<<

American Journal of Medical Part C (Seminars in ) 157:215–226 (2011) ARTICLE

Genetic Basis of Susceptibility to Teratogen Induced Birth Defects BOGDAN J. WLODARCZYK,* ANA M. PALACIOS, CLAUDIA J. CHAPA, HUIPING ZHU, TIMOTHY M. GEORGE, AND RICHARD H. FINNELL

Birth defects remain the leading cause of infant death in US. The field of has been focused on the causes and underlying mechanisms of birth defects for decades, yet our understanding of these critical issues remain unacceptably vague. Conclusions from years of animal and studies made it clear that the vast majority of birth defects have multifactorial origins, with contributions from environmental and genetic factors. The environment comprises not only of the physical, biological, and chemical external environment surrounding the pregnant woman, but it also includes the internal environment of the woman’s body that interact with the developing in a complex fashion. The importance of maternal and embryonic genetic factors consisting of countless genetic variants/mutations that exist within every individual contribute to susceptibility is only now being more fully appreciated. This great complexity of the genome and its diversity within individuals and populations seems to be the principal reason why the same teratogenic exposure can induce severe malformation in one embryo, while fail to do so to other exposed . As the interaction between genetic and environmental factors has long been recognized as the first ‘‘Principle of Teratology’’ by Wilson and Warkany [1965. Teratology: Principles and techniques. Chicago: University of Chicago Press], it is only recently that the appropriate investigative tools have been developed with which to fully investigate this fundamental principle. The introduction of high throughput technologies like whole genome sequencing or genome-wide association studies are promising to deliver an enormous amount of new data that will shed light on the genomic factors that contribute susceptibility to environmental teratogens. In this review, we attempt to summarize the epidemiological and experimental literature concerning birth defects whose phenotypic expression can be clearly related to the interactions between several select environmental factors and those genetic pathways in which they are most likely to have significant modifying effects. ß 2011 Wiley-Liss, Inc.

KEY WORDS: birth defects; teratogen; genome; mutations; gene–environment interaction How to cite this article: Wlodarczyk B, Palacios A, Chapa C, Zhu H, George TM, Finnell RH. 2011. Genetic basis of susceptibility to teratogen induced birth defects. Am J Med Genet Part C Semin Med Genet 157:215–226.

INTRODUCTION ple organs including: the brain, heart, including inherited (genetic) condi- lungs, liver, bones, intestinal tract, limbs tions, toxic exposure of the embryo Birth defects are abnormalities present at and other structures. These defects and , and as is most often the case, or before birth, and may involve multi- can occur for a variety of reasons for as yet unknown reasons. In the

Bogdan J. Wlodarczyk is an assistant Professor at the Dell Pediatric Research Institute and holds a faculty position in the Department of Nutritional Sciences, The University of Texas at Austin. He is a reproductive toxicologist and uses animal models to study the mechanisms of birth defects. Ana M. Palacios is a physician whose clinical activities and research have focused mainly on clinical genetics and neural tube defects. She has been involved in the creation and consolidation of two independent birth defect surveillance programs in Colombia and Nicaragua. Currently she is a graduate student in Dr. Richard Finnell’s laboratory. Claudia J. Chapa is a physician currently involved in the Medical Genetics research team in Dr. Richard Finnell’s laboratory. Her main research interest is in identifying genetic polymorphisms in patients with birth defects. Huiping Zhu is an assistant Professor at the Dell Pediatric Research Institute, Department of Nutritional Sciences, The University of Texas at Austin. Dr. Zhu has a doctoral degree in , and has been working on birth defects etiology, especially gene-environment interactions. Timothy M. George is the Chief of Pediatric Neurosurgery and at the Dell Children’s Medical Center in Austin, Additionally; he is an Adjunct Professor of Cell and Molecular at the University of Texas at Austin and serves as Director of the Pediatric Educational and Translational Research Institute. Dr George serves on Biomedical Engineering Advisory Board and the McComb Business School Health Care Initiative Advisory Board at the University of Texas at Austin. His research focuses on the molecular genetics of neural tube defects. Richard H. Finnell is a Professor in the Department of Nutritional Sciences and in the Department of Chemistry and at the University of Texas at Austin, and serves as the Director of Genomic Research at Dell Children’s Medical Center. A pediatric geneticist, he has a distinguished career researching environmentally induced birth defects. Research in the Finnell Laboratory focuses on the interaction between specific genes and nutritional factors as they influence normal and susceptibility to complex birth defects. *Correspondence to: Bogdan Wlodarczyk, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd., Austin, TX 78723. E-mail: [email protected] DOI 10.1002/ajmg.c.30314 Published online 15 July 2011 in Wiley Online Library (wileyonlinelibrary.com).

