastal areas. The prevalence increased with age and was significantly higher in men than in women. Rural residents had a slightly higher HHcy prevalence than urban residents, and the studies conducted during 2006 to 2012 presented a higher HHcy prevalence than those during 1990 to 2005. In summary, the prevalence of HHcy in China is high, particularly in northern populations, the inlanders, males, and the elderly. Homocysteine-lowering strategies are necessary to reduce this highly preventable disorder.
Collapse
Affiliation(s)
- Boyi Yang
- Research Center of Environment and Non-Communicable Disease, School of Public Health, China Medical University, Shenyang 110013, China.
| | - Shujun Fan
- Research Center of Environment and Non-Communicable Disease, School of Public Health, China Medical University, Shenyang 110013, China.
| | - Xueyuan Zhi
- Research Center of Environment and Non-Communicable Disease, School of Public Health, China Medical University, Shenyang 110013, China.
| | - Yinuo Wang
- Division of Molecular Preventive Medicine, Shanghai Institute of Targeted Therapy and Molecular Medicine, Shanghai 200433, China.
| | - Yanxun Wang
- Division of Molecular Preventive Medicine, Shanghai Institute of Targeted Therapy and Molecular Medicine, Shanghai 200433, China.
| | - Quanmei Zheng
- Research Center of Environment and Non-Communicable Disease, School of Public Health, China Medical University, Shenyang 110013, China.
| | - Guifan Sun
- Research Center of Environment and Non-Communicable Disease, School of Public Health, China Medical University, Shenyang 110013, China.
| |
Collapse
|
376
|
Montalvão-de-Azevedo R, Vasconcelos GM, Vargas FR, Thuler LC, Pombo-de-Oliveira MS, de Camargo B. RFC-1 80G>A polymorphism in case-mother/control-mother dyads is associated with risk of nephroblastoma and neuroblastoma. Genet Test Mol Biomarkers 2014; 19:75-81. [PMID: 25536437 DOI: 10.1089/gtmb.2014.0177] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIM Embryonic tumors are associated with an interruption during normal organ development; they may be related to disturbances in the folate pathway involved in DNA synthesis, methylation, and repair. Prenatal supplementation with folic acid is associated with a decreased risk of neuroblastoma, brain tumors, retinoblastoma, and nephroblastoma. The aim of this study was to investigate the association between MTHFR rs1801133 (C677T) and RFC-1 rs1051266 (G80A) genotypes with the risk of developing nephroblastoma and neuroblastoma. MATERIALS AND METHODS Case-mother/control-mother dyad study. Samples from Brazilian children with nephroblastoma (n=80), neuroblastoma (n=66), healthy controls (n=453), and their mothers (case n=93; control n=75) were analyzed. Genomic DNA was isolated from peripheral blood cells and/or buccal cells and genotyped to identify MTHFR C677T and RFC-1 G80A polymorphisms. Differences in genotype distribution between patients and controls were tested by multiple logistic regression analysis. RESULTS Risk for nephroblastoma and neuroblastoma was two- to fourfold increased among children with RFC-1 polymorphisms. An increased four- to eightfold risk for neuroblastoma and nephroblastoma was seen when the child and maternal genotypes were combined. CONCLUSION Our results suggest that mother and child RFC-1 G80A genotypes play a role on the risk of neuroblastoma and nephroblastoma since this polymorphism may impair the intracellular levels of folate, through carrying fewer folate molecules to the cell interior, and thus, the intracellular concentration is not enough to maintain regular DNA synthesis and methylation pathways.
Collapse
|
377
|
Guan Z, Wang X, Dong Y, Xu L, Zhu Z, Wang J, Zhang T, Niu B. dNTP deficiency induced by HU via inhibiting ribonucleotide reductase affects neural tube development. Toxicology 2014; 328:142-51. [PMID: 25527867 DOI: 10.1016/j.tox.2014.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/12/2014] [Accepted: 12/01/2014] [Indexed: 12/22/2022]
Abstract
Exposure to environmental toxic chemicals in utero during the neural tube development period can cause developmental disorders. To evaluate the disruption of neural tube development programming, the murine neural tube defects (NTDs) model was induced by interrupting folate metabolism using methotrexate in our previous study. The present study aimed to examine the effects of dNTP deficiency induced by hydroxyurea (HU), a specific ribonucleotide reductase (RNR) inhibitor, during murine neural tube development. Pregnant C57BL/6J mice were intraperitoneally injected with various doses of HU on gestation day (GD) 7.5, and the embryos were checked on GD 11.5. RNR activity and deoxynucleoside triphosphate (dNTP) levels were measured in the optimal dose. Additionally, DNA damage was examined by comet analysis and terminal deoxynucleotidyl transferase mediated dUTP nick end-labeling (TUNEL) assay. Cellular behaviors in NTDs embryos were evaluated with phosphorylation of histone H3 (PH-3) and caspase-3 using immunohistochemistry and western blot analysis. The results showed that NTDs were observed mostly with HU treatment at an optimal dose of 225 mg/kg b/w. RNR activity was inhibited and dNTP levels were decreased in HU-treated embryos with NTDs. Additionally, increased DNA damage, decreased proliferation, and increased caspase-3 were significant in NTDs embryos compared to the controls. Results indicated that HU induced murine NTDs model by disturbing dNTP metabolism and further led to the abnormal cell balance between proliferation and apoptosis.
Collapse
Affiliation(s)
- Zhen Guan
- Department of Biotechnology, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Xiuwei Wang
- Department of Biotechnology, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Yanting Dong
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Lin Xu
- Department of Biotechnology, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Zhiqiang Zhu
- Department of Biotechnology, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Jianhua Wang
- Department of Biotechnology, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Ting Zhang
- Department of Biotechnology, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China.
| | - Bo Niu
- Department of Biotechnology, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China.
| |
Collapse
|
378
|
|
379
|
Zheng J, Lu X, Liu H, Zhao P, Li K, Li L. MTHFD1 polymorphism as maternal risk for neural tube defects: a meta-analysis. Neurol Sci 2014; 36:607-16. [PMID: 25502174 DOI: 10.1007/s10072-014-2035-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 12/06/2014] [Indexed: 11/27/2022]
Abstract
Recently, the association between methylenetetrahydrofolate dehydrogenase 1 (MTHFD1) G1958A polymorphism and neural tube defects (NTD) susceptibility has been widely investigated; however, the results remained inconclusive. Hence, we conducted a meta-analysis to evaluate the effect of MTHFD1 G1958A polymorphism on NTD. The relative literatures were identified by search of the electronic databases PubMed, MEDLINE, and EMBASE. The extracted data were statistically analyzed, and pooled odds ratios (ORs) with 95 % confidence intervals (CIs) were calculated to estimate the association strength using Stata version 11.0 software. Finally, ten studies met our inclusion criteria, including 2,132/4,082 in NTD infants and controls; 1,402/3,136 in mothers with NTD offspring and controls; and 993/2,879 in fathers with NTD offspring and controls. This meta-analysis showed that, compared with the mothers with GG genotype, the women with AA genotype had an increased risk of NTD in their offspring, with OR values and 95 % CI at 1.39 (1.16-1.68), p < 0.001. Interestingly, fathers with AG genotype had a significant decreased risk of NTD offspring (OR = 0.79, 95 % CI = 0.66-0.94, p = 0.009). However, there was no significant association between the MTHFD1 G1958A polymorphism in NTD patients and the risk of NTD. In conclusion, the present meta-analysis provided evidence of the association between maternal MTHFD1 G1958A polymorphism and NTD susceptibility.