ß 2011 Wiley-Liss, Inc. 216 AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) ARTICLE

United States, 1 out of every 33 babies is variants from multiple genes have been Finnell et al., 2004; Graham and Shaw, born with a major birth defect (http:// linked to NTDs risk individually with 2005; Shi et al., 2008]. One commonly www.nbdpn.org/docs/US_2010_C.pdf). rather small effect [van der Put et al., cited birth defect-oriented example of a The most common types of birth defects 1995; Ou et al., 1996; Volcik et al., GxE interaction is the effect of maternal include congenital heart defects (CHDs) 2003a,b; Zhu et al., 2003, 2005, 2006, smoking and an important detoxifica- and neural tube defects (NTDs), and 2007; van der Linden et al., 2006, 2007; tion gene, CYP1A1, on risk for oral these defects can be very serious, often Parle-McDermott et al., 2007; David- clefts in the exposed embryos [van Rooij life-threatening. Another group of son et al., 2008], leading us to believe et al., 2001; Shi et al., 2007]. This is common birth defects, craniofacial that these small effects interact with consistent with the extreme complexity malformations, include cleft lip with or select environmental factors to create a of the developmental processes during without cleft palate (CL/P) and cleft more significant developmental disequi- early embryogenesis. palate only (CPO), while less commonly librium beyond which these genetic Biological interactions and their life-threatening, they require compre- factors could have produced individual- role in etiological mechanisms require hensive medical and surgical attention. ly, disrupting normal development, and further functional studies [Ottman, Currently, about 70% of the causes resulting in the abnormal phenotype 1990]. Herein we will review both of birth defects are unknown. The [Prescott et al., 2001; Murray, 2002; human and animal studies on the genetic etiology of the majority of birth defects basis of environmentally induced birth whose causes are unclear is believed to defects. Inherited diseases caused by involve both environmental and genetic For example, more than chromosomal abnormalities, as well as factors. The list of environmental factors defects caused by single genes are not that have been associated with birth 240 spontaneous and within the scope of this review. defects is long and remarkably varied genetically modified mouse [Blom et al., 2006]. Some examples models of NTDs exist, with 205 FOLATE PATHWAY GENES include: maternal smoking [Shi et al., AND NTDs 2007; Shea and Steiner, 2008; Suarez of them representing specific et al., 2008, 2011], organic solvents It has been demonstrated that folic [Brender and Suarez, 1990; Brender genes. These models provided acid supplementation prevents NTDs et al., 2002], environmental toxicants abundant candidate genes for [Smithells et al., 1980; Czeizel, 2000; Au like pesticides, nitrates, and heavy metals human studies; however, very et al., 2010; Blencowe et al., 2010; [White et al., 1988; Sever, 1995; Shaw Obican et al., 2010; Collins et al., 2011] et al., 1999b], inadequate intake of few mutations have been and other birth defects such as cono- nutrients [Shaw et al., 1999a; Carmi- found in these genes providing truncal heart defects [Botto et al., 2003b; chael et al., 2003], maternal fever/ Bailey and Berry, 2005]. Folate metab- influenza [Shaw et al., 1998a; Lundberg evidence as having a major role olism is a complex process involving a et al., 2003], maternal and for human NTDs. Instead, large number of enzymatic reactions, [Watkins and Botto, 2001; variants from multiple genes producing a variety of intermediates that Watkins et al., 2003; Waller et al., function in vital physiological processes 2007]. On the other hand, there is an have been linked to NTDs risk such as nucleotide synthesis, cell divi- abundance of supportive evidence that individually with rather small sion, and . Perturbation of complex birth defects have a genetic this complex process may result in frank component [Campbell et al., 1986; effect, leading us to believe that folate deficiency and/or accumulation Shaw et al., 1994; Blom et al., 2006]. these small effects interact with of homocysteine. Animal studies have One important source of scientific generated several hypotheses regarding evidence supporting a genetic contribu- select environmental factors the mechanisms underlying the preven- tion to the etiology of birth defects to create a more significant tive effect of folic acid. Inactivation of Folr1 Folr2 comes from studies involving animal developmental disequilibrium folate pathway genes such as , , models. For example, more than 240 or Mthfr renders the developing embryo spontaneous and genetically modified beyond which these genetic susceptible to an increased risk of NTDs mouse models of NTDs exist, with 205 factors could have produced following in utero exposure to specific of them representing specific genes teratogens. Human studies exploring the [Harris and Juriloff, 2010]. These mod- individually, disrupting interaction between folate pathway els provided abundant candidate genes normal development, genes and teratogens on NTDs risk have for human studies; however, very few and resulting in the been limited by the insufficiency of mutations have been found in these statistical power due to small sample size. genes providing evidence as having a abnormal phenotype. There have been reports of possible major role for human NTDs. Instead, interactions between the MTHFR ARTICLE AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) 217