Collapse
Affiliation(s)
- Jinyu Zheng
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | | | | | | | | | | |
Collapse
|
380
|
Lyu Y, Mirea L, Yang J, Warre R, Zhang J, Lee SK, Li Z. Secular trends in age at menarche among women born between 1955 and 1985 in Southeastern China. BMC WOMENS HEALTH 2014; 14:155. [PMID: 25495097 PMCID: PMC4275952 DOI: 10.1186/s12905-014-0155-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/19/2014] [Indexed: 11/12/2022]
Abstract
Background Improvements in socioeconomic conditions and population health have been linked to declining age at menarche. In China, secular trends in age at menarche following extensive economic reform during recent decades have not been thoroughly investigated. This study examined the overall trend in age at menarche and assessed differences in the rate of change of age at menarche over time, and between urban and rural populations and education levels in southeastern China. Methods Age at menarche was retrospectively collected from 1,167,119 Han Chinese women born 1955–1985, who registered in the Perinatal Health Care Surveillance System in 19 cities and counties in two southeast provinces during 1993–2005. Multivariable linear regression was used to estimate trends in age at menarche overall and stratified by urban/rural residence and education level. Results Age at menarche declined by 0.33 [95% CI 0.33, 0.32] years/decade overall, with the fastest decline in women born in 1966–1975. For the earliest birth cohorts (1955–1965), age at menarche declined faster in urban versus rural regions, and for women with high school education or above versus primary school or less. In contrast, age at menarche declined slower among urban women born 1976–1985, and among those with higher education born 1966–1985. Conclusions Mean age at menarche declined for women born in 1955–1985 in southeast China. Further study is warranted to identify specific factors contributing to earlier age at menarche and associated health outcomes. Electronic supplementary material The online version of this article (doi:10.1186/s12905-014-0155-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yanyu Lyu
- Department of Child Health Development, Capital Institute of Pediatrics, Beijing, China. .,School of Public Health, Peking University Health Science Center, Beijing, China. .,Maternal-Infant Care Research Centre, Mount Sinai Hospital, Toronto, Ontario, Canada.
| | - Lucia Mirea
- Maternal-Infant Care Research Centre, Mount Sinai Hospital, Toronto, Ontario, Canada. .,Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada.
| | - Junmin Yang
- Maternal-Infant Care Research Centre, Mount Sinai Hospital, Toronto, Ontario, Canada.
| | - Ruth Warre
- Maternal-Infant Care Research Centre, Mount Sinai Hospital, Toronto, Ontario, Canada.
| | - Jun Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Shoo K Lee
- Maternal-Infant Care Research Centre, Mount Sinai Hospital, Toronto, Ontario, Canada. .,Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada.
| | - Zhu Li
- School of Public Health, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
381
|
Choi JH, Yates Z, Veysey M, Heo YR, Lucock M. Contemporary issues surrounding folic Acid fortification initiatives. Prev Nutr Food Sci 2014; 19:247-60. [PMID: 25580388 PMCID: PMC4287316 DOI: 10.3746/pnf.2014.19.4.247] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 12/07/2014] [Indexed: 12/20/2022] Open
Abstract
The impact of folate on health and disease, particularly pregnancy complications and congenital malformations, has been extensively studied. Mandatory folic acid fortification therefore has been implemented in multiple countries, resulting in a reduction in the occurrence of neural tube defects. However, emerging evidence suggests increased folate intake may also be associated with unexpected adverse effects. This literature review focuses on contemporary issues of concern, and possible underlying mechanisms as well as giving consideration the future direction of mandatory folic acid fortification. Folate fortification has been associated with the presence of unmetabolized folic acid (PteGlu) in blood, masking of vitamin B12 deficiency, increased dosage for anti-cancer medication, photo-catalysis of PteGlu leading to potential genotoxicity, and a role in the pathoaetiology of colorectal cancer. Increased folate intake has also been associated with twin birth and insulin resistance in offspring, and altered epigenetic mechanisms of inheritance. Although limited data exists to elucidate potential mechanisms underlying these issues, elevated blood folate level due to the excess use of PteGlu without consideration of an individual's specific phenotypic traits (e.g. genetic background and undiagnosed disease) may be relevant. Additionally, the accumulation of unmetabolized PteGlu may lead to inhibition of dihydrofolate reductase and other enzymes. Concerns notwithstanding, folic acid fortification has achieved enormous advances in public health. It therefore seems prudent to target and carefully monitor high risk groups, and to conduct well focused further research to better understand and to minimize any risk of mandatory folic acid fortification.