C677T polymorphism and maternal When the Folr2 knockout mice, NTDs and some CHDs [Kallen, 1998; smoking [Hobbs et al., 2006], BHMT which do not present with any patho- Grewal et al., 2008], the vast majority of and obesity, as well as transcobalamin II logical phenotype even in the nullizy- studies have reported an increased risk of (TCII) and maternal use [Hobbs gous state, were administered sodium adverse outcomes including: et al., 2010], on CHD risk. Blanton et al. arsenate, the treatment was significantly birth defects, preterm delivery, sponta- / [2011] recently explored a number of more teratogenic to Folr2 mice than neous , growth restriction, or þ/þ folate pathway genes (including folate to wildtype (Folr2 ) mice. To further negative neurodevelopmental outcomes transport genes) as risk factors for non- investigate how additional environmen- in the exposed offspring of women who syndromic CLP, and found possible tal factors (potentially teratogenic) can smoked [Shea and Steiner, 2008; Suarez interactions between polymorphisms in modulate this gene–teratogen interac- et al., 2008, 2011; Shaw et al., 2009], or FOLR1 and FOLR2 and maternal tion, the Folr2 knockout mice were fed a were breathing in second hand smoke smoking. folate deficient diet during the pericon- [Venners et al., 2004; Meeker et al., Data from animal studies concern- ceptional period and throughout the 2007; Li et al., 2008; Suarez et al., 2008, ing the possible involvement of folate pregnancy and the arsenate was admin- 2011]. Association between maternal pathway genes in teratogen-induced istered during the period of neural tube cigarette smoking and non-syndromic birth defects remain equivocal. In our closure. In this experiment, the terato- orofacial clefts (NSOFC) has been / laboratory,we have treated four different genic effect observed in Folr2 was observed in a number of epidemiological knockout mouse strains (Rfc1, Folr1, considerably exacerbated by the low studies [Lorente et al., 2000; Little et al., Folr2, and Mthfr) that are involved folate diet (exencephaly rate increased 2004; Shaw et al., 2009]. þ/þ in folate transport or metabolism with from 40 to 64%), whereas in Folr2 Hwang et al. [1995] was the first to sodium arsenate, a known mouse neural mice, the rate of NTDs did not change suggest a possible interaction between tube teratogen [Wlodarczyk et al., 2001, [Wlodarczyk et al., 2001]. These results the transforming growth factor alpha 2006a; Spiegelstein et al., 2005]. The illustrate that the gene–teratogen inter- (TGFa) Taq1 variant and maternal Rfc1 knockout mice treated with 40 mg/ actions can be further complicated when smoking on NSOFC [Hwang et al., kg of sodium arsenate, failed to show more than one teratogen is considered. 1995]. TGFa is a secretory protein that any dependent differences Some teratogens can act in a synergistic binds to the epidermal growth factor in teratogenic outcomes. The rates of manner in individuals with ‘‘sensitive’’ receptor. It plays important roles in cell early embryonic loss (resorptions) and genotype, as exemplified by arsenic and migration and differentiation, and is / exencephaly (NTDs) among folate deficiency in Folr2 nullizygous also a potent promoter of angiogenesis from treated wildtype and heterozygous mice. [Kumar et al., 1995]. Hwang reported Rfc1 dams did not differ significantly. The results following the treatment that TGFa Taq1 variant was associated However, there was a substantial differ- of Mthfr knockout mice in utero with with a higher risk of non-syndromic ence seen in arsenic embryotoxicity arsenic were somewhat similar to those cleft lip with or without cleft palate in þ/ between heterozygous Rfc1 and observed in the Rfc1 mice. There was no mothers who smoked 20 or more þ/ heterozygous Folr1 dams, another difference in teratogenic response to cigarettes/day. Infants carrying the less folate transport mouse line arsenic when the pregnancy outcomes common C2 allele who were exposed to described in this study [Spiegelstein in dams of different Mthfr (þ/ maternal smoking of 10 or less cigarettes þ/ et al., 2005]. In Rfc1 mice treated þ, þ/, and /) were compared. per day showed a 6.16-fold increase in with arsenic, 12–22% of the embryos However the Mthfr mutant mice were the risk for CPO [95% confidence þ/ were resorbed, whereas in Folr1 significantly more sensitive to arsenic interval (CI): 1.09–34.7], while the þ/ mice embryolethality reached 82% of teratogenicity than were the Rfc1 or risk was 8.69-fold higher (95%CI: / the conceptuses. Despite the fact that Folr2 knockout mice [Wlodarczyk 1.57–47.8) if the mothers smoked these two mutant mouse strains were not and Finnell, unpublished data]. more than 10 cigarettes/day. compared directly in that study, this is More recently in a population- still a compelling example that genetic based case control study in California, variants can deeply modulate the out- MATERNAL EXPOSURE TO Shaw et al. [1998b] observed an elevated come to a teratogen exposure. It should CIGARETTE SMOKE risk associated with the same TGFa be also noted, that only wildtype and Taq1 C2 allele in smoking mothers, heterozygous animals were used in this Embryonic exposure to cigarette smoke resulting in an OR of 6.1 (95%CI: 1.1– study, since Rfc1 and Folr1 homozygous (first-hand and/or passive) is a known 36.1) for cleft lip with or without cleft mutations are embryolethal. Having one risk factor for structural defects such as palate (CL/P), and an OR of 9 (95%CI: active copy of these genes is apparently orofacial clefts [Lie et al., 2008; Shi et al., 1.4–61.9) for CPO. It is noteworthy that sufficient for viability and it appears that 2008] and gastroschisis [Feldkamp et al., when analyzing interactions, the ORs, the heterozygous mice can compensate 2008]. Although there are a few studies although elevated, often have wide for their genetic deficit in folate metab- suggesting that maternal exposure to CIs, indicating the rather imprecise risk olism [Spiegelstein et al., 2005]. cigarette smoking is ‘‘protective’’ against estimations. 218 AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) ARTICLE

A meta-analysis examining five Maternal smoking has been identi- been associated with birth defects that case–control studies found a significant fied as a risk factor for defects other than are not typically considered to be a part overall smoking effect for CL/P craniofacial, including CHD [Malik of the FAS. For example, one study (OR ¼ 1.64, 95%CI ¼ 1.33–2.01) and et al., 2008; Kuciene and Dulskiene, found that women who consumed CPO (OR ¼ 1.42, 95%CI ¼ 1.06– 2010] and gastroschisis [Lammer et al., alcohol at least once a week had a 2.1- 1.90). There was no evidence of in- 2008; Chabra et al., 2011]. In a popula- fold increased risk (95%CI: 1.1, 4.0) of creased risk for CPO if the infant carried tion-based case–control study, women having a child affected with an NTD the C2 allele among non-smoking who smoked and carried a TCII—776 [Grewal et al., 2008]. mothers. However, if the mother CG or GG genotype—were 1.8 times Studies on attempting to under- reported smoking and the infant carried more likely to have a CHD-affected stand the genetic basis of susceptibility the C2 allele, there was an overall fetus than were women who smoked and to the etiology of this condition have increased risk for CP, with an OR of carried a CC genotype. The CHDs not been successful. Although a recent 1.95 (95%CI: 1.22–3.10). Based on were: non-syndromic septal, conotrun- population-based case–control study this meta-analysis, the TGFa genotype cal, right or left-sided obstructive heart reported that variants in the alcohol failed to demonstrate an increased risk defects [Hobbs et al., 2010]. TCII is dehydrogenase 1C (ADH1C) gene may for CL/P, regardless of maternal smok- an important enzymatic transporter of modify the association between alcohol ing status. The authors concluded that cobalamin from the intestinal lumen to and oral clefts, it did not extend to gene–environment interactions between the bloodstream, and eventually to target the whole pattern of FAS dysmorphic the infant’s TGFa Taq1 polymorphism tissues, suggesting a role for vitamin B12 features [Boyles et al., 2010]. Alcohol and maternal smoking were limited to in the etiology of this group of defects dehydrogenase (ADH) is an enzyme that CPO [Zeiger et al., 2005]. [Quadros et al., 1999]. Torfset al. [2006] oxidizes ethanol to form acetaldehyde Folic acid is an essential B vitamin assessed the interaction between mater- during its metabolism. Functional SNPs required for both amino and nucleic acid nal smoking and selected candidate in ADH1C gene are associated with a biosynthesis. It also participates in the genes in a case–control study of 57 cases reduced ratio of alcohol oxidation. In one-carbon unit transfers, a key step of gastroschisis and 506 controls. Thirty- this study, mothers who consumed five in biomethylation reactions. Quickly two genes involved in angiogenesis, or more alcoholic drinks per sitting proliferating cells of a rapidly growing blood vessel integrity, inflammation, during the first trimester of pregnancy embryo/fetus have a high demand for wound repair, and dermal/epidermal had an elevated risk for a malformed folate; therefore adequate supply of folic strength were interrogated for variants. infant with an oral cleft (OR of 2.6, acid is especially important for women Of these genes, NOS3 glu298asp, 95%CI: 1.4–4.7). This increased risk during pregnancy. Cigarette smoking ICAM1 gly241arg, and NPPA T2238C was evident only in mothers or children has been documented to lower folate showed a strong interaction with mater- who carried ADH1C haplotype that was and increase homocysteine blood levels nal smoking. Women who smoked and associated with slow alcohol metabolism [Ozerol et al., 2004; Jauniaux and carried one or two of the aforemen- (OR of 3.0, 95%CI: 1.4–6.8) [Boyles Burton, 2007]. High level of homocys- tioned variants were at a significantly et al., 2010]. There was no evidence of teine during pregnancy might affect higher risk of having an infant with alcohol-related risk when both mother important developmental processes gastroschisis, when compared to women and infant carried only the rapid meta- such as cell motility and with wildtype alleles who did not smoke bolism ADH1C variant (OR of 0.9, migration [Brauer and Tierney, 2004]. [NOS3 OR ¼ 5.2 (95%CI: 2.4–11.4); 95%CI: 0.2–4.1). Hobbs et al. [2010] Additionally, high level of homocysteine ICAM1 OR ¼ 5.2 (95%CI: 2.1–12.7); reported a possible gene–environment and its conversion to homocysteine– and NPPA OR ¼ 6.4 (95%CI: 2.8– interaction in infants with CHDs ex- thiolactone, have been associated with 14.6)] [Torfs et al., 2006]. posed in utero to alcohol. Women who post-translational modification of the consumed alcohol while pregnant and folate receptors. As a result, creation