Collapse
Affiliation(s)
- Jeong-Hwa Choi
- School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW 2258, Australia
- Human Economics Research Institute, Chonnam National University, Gwangju 500-757, Korea
| | - Zoe Yates
- Biomedical Sciences and Pharmacy, University of Newcastle, Ourimbah, NSW 2258, Australia
| | - Martin Veysey
- Teaching and Research Unit, Central Coast Local Health District, Gosford, NSW 2250, Australia
| | - Young-Ran Heo
- Department of Food and Nutrition, Chonnam National University, Gwangju 500-757, Korea
| | - Mark Lucock
- School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW 2258, Australia
| |
Collapse
|
382
|
Ouyang F, Longnecker MP, Venners SA, Johnson S, Korrick S, Zhang J, Xu X, Christian P, Wang MC, Wang X. Preconception serum 1,1,1-trichloro-2,2,bis(p-chlorophenyl)ethane and B-vitamin status: independent and joint effects on women's reproductive outcomes. Am J Clin Nutr 2014; 100:1470-8. [PMID: 25411282 PMCID: PMC4232015 DOI: 10.3945/ajcn.114.088377] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Although preconception 1,1,1-trichloro-2,2,bis(p-chlorophenyl)ethane (DDT) exposure and B-vitamin deficiencies have each been shown to negatively affect human reproductive outcomes, little is known about their joint effect. OBJECTIVE We sought to examine whether B-vitamin sufficiency protects against adverse effects of DDT on clinical pregnancy (CP) and subclinical early pregnancy loss (EPL). DESIGN We measured preconception concentrations of plasma B vitamins (vitamin B-6, vitamin B-12, and folate) and serum total DDT [sum of p,p' and o,p' isomers of DDT and 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene] in 291 nulligravid women from Anhui, China, who were studied in 1996-1998. The women were followed prospectively from the time they stopped contraception until CP (gestational age ≥42 d) or 12 mo (whichever occurred first). EPL was identified by using daily urinary human chorionic gonadotropin. The women were categorized according to B-vitamin status (deficiency compared with sufficiency) and DDT concentration (high compared with low). RESULTS Of 291 study women, a total of 385 conceptions (31% of which ended in EPL) and 265 CPs occurred. Compared with women with adequate B-vitamins and low DDT, incidence rates of CP were reduced in women with B-vitamin deficiency and a high DDT concentration (P < 0.05 for all). Most notably, in women with sufficient vitamin B-12, DDT was not associated with the incidence of CP; in contrast, in women with vitamin B-12 deficiency, high DDT was associated with a lower incidence of CP (HR: 0.44; 95% CI: 0.23, 0.84); and the test for interaction was significant (P < 0.05). The odds of EPL decreased by 45% (95% CI: 21%, 62%) for each interquartile distance increase in folate in women with high DDT concentrations, and the test for interaction was significant (P = 0.006). CONCLUSIONS Our results provide suggestive evidence that vitamin B-12 and folate sufficiency may help protect against adverse reproductive effects of DDT exposure. Additional studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Fengxiu Ouyang
- From the Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (FO); the Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Department of Health and Human Services, Research Triangle Park, NC (MPL); the Simon Fraser University, Faculty of Health Sciences, Burnaby, Canada (SAV); the Division of General Pediatrics & Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD (SJ and XW); the Department of Environmental Health, Harvard School of Public Health, Boston, MA (SK); the Shanghai Jiaotong University School of Public Health, Shanghai, China (JZ); the National Clinical Research Center of Kidney Disease; State Key Laboratory of Organ Failure Research; Nanfang Hospital, Southern Medical University, Guangzhou, China (XX); and the Departments of International Health (PC) and Biostatistics (M-CW) and Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health (XW), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Matthew P Longnecker
- From the Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (FO); the Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Department of Health and Human Services, Research Triangle Park, NC (MPL); the Simon Fraser University, Faculty of Health Sciences, Burnaby, Canada (SAV); the Division of General Pediatrics & Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD (SJ and XW); the Department of Environmental Health, Harvard School of Public Health, Boston, MA (SK); the Shanghai Jiaotong University School of Public Health, Shanghai, China (JZ); the National Clinical Research Center of Kidney Disease; State Key Laboratory of Organ Failure Research; Nanfang Hospital, Southern Medical University, Guangzhou, China (XX); and the Departments of International Health (PC) and Biostatistics (M-CW) and Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health (XW), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Scott A Venners
- From the Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (FO); the Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Department of Health and Human Services, Research Triangle Park, NC (MPL); the Simon Fraser University, Faculty of Health Sciences, Burnaby, Canada (SAV); the Division of General Pediatrics & Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD (SJ and XW); the Department of Environmental Health, Harvard School of Public Health, Boston, MA (SK); the Shanghai Jiaotong University School of Public Health, Shanghai, China (JZ); the National Clinical Research Center of Kidney Disease; State Key Laboratory of Organ Failure Research; Nanfang Hospital, Southern Medical University, Guangzhou, China (XX); and the Departments of International Health (PC) and Biostatistics (M-CW) and Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health (XW), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Sara Johnson
- From the Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (FO); the Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Department of Health and Human Services, Research Triangle Park, NC (MPL); the Simon Fraser University, Faculty of Health Sciences, Burnaby, Canada (SAV); the Division of General Pediatrics & Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD (SJ and XW); the Department of Environmental Health, Harvard School of Public Health, Boston, MA (SK); the Shanghai Jiaotong University School of Public Health, Shanghai, China (JZ); the National Clinical Research Center of Kidney Disease; State Key Laboratory of Organ Failure Research; Nanfang Hospital, Southern Medical University, Guangzhou, China (XX); and the Departments of International Health (PC) and Biostatistics (M-CW) and Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health (XW), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Susan Korrick
- From the Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (FO); the Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Department of Health and Human Services, Research Triangle Park, NC (MPL); the Simon Fraser University, Faculty of Health Sciences, Burnaby, Canada (SAV); the Division of General Pediatrics & Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD (SJ and XW); the Department of Environmental Health, Harvard School of Public Health, Boston, MA (SK); the Shanghai Jiaotong University School of Public Health, Shanghai, China (JZ); the National Clinical Research Center of Kidney Disease; State Key Laboratory of Organ Failure Research; Nanfang Hospital, Southern Medical University, Guangzhou, China (XX); and the Departments of International Health (PC) and Biostatistics (M-CW) and Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health (XW), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Jun Zhang
- From the Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (FO); the Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Department of Health and Human Services, Research Triangle Park, NC (MPL); the Simon Fraser University, Faculty of Health Sciences, Burnaby, Canada (SAV); the Division of General Pediatrics & Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD (SJ and XW); the Department of Environmental Health, Harvard School of Public Health, Boston, MA (SK); the Shanghai Jiaotong University School of Public Health, Shanghai, China (JZ); the National Clinical Research Center of Kidney Disease; State Key Laboratory of Organ Failure Research; Nanfang Hospital, Southern Medical University, Guangzhou, China (XX); and the Departments of International Health (PC) and Biostatistics (M-CW) and Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health (XW), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Xiping Xu
- From the Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (FO); the Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Department of Health and Human Services, Research Triangle Park, NC (MPL); the Simon Fraser University, Faculty of Health Sciences, Burnaby, Canada (SAV); the Division of General Pediatrics & Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD (SJ and XW); the Department of Environmental Health, Harvard School of Public Health, Boston, MA (SK); the Shanghai Jiaotong University School of Public Health, Shanghai, China (JZ); the National Clinical Research Center of Kidney Disease; State Key Laboratory of Organ Failure Research; Nanfang Hospital, Southern Medical University, Guangzhou, China (XX); and the Departments of International Health (PC) and Biostatistics (M-CW) and Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health (XW), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Parul Christian