of were carriers of the TCII—776 CG or neoantigens might be inducing a patho- MATERNAL ALCOHOL GG genotype—were 1.7 times more logical maternal immune response that (ETHANOL) EXPOSURE likely to have a CHD-affected fetus, than ultimately will impair folic acid uptake were women who drank and carried a [Jakubowski et al., 2009]. In a recently Fetal alcohol (FAS) was CC genotype. published study, Blanton et al. [2011] initially described in the French medical analyzed 89 single nucleotide polymor- literature by Lemoine et al. [1968]. phisms (SNPs) in fourteen folate metab- Subsequently, Jones and collaborators MATERNAL olism-related genes in non-Hispanic were the first to systematically define the HYPERGLYCEMIA/ white and Hispanic NSCLP families. association between maternal alcohol GLUCOSE METABOLISM Evidence was documented for interac- abuse and the pattern of birth defects tions with smoking, and SNPs in the associated with the FAS [Jones et al., An extensive epidemiological literature FOLR1 and FOLR2 genes. 1973]. Maternal alcohol use has also indicates that maternal diabetes is a ARTICLE AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) 219 strong risk factor for birth defects transcriptional programming in devel- and perinatal mortality [Mills et al., There is sufficient evidence to oping embryos by down-regulating a 1979; Myrianthopoulos and Melnick, suggest that compromised large group of transcriptional factors 1987; Reece and Homko, 1993; Waller [Pavlinkova et al., 2009; Salbaum and et al., 1994; Shaw et al., 1996, 2000; maternal glucose control Kappen, 2010]. Werler et al., 1996; Hendricks et al., predisposes embryos to Studies have shown that the in- 2001; Eriksson et al., 2003; Honein creased level of oxygen free radicals in et al., 2003; Watkins et al., 2003; developmental anomalies, embryos of diabetic rats are contributing Anderson et al., 2005]. Maternal meta- even among non-diabetic to the observed malformations, and that bolic syndrome, obesity and pre-preg- women. Maternal diabetes, the removal of these reactive oxygen nant weight are also associated with risk species reduces the likelihood of fetuses of central nervous system birth defects obesity, and metabolic expressing the diabetic embryopathy [Waller et al., 1994; Shaw et al., 1996; syndrome create a compromised [Eriksson and Borg, 1991; Hagay et al., Watkins et al., 1996; Werler et al., 1996; 1995; Eriksson and Siman, 1996; Sivan Kallen, 1998; Hendricks et al., 2001; in utero environment for et al., 1996; Wentzel et al., 1997; Fine Ray et al., 2007; Correa et al., 2008] as the developing embryo. The et al., 1999]. Increased glucose delivery well as other birth defects such as CHDs, to embryos, or activation of pathways omphalocele and instances where the genetic basis underlying the that are stimulated by high glucose such child presents with multiple anomalies susceptibility to abnormal as the hexosamine biosynthetic pathway [Watkinsand Botto, 2001; Watkins et al., glucose metabolism induced or hypoxia, increase oxidative stress in 2003]. Gestational diabetes mellitus embryos. Blocking these pathways, or (GDM) may also be associated with birth defects involves both providing antioxidants such as reduced increased risk of central nervous system maternal and embryonic glutathione or vitamin E, suppress birth defects [Anderson et al., 2005]; the adverse effects of excess glucose however, the increased risk may in fact gene variants. [Loeken, 2005, 2006]. Impaired embry- be due to undiagnosed type 2 diabetes. onic gene expression resulting from Therefore, GDM has not been recog- oxidative stress and consequent apopto- nized as a risk factor due to inconsistent Early human embryos do not sis or disturbed organogenesis, may findings [Janssen et al., 1996; Savona- have pancreatic function until the be a general mechanism to explain Ventura and Gatt, 2004; Mitanchez, development of b-cells, usually after the features observed in the diabetic 2010]. In vivo and in vitro studies using gestational week 7. Maternal hypergly- embryopathy [Kambe et al., 1996; Wu animal models suggested that hyper- cemia forces an increased glucose flux et al., 1996; Phelan et al., 1997; Fine glycemia is a major teratogen capable into embryonic cells through glucose et al., 1999; Pani et al., 2002a,b; King of inducing malformations in diabetic transporters during early organogenesis; and Loeken, 2004; Loeken, 2006; animal models, while other associated therefore, the embryonic cells suffer Dheen et al., 2009]. For example, factors such as hyperinsulinemia [Hen- from an increased metabolic overload oxidative stress induced by excess em- dricks et al., 2001], ketone bodies, which is directed at their mitochondria, bryonic glucose metabolism inhibits the branched amino acids, and triglycerides leading to increased formation of reac- expression of Pax3, a critical develop- may also exert some adverse effects on tive oxygen and oxidative stress [Eriks- mental gene, leading to the appearance the developing embryos [Styrud and son et al., 2000; Wentzel et al., 2001; of birth defects [Morgan et al., 2008; Eriksson, 1992; Suzuki et al., 1996; Beemster et al., 2002]. Hyperglycemia Zabihi and Loeken, 2010]. Reece et al., 1996a; Eriksson et al., causes a myo-inositol deficiency with a PAX3 gene encodes a transcription 2000; Zhao and Reece, 2005]. There competitive inhibition of ambient glu- factor required for normal neural tube is sufficient evidence to suggest that cose, which might have