- From the Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (FO); the Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Department of Health and Human Services, Research Triangle Park, NC (MPL); the Simon Fraser University, Faculty of Health Sciences, Burnaby, Canada (SAV); the Division of General Pediatrics & Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD (SJ and XW); the Department of Environmental Health, Harvard School of Public Health, Boston, MA (SK); the Shanghai Jiaotong University School of Public Health, Shanghai, China (JZ); the National Clinical Research Center of Kidney Disease; State Key Laboratory of Organ Failure Research; Nanfang Hospital, Southern Medical University, Guangzhou, China (XX); and the Departments of International Health (PC) and Biostatistics (M-CW) and Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health (XW), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Mei-Cheng Wang
- From the Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (FO); the Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Department of Health and Human Services, Research Triangle Park, NC (MPL); the Simon Fraser University, Faculty of Health Sciences, Burnaby, Canada (SAV); the Division of General Pediatrics & Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD (SJ and XW); the Department of Environmental Health, Harvard School of Public Health, Boston, MA (SK); the Shanghai Jiaotong University School of Public Health, Shanghai, China (JZ); the National Clinical Research Center of Kidney Disease; State Key Laboratory of Organ Failure Research; Nanfang Hospital, Southern Medical University, Guangzhou, China (XX); and the Departments of International Health (PC) and Biostatistics (M-CW) and Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health (XW), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Xiaobin Wang
- From the Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (FO); the Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Department of Health and Human Services, Research Triangle Park, NC (MPL); the Simon Fraser University, Faculty of Health Sciences, Burnaby, Canada (SAV); the Division of General Pediatrics & Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD (SJ and XW); the Department of Environmental Health, Harvard School of Public Health, Boston, MA (SK); the Shanghai Jiaotong University School of Public Health, Shanghai, China (JZ); the National Clinical Research Center of Kidney Disease; State Key Laboratory of Organ Failure Research; Nanfang Hospital, Southern Medical University, Guangzhou, China (XX); and the Departments of International Health (PC) and Biostatistics (M-CW) and Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health (XW), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
383
|
Gallot V, Nedellec S, Capmas P, Legendre G, Lejeune-Saada V, Subtil D, Nizard J, Levêque J, Deffieux X, Hervé B, Vialard F. Fausses couches précoces « à répétition » : bilan et prise en charge. ACTA ACUST UNITED AC 2014; 43:812-41. [DOI: 10.1016/j.jgyn.2014.09.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
384
|
|
385
|
Dang S, Yan H, Zeng L, Wang Q, Li Q, Xiao S, Fan X. The status of vitamin B12 and folate among Chinese women: a population-based cross-sectional study in northwest China. PLoS One 2014; 9:e112586. [PMID: 25390898 PMCID: PMC4229226 DOI: 10.1371/journal.pone.0112586] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 10/08/2014] [Indexed: 01/08/2023] Open
Abstract
Objective To assess the status of the vitamin B12 and folate of Chinese women living in northwest China. Methods A population-based cross-sectional study was conducted in 2008 among Chinese women aged 10–49 years living in Shaanxi province of northwest China. A stratified multistage random sampling method was adopted to obtain a sample of 1170 women. The women were interviewed for collection of their background information and their plasma vitamin B12 and folate were measured with the immunoassay method. The status of both vitamins was evaluated and the prevalence of deficiency was estimated. Results The median value of the women was 214.5 pg/mL for vitamin B12 and 4.6 ng/mL for folate. The urban women had a significantly higher vitamin B12 (254.1 vs. 195.9 pg/mL) but lower folate (4.4 vs. 4.7 ng/mL) than rural women. Total prevalence of deficiency was 45.5% (95% CI: 42.6%∼48.4%) for vitamin B12 and 14.7% (95% CI: 12.6%∼16.8%) for folate. About 36% of women presented vitamin B12 deficiency alone, 5.2% belonged to folate deficiency alone and 9.5% was combined deficiency in both vitamins. More than 25% of the women were in marginal vitamin B12 status (200–299 pg/mL) and 60% in marginal status of folate (3–6 ng/mL). About 75.2% of rural women with folate deficiency were deficient in vitamin B12 and 46% for urban women. Quantile regression model found decreasing coefficient of folate status across 73 different quantiles of vitamin B12, which indicated that the women with folate deficiency had lower vitamin B12 significantly compared with those with no deficiency. Conclusions The deficiency of vitamin B12 and folate is still prevalent among the Chinese women in northwest China. Vitamin B12 deficiency could be more serious and the improvement of poor vitamin B12 status should be invoked when practicing the supplementation of folate against the neural tube defects in northwest China.
Collapse
Affiliation(s)
- Shaonong Dang
- Department of Epidemiology and Health Statistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Hong Yan
- Department of Epidemiology and Health Statistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
- * E-mail:
| | - Lingxia Zeng
- Department of Epidemiology and Health Statistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Quanli Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Qiang Li
- Department of Epidemiology and Health Statistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Shengbin Xiao
- Department of Epidemiology and Health Statistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Xiaojing Fan
- Department of Epidemiology and Health Statistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
386
|
Neggers Y. The Relationship between Folic Acid and Risk of Autism Spectrum Disorders. Healthcare (Basel) 2014; 2:429-44. [PMID: 27429286 PMCID: PMC4934568 DOI: 10.3390/healthcare2040429] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 09/18/2014] [Accepted: 10/13/2014] [Indexed: 11/17/2022] Open
Abstract
There is considerable scientific evidence that many aspects of diet influence the occurrence of human disease. Many factors such as genetic, psychological, environmental and behavioral characteristics influence development of human disease, and there is a close relationship between nutrition and disease. Though typical Western diets are not overtly deficient in essential nutrients, nutriture of a few micro nutrients such as folic acid has been reported to be sub-optimal, particularly in women of childbearing age. The role of folic acid in the prevention of macrocytic anemia and neural tube defects is well established. However, the relationship between folic acid and risk of autism is still evolving. Furthermore, environmental as well as nutritional factors such as folic acid are now well acknowledged as interacting with the individual genetic background in development of several diseases. In this article, recent research regarding the relationship between folic acid and risk of autism is evaluated.
Collapse
Affiliation(s)
- Yasmin Neggers
- Department of Human Nutrition, University of Alabama, Box 870311, 504 University Blvd, Tuscaloosa, AL 35487, USA.
| |
Collapse
|
387
|
Huang C, Guo C, Nichols C, Chen S, Martorell R. Elevated levels of protein in urine in adulthood after exposure to the Chinese famine of 1959-61 during gestation and the early postnatal period. Int J Epidemiol 2014; 43:1806-14. [PMID: 25298393 DOI: 10.1093/ije/dyu193] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Animal models have suggested that undernutrition during gestation and the early postnatal period may adversely affect kidney development and compromise renal function. As a natural experiment, famines provide an opportunity to test such potential effects in humans. We assessed whether exposure to the Chinese famine of 1959-1961 during gestation and early postnatal life was associated with the levels of proteinuria among female adults three decades after exposure to the famine. METHODS We measured famine intensity using the cohort size shrinkage index and we constructed a difference-in-difference model to compare the levels of proteinuria, measured with a dipstick test of random urine specimens, among Chinese women (n = 70 543) whose exposure status to the famine varied across birth cohorts (born before, during or after the famine) and counties of residence with different degrees of famine intensity. RESULTS Famine exposure was associated with a greater risk [odds ratio (OR) = 1.54; 95% confidence interval (CI): 1.04, 2.28; P = 0.029) of having higher level of proteinuria among women born during the famine years (1959-61) compared with the unexposed post famine-born cohort (1964-65) in rural samples. No association was observed among urban samples. Results were robust to adjustment for covariates. CONCLUSIONS Severe undernutrition during gestation and the early postnatal period may have long-term effects on levels of proteinuria in humans, but the effect sizes may be small.
Collapse
Affiliation(s)
- Cheng Huang
- George Washington University - Department of Global Health, Washington, District of Columbia, United States, UC Berkeley School of Public Health - Health Services & Policy Analysis, Berkeley, California, United States, University of Maryland School of Public Health, College Park, Maryland, United States and Emory University - Hubert Department of Global Health, Atlanta, Georgia, United States
| | - Chaoran Guo
- George Washington University - Department of Global Health, Washington, District of Columbia, United States, UC Berkeley School of Public Health - Health Services & Policy Analysis, Berkeley, California, United States, University of Maryland School of Public Health, College Park, Maryland, United States and Emory University - Hubert Department of Global Health, Atlanta, Georgia, United States
| | - Catherine Nichols
- George Washington University - Department of Global Health, Washington, District of Columbia, United States, UC Berkeley School of Public Health - Health Services & Policy Analysis, Berkeley, California, United States, University of Maryland School of Public Health, College Park, Maryland, United States and Emory University - Hubert Department of Global Health, Atlanta, Georgia, United States
| | - Shuo Chen
- George Washington University - Department of Global Health, Washington, District of Columbia, United States, UC Berkeley School of Public Health - Health Services & Policy Analysis, Berkeley, California, United States, University of Maryland School of Public Health, College Park, Maryland, United States and Emory University - Hubert Department of Global Health, Atlanta, Georgia, United States
| | - Reynaldo Martorell
- George Washington University - Department of Global Health, Washington, District of Columbia, United States, UC Berkeley School of Public Health - Health Services & Policy Analysis, Berkeley, California, United States, University of Maryland School of Public Health, College Park, Maryland, United States and Emory University - Hubert Department of Global Health, Atlanta, Georgia, United States
| |
Collapse
|
388
|
Pangilinan F, Molloy AM, Mills JL, Troendle JF, Parle-McDermott A, Kay DM, Browne ML, McGrath EC, Abaan HO, Sutton M, Kirke PN, Caggana M, Shane B, Scott JM, Brody LC. Replication and exploratory analysis of 24 candidate risk polymorphisms for neural tube defects. BMC MEDICAL GENETICS 2014; 15:102. [PMID: 25293959 PMCID: PMC4411759 DOI: 10.1186/s12881-014-0102-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 08/19/2014] [Indexed: 12/14/2022]
Abstract
Background Neural tube defects (NTDs), which are among the most common congenital malformations, are influenced by environmental and genetic factors. Low maternal folate is the strongest known contributing factor, making variants in genes in the folate metabolic pathway attractive candidates for NTD risk. Multiple studies have identified nominally significant allelic associations with NTDs. We tested whether associations detected in a large Irish cohort could be replicated in an independent population. Methods Replication tests of 24 nominally significant NTD associations were performed in racially/ethnically matched populations. Family-based tests of fifteen nominally significant single nucleotide polymorphisms (SNPs) were repeated in a cohort of NTD trios (530 cases and their parents) from the United Kingdom, and case–control tests of nine nominally significant SNPs were repeated in a cohort (190 cases, 941 controls) from New York State (NYS). Secondary hypotheses involved evaluating the latter set of nine SNPs for NTD association using alternate case–control models and NTD groupings in white, African American and Hispanic cohorts from NYS. Results Of the 24 SNPs tested for replication, ADA rs452159 and MTR rs10925260 were significantly associated with isolated NTDs. Of the secondary tests performed, ARID1A rs11247593 was associated with NTDs in whites, and ALDH1A2 rs7169289 was associated with isolated NTDs in African Americans. Conclusions We report a number of associations between SNP genotypes and neural tube defects. These associations were nominally significant before correction for multiple hypothesis testing. These corrections are highly conservative for association studies of untested hypotheses, and may be too conservative for replication studies. We therefore believe the true effect of these four nominally significant SNPs on NTD risk will be more definitively determined by further study in other populations, and eventual meta-analysis. Electronic supplementary material The online version of this article (doi:10.1186/s12881-014-0102-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Faith Pangilinan
- Molecular Pathogenesis Section, Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Room 5306, 50 South Drive, Bethesda, MD, 20892-8004, USA.
| | - Anne M Molloy
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland.
| | - James L Mills
- Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| | - James F Troendle
- Office of Biostatistics Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | | | - Denise M Kay
- New York State Department of Health, Division of Genetics, Wadsworth Center, Albany, NY, 12201, USA.
| | - Marilyn L Browne
- New York State Department of Health, Congenital Malformations Registry, Troy, NY, 12180, USA. .,Department of Epidemiology and Biostatistics, School of Public Health, University at Albany, Rensselaer, NY, 12144, USA.
| | - Emily C McGrath
- New York State Department of Health, Division of Genetics, Wadsworth Center, Albany, NY, 12201, USA.
| | - Hatice Ozel Abaan
- Molecular Pathogenesis Section, Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Room 5306, 50 South Drive, Bethesda, MD, 20892-8004, USA.
| | - Marie Sutton
- Evidence Centre, Health Research Board, Dublin, Ireland.
| | - Peadar N Kirke
- Child Health Epidemiology Unit, Health Research Board, Dublin, Ireland.
| | - Michele Caggana
- New York State Department of Health, Division of Genetics, Wadsworth Center, Albany, NY, 12201, USA.
| | - Barry Shane
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, 94720-3104, USA.
| | - John M Scott
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland.
| | - Lawrence C Brody
- Molecular Pathogenesis Section, Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Room 5306, 50 South Drive, Bethesda, MD, 20892-8004, USA.