been associated closure. Murine embryos with a non- compromised maternal glucose control with a diminished phosphoinositide functional Pax3 gene exhibit open predisposes embryos to developmental signal transduction. Dietary supplemen- NTDs, exencephaly and spina bifida, anomalies, even among non-diabetic tation of deficient substrates for example, with 100% penetrance [Pani et al., women [Groenen et al., 2003; Shaw arachidonic acid or myo-inositol [Gold- 2002a; Wlodarczyk et al., 2006b; Burren et al., 2003]. Maternal diabetes, obesity, man et al., 1985; Reece et al., 1988, et al., 2008]. PAX3 protein is required and metabolic syndrome create a com- 1996b; Baker et al., 1990; Khandelwal during neural tube development to promised in utero environment for the et al., 1998], either in vitro or in vivo, has suppress p53-dependent cell death and developing embryo. The genetic basis been shown to reduce the incidence of consequent failure to close the neural underlying the susceptibility to abnor- diabetes-related malformations in the tube. PAX3 decreases steady-state levels mal glucose metabolism induced birth offspring of diabetic pregnant animals. of the p53 tumor-suppressor protein, defects involves both maternal and Gene expression profiling studies such that when PAX3 is deficient, p53 embryonic gene variants. showed that maternal diabetes alters protein increases, leading to increased 220 AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) ARTICLE neuroepithelial which may including: intrauterine growth retarda- About 3–10% of these infants present occur prior to the completion of neural tion (IUGR), caudal regression, anen- with birth defects, and approximately tube closure. Supplementation with cephaly with absence of the head, 11–20% of them exhibit neurodevelop- folic acid or 5-methyltetrahydrofolate microphthalmia, and micrognathia, mental impairment, with or without rescues the normal phenotype in spon- which are similar to those defects associated structural abnormalities [Fin- taneous Pax3 mutant (Sp/Sp) embryos observed in the diabetic embryopathy nell, 1991; Kaneko et al., 1999; Dean [Wlodarczyk et al., 2006b]. A recent [Heilig et al., 2003]. A recent study et al., 2002; Mortensen et al., 2003; mouse study suggested the presence of a reported that one silent variant, Pro196, Adab et al., 2004; Gaily et al., 2004; gene–environment interaction between may be associated with risk of spina Vinten et al., 2005]. Intrauterine expo- embryonic folate levels and the Pax3 bifida by affecting the transcription and sure to valproate or to multiple AEDs genotype. Folate deficiency does not spicing of the SLC2A1 (alias: GLUT1) seems to represent the highest malfor- cause NTDs in wildtype mice, but gene [Davidson et al., 2008]. mation risk [Breen and Davenport, causes a significant increase in cranial A recent report suggested that 2006; Pennell, 2006; Veiby et al., NTDs among mutant embryos that GLUT2 may be a risk factor involved 2009]. Irrespective of which AED was carry an intragenic deletion in the Pax3 in NTD occurrence in diabetic preg- studied, the results consistently showed gene (Sp2H). Control treatments, in nancies [Li et al., 2007]. In animal that not all exposed fetuses were born which intermediate levels of folate are models, maternal diabetes and hypergly- with birth defects [German et al., 1970; supplied, failed to induce NTDs, sug- cemia do not down-regulate Glut1 and Jones et al., 1989; Dansky and Finnell, gesting that NTD risk is related to Glut2 expression in the embryos and 1991]. These observations indicate that embryonic folate concentration, not visceral yolk sac during the critical in most instances, AEDs alone are maternal blood folate concentration. periods of organogenesis, as they do in insufficient to induce teratogenic effects This study suggested that folate deficien- pre-implantation embryos; therefore, in exposed fetuses. The observed birth cy increases the risk of NTDs in embryonic glucose transport continues defects are multifactorial in their origin genetically predisposed Splotch embry- and results in increased glucose flux into and beside exposure to AEDs, some os, probably via embryonic growth the cells [Maeda et al., 1993; Takao et al., other interacting components, most retardation [Burren et al., 2008]. Var- 1993; Trocino et al., 1994; Li et al., likely genetic, must be involved in order iants in human PAX3 gene potentially 2007]. Inactivation of Glut2 gene in a to predispose the developing embryo to associated with risk of spina bifida have mouse strain protects embryos from express an abnormal phenotype follow- been suggested [Lu et al., 2007]; there- maternal diabetes-induced NTDs [Li ing the in utero AED exposure. It seems fore, interaction between these variants et al., 2007]. The interactions between and environmental factors including human SLC2A2 (alias: GLUT2) and folic acid use and maternal hyperglyce- maternal glucose metabolism; however, mia merit evaluation. have not yet been evaluated. Intrauterine exposure to Glucose is transported across the valproate or to multiple AEDs lipid bi-layers of cell membranes by a EXPOSURE TO family of structurally related membrane ANTIEPILEPTIC DRUGS seems to represent the highest spanning glycoproteins called glucose transporters [Joost and Thorens, 2001; Epilepsy is the most common neuro- malformation risk. Irrespective Joost et al., 2002]. Erythrocyte-type logic disorder requiring medical treat- of which AED was studied, the glucose transporter (GLUT1) is the ment in pregnant women [Hvas et al., results consistently showed that major type of glucose transporter 2000]. Antiepileptic drugs (AEDs) have expressed during early organogenesis long been recognized as having a not all exposed fetuses were [Maeda et al., 1993; Matsumoto et al., teratogenic potential capable of disrupt- born with birth defects. These 1995]. Consistent with the high demand ing normal development in exposed for glucose, GLUT1 protein is expressed infants. Several epidemiological studies observations indicate that in at a high level in the embryonic tissues indicated that infants of mothers treated most instances, AEDs alone undergoing differentiation, mainly in with AEDs during pregnancy have a are insufficient to induce the neural tube, while expression de- two- to threefold higher risk of having clined as gestational age progressed birth defects when compared to the teratogenic effects in exposed [Shepard et al., 1970; Tanimura and general population [Holmes et al., 2001; fetuses. The observed birth Shepard, 1970; Akazawa et al., 1989; Perucca, 2005]. In the USA annually Matsumoto et al., 1995]. Homozygous almost 25,000 children are born to defects are multifactorial Glut1 knockout mice die in utero women with epilepsy and most of them in their origin and beside [Ohtsuki et al., 2006]. Glut1-deficient require AEDs such as valproic acid, mice generated by antisense-Glut1 carbamazepine, , phenobarbi- exposure to AEDs, some other exhibit developmental malformations tal, or lamotrigine [Meador et al., 2008]. interacting components, most ARTICLE AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) 221 likely genetic, must be involved pine, comparing to a control group of slight strain dependent variation in healthy children [Vurucu et al., 2008]. timing of neural tube closure, as evi- in order to predispose the Based on this finding, one can speculate denced by the study which failed to developing embryo to express an that VPA lowers an already limited produce at least moderately high level supply of folate in pregnant MTHFR of NTDs in C57 mice treated at abnormal phenotype following 677TT homozygous women, pushing various timepoints during pregnancy the in utero AED exposure. the developing embryo beyond the [Beck, 1999]. The observed strain spe- threshold of normal development and cific sensitivity to VPA was also con- ultimately leading to NTDs. Insight firmed in an in vitro study where 6–8 from the animal studies demonstrating somite stage mouse embryos of two that either the mother or the embryo that the teratogenicity of VPA can be different strains; SWV and C57BL/6J, carry genetic factors determining sus- prevented/diminished by folic acid sup- were cultured in the serum supple- ceptibility to AED induced adverse plementation tend to provide biological mented with VPA for 48 hr. As in the effects. Epidemiological study showed plausibility to this hypothesis [Padma- in vivo studies, the SWV embryos were that the recurrence risk of NTDs among nabhan and Shafiullah, 2003; Dawson more likely to have open NTDs than pregnant women who were exposed to et al., 2006]. Unfortunately, there is no were the C57BL/6J embryos. Addition- VPAwas higher than the occurrence risk concrete evidence that supplemental ally, the minimum lethal dose of VPA of NTD in women exposed to VPA, folic acid protects against NTDs in was also significantly lower for SWV suggesting possible genetic contribu- [Ornoy, 2009]. On the other cultured embryos [Naruse et al., 1988]. tions to VPA sensitivity [Dansky and hand, rather surprising results were Results of this study point out that Finnell, 1991]. Other studies concern- reported from studies on Mthfr knockout besides variation in the maternal genes þ/þ ing NTDs demonstrated that these mice treated with VPA. Mthfr wild- that interact with environmental factors, defects are more common if there was type mice turned out to be more the embryonic genetic component þ/ an existing family history of NTDs sensitive than Mthfr heterozygotes alone can modulate the teratogenic [Delgado-Escueta and Janz, 1992; Malm when treated with VPA. There were no outcome. et al., 2002]. cases of exencephaly in the fetuses from Complex birth defects are known þ/ Methylenetetrahydrofolate reduc- Mthfr dams, whereas 21% of fetuses to have multifactorial etiologies and the þ/þ tase (MTHFR) is an enzyme that from Mthfr had NTDs. Further combination of genetic susceptibility as plays a key role in folate metabolism. analysis revealed that VPA lowered well as the dose and the time of exposure It catalyzes the conversion of 5,10- homocysteine plasma level and increased to certain environmental compounds methylenetetrahydrofolate (5,10-MTHF) the Mthfr expression which resulted in (e.g., drugs and other toxic xenobiotics) to 5-methyltetrahydrofolate (5-MTHF), lower teratogenicity in Mthfr deficient appear to account for significant varia- which is the main methyl donor in the mice [Roy et al., 2008]. Much of our tion in the human response. Both homocysteine to methionine remethy- understanding of how genetic factors maternal and fetal genotypes can affect lation cycle. Studies of MTHFR in modulate teratogenic action of drugs or placental transport, absorption, metabo- human revealed a common polymor- other chemicals comes from studies on lism, distribution, and receptor binding phism (677C!T substitution) in this animal models. Results of VPA terato- of the drug, influencing its teratogenici- gene that is