| |
Collapse
|
389
|
Ban L, West J, Abdul Sultan A, Dhalwani NN, Ludvigsson JF, Tata LJ. Limited risks of major congenital anomalies in children of mothers with coeliac disease: a population-based cohort study. BJOG 2014; 122:1833-41. [DOI: 10.1111/1471-0528.13102] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2014] [Indexed: 01/08/2023]
Affiliation(s)
- L Ban
- Division of Epidemiology & Public Health; School of Medicine; Nottingham City Hospital; University of Nottingham; Nottingham UK
| | - J West
- Division of Epidemiology & Public Health; School of Medicine; Nottingham City Hospital; University of Nottingham; Nottingham UK
| | - A Abdul Sultan
- Division of Epidemiology & Public Health; School of Medicine; Nottingham City Hospital; University of Nottingham; Nottingham UK
| | - NN Dhalwani
- Division of Epidemiology & Public Health; School of Medicine; Nottingham City Hospital; University of Nottingham; Nottingham UK
| | - JF Ludvigsson
- Department of Medical Epidemiology and Biostatistics; Karolinska Institutet; Karolinska University Hospital; Stockholm Sweden
- Department of Paediatrics; Örebro University Hospital; Örebro Sweden
| | - LJ Tata
- Division of Epidemiology & Public Health; School of Medicine; Nottingham City Hospital; University of Nottingham; Nottingham UK
| |
Collapse
|
390
|
Abstract
INTRODUCTION There is increasingly a double burden of under-nutrition and obesity in women of reproductive age. Preconception underweight or overweight, short stature and micronutrient deficiencies all contribute to excess maternal and fetal complications during pregnancy. METHODS A systematic review and meta-analysis of the evidence was conducted to ascertain the possible impact of preconception care for adolescents, women and couples of reproductive age on maternal, newborn and child health (MNCH) outcomes. A comprehensive strategy was used to search electronic reference libraries, and both observational and clinical controlled trials were included. Cross-referencing and a separate search strategy for each preconception risk and intervention ensured wider study capture. RESULTS Maternal pre-pregnancy weight is a significant factor in the preconception period with underweight contributing to a 32% higher risk of preterm birth, and obesity more than doubling the risk for preeclampsia, gestational diabetes. Overweight women are more likely to undergo a Cesarean delivery, and their newborns have higher chances of being born with a neural tube or congenital heart defect. Among nutrition-specific interventions, preconception folic acid supplementation has the strongest evidence of effect, preventing 69% of recurrent neural tube defects. Multiple micronutrient supplementation shows promise to reduce the rates of congenital anomalies and risk of preeclampsia. Although over 40% of women worldwide are anemic in the preconception period, only one study has shown a risk for low birth weight. CONCLUSION All women, but especially those who become pregnant in adolescence or have closely-spaced pregnancies (inter-pregnancy interval less than six months), require nutritional assessment and appropriate intervention in the preconception period with an emphasis on optimizing maternal body mass index and micronutrient reserves. Increasing coverage of nutrition-specific and nutrition-sensitive strategies (such as food fortification; integration of nutrition initiatives with other maternal and child health interventions; and community based platforms) is necessary among adolescent girls and women of reproductive age. The effectiveness of interventions will need to be simultaneously monitored, and form the basis for the development of improved delivery strategies and new nutritional interventions.
Collapse
Affiliation(s)
- Sohni V Dean
- Division of Women and Child Health, Aga Khan University Karachi, Pakistan
| | - Zohra S Lassi
- Division of Women and Child Health, Aga Khan University Karachi, Pakistan
| | - Ayesha M Imam
- Division of Women and Child Health, Aga Khan University Karachi, Pakistan
| | - Zulfiqar A Bhutta
- Division of Women and Child Health, Aga Khan University Karachi, Pakistan
| |
Collapse
|
391
|
Mavrogenis S, Urban R, Czeizel AE, Ács N. Possible preventive effect of high doses of folic acid for isolated hypospadias: A national population-based case-control study. Am J Med Genet A 2014; 164A:3108-14. [DOI: 10.1002/ajmg.a.36781] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 08/23/2014] [Indexed: 02/01/2023]
Affiliation(s)
- Stelios Mavrogenis
- Department of Uro-oncology; National Institute of Oncology; Budapest Hungary
| | - Róbert Urban
- Department of Personality and Health Psychology; Eötvös Loránd University; Budapest Hungary
| | - Andrew E. Czeizel
- Foundation for the Community Control of Hereditary Diseases; Budapest; Hungary
| | - Nándor Ács
- Second Department of Obstetrics and Gynecology; Semmelweis University, School of Medicine; Budapest Hungary
| |
Collapse
|
392
|
Dong Y, Wang X, Zhang J, Guan Z, Xu L, Wang J, Zhang T, Niu B. Raltitrexed's effect on the development of neural tube defects in mice is associated with DNA damage, apoptosis, and proliferation. Mol Cell Biochem 2014; 398:223-31. [PMID: 25245820 DOI: 10.1007/s11010-014-2222-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 09/15/2014] [Indexed: 12/13/2022]
Abstract
The causal metabolic pathway and the underlying mechanism between folate deficiency and neural tube defects (NTDs) remain obscure. Thymidylate (dTMP) is catalyzed by thymidylate synthase (TS) using the folate-derived one-carbon unit as the sole methyl donor. This study aims to examine the role of dTMP biosynthesis in the development of neural tube in mice by inhibition of TS via a specific inhibitor, raltitrexed (RTX). Pregnant mice were intraperitoneally injected with various doses of RTX on gestational day 7.5, and embryos were examined for the presence of NTDs on gestational day 11.5. TS activity and changes of dUMP and dTMP levels were measured following RTX treatment at the optimal dose. DNA damage was determined by detection of phosphorylated replication protein A2 (RPA2) and γ-H2AX in embryos with NTDs induced by RTX. Besides, apoptosis and proliferation were also analyzed in RTX-treated embryos with NTDs. We found that NTDs were highly occurred by the treatment of RTX at the optimal dose of 11.5 mg/kg b/w. RTX treatment significantly inhibited TS activity. Meanwhile, dTMP was decreased associated with the accumulation of dUMP in RTX-treated embryos. Phosphorylated RPA2 and γ-H2AX were significantly increased in RTX-treated embryos with NTDs compared to control. More apoptosis and decreased proliferation were also found in embryos with NTDs induced by RTX. These results indicate that impairment of dTMP biosynthesis caused by RTX led to the development of NTDs in mice. DNA damage and imbalance between apoptosis and proliferation may be potential mechanisms.
Collapse
Affiliation(s)
- Yanting Dong
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, China
| | | | | | | | | | | | | | | |
Collapse
|
393
|
Binia A, Contreras AV, Canizales-Quinteros S, Alonzo VA, Tejero ME, Silva-Zolezzi I. Geographical and ethnic distribution of single nucleotide polymorphisms within genes of the folate/homocysteine pathway metabolism. GENES & NUTRITION 2014; 9:421. [PMID: 25106483 PMCID: PMC4172644 DOI: 10.1007/s12263-014-0421-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 07/25/2014] [Indexed: 12/22/2022]
Abstract
High levels of plasma homocysteine are associated with an increased risk of many health conditions influenced by both environmental and genetic factors. The objective of this study was to provide the geographical distribution of folate pathway genetic polymorphisms in Mexico and the comparison with the reported frequencies in different continental populations. This study included the analysis of the genotypic frequencies of eight polymorphisms in genes of the folate/homocysteine metabolic pathway in 1,350 Mestizo and Amerindian subjects from different regions in Mexico and 836 individuals from European, African and Asian populations of the 1,000 Genomes Project. In Mexican Mestizo and Amerindian populations, the MTHFR C677T risk genotype (TT) was highly prevalent (frequency: 25 and 57 %, respectively). In Mestizos, the frequency showed clear regional variation related to ancestry; the Guerrero subpopulation with the highest Amerindian contribution had the highest TT frequency (33 %). The MTHFD1 G1958A AA risk genotype was also enriched in Mexican Mestizos and Amerindians (frequency: 34 and 58 %, respectively), whereas in African and Asian ancestry populations the frequency for AA was low (~4 %). All together risk genotypes showed regional differences, and Sonora had significantly different genetic frequencies compared with the other regions (P value <0.05). Our study illustrates differential geographical distribution of the risk variants in the folate/homocysteine metabolic pathway relative to ethnic background. This work supports that certain areas of the world have increased needs for folic acid and vitamin B supplementation, and this information needs to be considered in public health guidelines and eventually policies.
Collapse
Affiliation(s)
- Aristea Binia
- Nutrition and Health Research, Nestlé Research Center, Lausanne, Switzerland,
| | | | | | | | | | | |
Collapse
|
394
|
Technology of foam fractionation coupled with crystallization for the enrichment and purification of folic acid. Sep Purif Technol 2014. [DOI: 10.1016/j.seppur.2014.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
395
|
Bigger babies born to women survivors of the 1959-1961 Chinese famine: a puzzle due to survival selection? J Dev Orig Health Dis 2014; 1:412-8. [PMID: 25142012 DOI: 10.1017/s2040174410000504] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The Chinese Famine of 1959-1961 caused up to 30 million deaths. It varied in intensity across China and affected rural areas disproportionately. Data from the China-U.S. Collaborative Project for Neural Tube Defect Prevention on 31, 449 women (born 1957-1963) and their offspring birth size were recorded in 1993-1996. We used a measure of famine intensity at county level based on the size of famine-born cohorts relative to cohorts preceding and following the famine in a difference-in-difference model that compared offspring birth size of pre-famine (1957-1958; exposed between 0.5 and 4.5 years), famine (1959-1961; prenatal and up to 2.5 years) and post-famine (1962; some exposed in early pregnancy) cohort groups to that of the unexposed 1963 cohort. The model corrected for age and cohort trends and estimated associations between maternal famine exposure and offspring birth size for the average level of famine intensity across counties, and included adjustment for clustering. In rural areas and in pre-famine and famine cohorts, exposure to famine was associated with larger weight (69 g; 95% CI 30, 108), length (0.3 cm; 95% CI -0.0, 0.5) and birth body mass index (0.1 kg/m2; 95% CI 0.0, 0.2). In urban areas, however, exposure to famine was not associated with offspring birth size. Our findings in rural areas suggest that severe and prolonged famine leads to larger newborn size in the offspring of mothers exposed to famine in utero and during the first few years of life; less severe famine in urban areas however, appeared to have no impact. The markedly increased mortality in rural areas may have resulted in the selection of hardier mothers with greater growth potential, which becomes expressed in their offspring.
Collapse
|
396
|
Tsurubuchi T, Allender EV, Siddiqui MR, Shim KW, Ichi S, Boshnjaku V, Mania-Farnell B, Xi G, Finnell RH, McLone DG, Tomita T, Mayanil CS. A critical role of noggin in developing folate-nonresponsive NTD in Fkbp8 -/- embryos. Childs Nerv Syst 2014; 30:1343-53. [PMID: 24817375 DOI: 10.1007/s00381-014-2428-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 04/16/2014] [Indexed: 01/07/2023]
Abstract
PURPOSE Maternal folate intake has reduced the incidence of human neural tube defects by 60-70 %. However, 30-40 % of cases remain nonresponsive to folate intake. The main purpose of this study was to understand the molecular mechanism of folate nonresponsiveness in a mouse model of neural tube defect. METHODS We used a folate-nonresponsive Fkbp8 knockout mouse model to elucidate the molecular mechanism(s) of folate nonresponsiveness. Neurospheres were grown from neural stem cells isolated from the lumbar neural tube of E9.5 Fkbp8 (-/-) and wild-type embryos. Immunostaining was used to determine the protein levels of oligodendrocyte transcription factor 2 (Olig2), Nkx6.1, class III beta-tubulin (TuJ1), O4, glial fibrillary acidic protein (GFAP), histone H3 Lys27 trimethylation (H3K27me3), ubiquitously transcribed tetratricopeptide repeat (UTX), and Msx2, and quantitative real-time (RT)-PCR was used to determine the message levels of Olig2, Nkx6.1, Msx2, and noggin in neural stem cells differentiated in the presence and absence of folic acid. RESULTS Fkbp8 (-/-)-derived neural stem cells showed (i) increased noggin expression; (ii) decreased Msx2 expression; (iii) premature differentiation--neurogenesis, oligodendrogenesis (Olig2 expression), and gliogenesis (GFAP expression); and (iv) increased UTX expression and decreased H3K27me3 polycomb modification. Exogenous folic acid did not reverse these markers. CONCLUSIONS Folate nonresponsiveness could be attributed in part to increased noggin expression in Fkbp8 (-/-) embryos, resulting in decreased Msx2 expression. Folate treatment further increases Olig2 and noggin expression, thereby exacerbating ventralization.
Collapse
Affiliation(s)
- Takao Tsurubuchi
- Developmental Neurobiology Laboratory, Department of Pediatric Neurosurgery, Developmental Biology Program, Lurie Children's Research Center, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, 60614, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
397
|
Crider KS, Devine O, Hao L, Dowling NF, Li S, Molloy AM, Li Z, Zhu J, Berry RJ. Population red blood cell folate concentrations for prevention of neural tube defects: Bayesian model. BMJ 2014; 349:g4554. [PMID: 25073783 PMCID: PMC4115151 DOI: 10.1136/bmj.g4554] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/23/2014] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To determine an optimal population red blood cell (RBC) folate concentration for the prevention of neural tube birth defects. DESIGN Bayesian model. SETTING Data from two population based studies in China. PARTICIPANTS 247,831 participants in a prospective community intervention project in China (1993-95) to prevent neural tube defects with 400 μg/day folic acid supplementation and 1194 participants in a population based randomized trial (2003-05) to evaluate the effect of folic acid supplementation on blood folate concentration among Chinese women of reproductive age. INTERVENTION Folic acid supplementation (400 μg/day). MAIN OUTCOME MEASURES Estimated RBC folate concentration at time of neural tube closure (day 28 of gestation) and risk of neural tube defects. RESULTS Risk of neural tube defects was high at the lowest estimated RBC folate concentrations (for example, 25.4 (95% uncertainty interval 20.8 to 31.2) neural tube defects per 10,000 births at 500 nmol/L) and decreased as estimated RBC folate concentration increased. Risk of neural tube defects was substantially attenuated at estimated RBC folate concentrations above about 1000 nmol/L (for example, 6 neural tube defects per 10,000 births at 1180 (1050 to 1340) nmol/L). The modeled dose-response relation was consistent with the existing literature. In addition, neural tube defect risk estimates developed using the proposed model and population level RBC information were consistent with the prevalence of neural tube defects in the US population before and after food fortification with folic acid. CONCLUSIONS A threshold for "optimal" population RBC folate concentration for the prevention of neural tube defects could be defined (for example, approximately 1000 nmol/L). Population based RBC folate concentrations, as a biomarker for risk of neural tube defects, can be used to facilitate evaluation of prevention programs as well as to identify subpopulations at elevated risk for a neural tube defect affected pregnancy due to folate insufficiency.
Collapse
Affiliation(s)
- Krista S Crider
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Owen Devine
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Ling Hao
- Peking University Health Science Center, Peking University, Beijing, China US Centers for Disease Control and Prevention, US Embassy, Beijing, China
| | - Nicole F Dowling
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Song Li
- Peking University Third Hospital, Beijing, China
| | - Anne M Molloy
- School of Medicine, Trinity College, Dublin, Ireland
| | - Zhu Li
- Peking University Health Science Center, Peking University, Beijing, China
| | - Jianghui Zhu
- Peking University Health Science Center, Peking University, Beijing, China Division of Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing, China
| | - Robert J Berry
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| |
Collapse
|
398
|
Meng Q, Zhang L, Liu J, Li Z, Jin L, Zhang Y, Wang L, Ren A. Dietary folate intake levels in rural women immediately before pregnancy in Northern China. ACTA ACUST UNITED AC 2014; 103:27-36. [PMID: 25066482 DOI: 10.1002/bdra.23280] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/04/2014] [Accepted: 06/11/2014] [Indexed: 01/24/2023]
Abstract
BACKGROUND The study aims to assess dietary folate levels and food sources in women immediately before pregnancy in a rural area of northern China associated with a high prevalence of neural tube defects. METHODS Information was collected by face-to-face interviews with women who sought premarital healthcare and planned to become pregnant within the next 12 months from November 2009 through December 2012. Information regarding food consumption was obtained by means of 24-hr dietary recall. Folate values were assigned to foods according to the China Food Composition 2004. Factors associated with dietary folate intake were analyzed by multiple linear regression. RESULTS Mean (± standard deviation) and median (interquartile range) daily folate intake levels were 114.3 ± 59.7 and 102.8 (69.3-146.8) μg/day, respectively. Over 99% of the subjects had an intake level below 320 μg/day, the estimated average requirement for nonpregnant women. Only 1% and 7% of the women consumed 75% and 50%, respectively, of the recommended daily folate intake of 400 μg for nonpregnant women. Over 80% of total folate consumption came from cereals, vegetables, and tubers, whereas fruit consumption was severely lacking. Underweight women, farmers, women enrolled during the winter, and women with access to fewer food types or daily meals were more likely to exhibit low folate intake levels. CONCLUSION Dietary folate intake among study participants was far below the recommended intake level. Folic acid fortification of cereals is advised to raise folate intake in rural Chinese women planning to become pregnant.
Collapse
Affiliation(s)
- Qinqin Meng
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Peking, China; Institute of Reproductive and Child Health/Ministry of Health Key Laboratory of Reproductive Health, Peking University; and Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
399
|
Affiliation(s)
- Joseph F Cubells
- Department of Human Genetics and Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
400
|
Zugna D, Richiardi L, Stephansson O, Pasternak B, Ekbom A, Cnattingius S, Ludvigsson JF. Risk of congenital malformations among offspring of mothers and fathers with celiac disease: a nationwide cohort study. Clin Gastroenterol Hepatol 2014; 12:1108-1116.e6. [PMID: 24161347 DOI: 10.1016/j.cgh.2013.10.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 10/02/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Many patients with celiac disease experience malabsorption, weight loss, and anemia; undiagnosed celiac disease during pregnancy has been linked with adverse outcomes. Studies of celiac disease and congenital malformations in offspring have been underpowered. We investigated the risk of congenital malformations among the offspring of parents with celiac disease. METHODS We performed a nationwide cohort study of data from linked health care registers in Sweden from 1973 through 2009. We collected histopathology data from 28 pathology departments in Sweden to identify individuals with celiac disease (based on the presence of villous atrophy). We estimated the risks of malformations in the offspring of mothers and fathers with and without celiac disease. Logistic regression was used to estimate adjusted prevalence odds ratios (aPORs) with 95% confidence intervals (CIs). RESULTS Among 11,382 offspring of mothers with celiac disease, there were 672 cases (5.9%) of malformation compared with 2098 cases (5.1%) among 40,922 offspring of mothers without celiac disease. Similarly, 352 (5.9%) of 6002 offspring of fathers with celiac disease and 1009 (5.1%) of 19,600 offspring of fathers without celiac disease had a malformation. In adjusted analyses, the offspring of mothers or fathers with celiac disease had a slightly increased risk of having children with malformations (for those with mothers with celiac disease: aPOR, 1.15; 95% CI, 1.05-1.26; for those with fathers with celiac disease: aPOR, 1.14; 95% CI, 1.00-1.29). However, these excess risks decreased or vanished entirely when we restricted our data to births since 2000 (for those with mothers with celiac disease: aPOR, 1.11; and 95% CI, 0.79-1.56; for those with fathers with celiac disease: aPOR, 1.01; 95% CI, 0.81-1.26). CONCLUSIONS In a nationwide study, we found an increased risk for malformation among the offspring of mothers or fathers with celiac disease. However, the excess risk is small; the upper limits of the CIs for malformation indicate a 29% maximum relative increase.
Collapse
Affiliation(s)
- Daniela Zugna
- Cancer Epidemiology Unit, CPO-Piemonte and University of Turin, Turin, Italy; Clinical Epidemiology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lorenzo Richiardi
- Cancer Epidemiology Unit, CPO-Piemonte and University of Turin, Turin, Italy
| | - Olof Stephansson
- Clinical Epidemiology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet, Stockholm, Sweden
| | - Björn Pasternak
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Anders Ekbom
- Clinical Epidemiology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sven Cnattingius
- Clinical Epidemiology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jonas F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Department of Pediatrics, Örebro University Hospital, Sweden.
| |
Collapse
|