associated with hyperhomo- genicity studies in mice clearly demon- ty [Hall et al., 1997; Spiegelstein et al., cysteinemia and a slightly increased risk strated a strong genetic component 2003]. Polymorphisms in maternal and for birth defects [Botto and Yang, 2000]. involved in determining sensitivity to fetal genes involved in xenobiotic de- Epileptic mothers taking AEDs who the induction of NTDs. Different strains toxification may modify risks of adverse were MTHFR 677T homozygotes were of mice reacted very differently to VPA pregnancy outcome in response to found to have a three- to fourfold higher treatment during pregnancy as evi- maternal use of AED. Many drugs and risk for having a child with the fetal denced by the presence of fetuses with endogenous compounds need to be anticonvulsant syndrome, compared exencephaly, a form of NTD equivalent metabolized in order to be activated or with 677CC homozygous mothers re- of human anencephaly [Finnell, 1991; inactivated and ultimately excreted. The ceiving AED treatment [Deen et al., Hall et al., 1997; Malm et al., 2002]. enzymes responsible for these reactions 1999; Kini et al., 2007; Dean et al., More specifically SWV/Fnn, AKR/J, belong to the extensive cytochrome 2008]. This syndrome consists of a C3H/HeJ, and P/J mice turned out to P450 (CYPs) gene family. The human characteristic pattern of neurodevelop- be the most sensitive; LM/Bc had an genome contains 115 cytochrome P450 mental impairment, facial features, and intermediate sensitivity, while DBA and genes, of which at least 57 are functional. major birth defects including NTDs, C57BL/6J were highly resistant [Azar- Among CYP families 1–3 (responsible orofacial clefts, heart, and renal defects. bayjani and Danielsson, 1998; Faiella for drug metabolism), there are 22 dif- It appears that there are significantly et al., 2000; Spiegelstein et al., 2003; ferent P450 isoforms and a high degree lower plasma folate concentrations in Wiltse, 2005]. These differences in of polymorphism has been identified children taking VPA and carbamaze- sensitivity cannot be explained by the [Ingelman-Sundberg and Sim, 2010; 222 AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) ARTICLE

Johansson and Ingelman-Sundberg, them were born without characteristic REFERENCES 2010]. Almost all of the P450 metabo- FSH features. This study indicated that lizing genes are polymorphic, and an adverse pregnancy outcome can be Adab N, Kini U, Vinten J, Ayres J, Baker G, Clayton-Smith J, Coyle H, Fryer A, Gorry J, the known allelic CYP variants are too predicted on the basis of an enzymatic Gregg J, Mawer G, Nicolaides P, Pickering extensive to describe here (see web biomarker. More studies are needed to L, Tunnicliffe L, Chadwick DW. 2004. The page http://www.cypalleles.ki.se/). This decipher all the genes engaged in drugs longer term outcome of children born to mothers with epilepsy. J Neurol Neurosurg polymorphism translates into pheno- metabolism and their genetic variants. Psychiatry 75:1575–1583. types that differ in the ability and/or This knowledge would be very helpful Akazawa M, Akazawa S, Hashimoto M, Akashi speed of drug metabolization, from ‘‘low for new drug development and for M, Yamazaki H, Tahara D, Yamamoto H, Yamaguchi Y, Nakanishi T, Nagataki S. metabolizers’’ having defective alleles clinicians in order to prescribe the right 1989. Effects of brief exposure to insulin- and inactive enzymes, to ‘‘ultrarapid medicine, compatible with the specific induced hypoglycemic serum during orga- metabolizers’’ carrying two or more patient genotype. nogenesis in rat embryo culture. Diabetes 38:1573–1578. active gene copies. Pregnant women Anderson JL, Waller DK, Canfield MA, Shaw who are so called ‘‘slow metabolizers’’ GM, Watkins ML, Werler MM. 2005. may be at increased risk of having an Maternal obesity, gestational diabetes, and CONCLUSIONS central nervous system birth defects. Epi- affected baby due to the drug/teratogen demiology 16:87–92. accumulation and slower excretion Experimental developmental Au KS, Ashley-Koch A, Northrup H. 2010. rate that extends the time when the studies repeatedly demonstrate the Epidemiologic and genetic aspects of spina bifida and other neural tube defects. Dev developing embryo is exposed to that unquestioned importance of genetic Disabil Res Rev 16:6–15. toxicant. Conversely, pregnant women variation in determining risks to tera- Azarbayjani F, Danielsson BR. 1998. Pharmaco- who are ‘‘fast metabolizers’’ can more togen-induced developmental abnor- logically induced embryonic dysrhythmia and episodes of hypoxia followed by reox- quickly activate some drugs, including malities. As described earlier, inbred ygenation: A common teratogenic mecha- proteratogens, resulting in higher strains of mice, representing different nism for antiepileptic drugs? Teratology plasma concentration of reactive inter- mouse genomes, show significant varia- 57:117–126. Bailey LB, Berry RJ. 2005. Folic acid supple- mediates or active teratogens which tion in sensitivity to teratogenic phar- mentation and the occurrence of congenital ultimately can also adversely affect the maceutical or xenobiotic agents. heart defects, orofacial clefts, multiple births, embryo. Clinical research in this area; however, and miscarriage. Am J Clin Nutr 81:1213S– 1217S. Two cases of intoxication with remains quite limited. Although we Baker L, Piddington R, Goldman A, Egler J, an AED drug—phenytoin, showing are still far away from individualized Moehring J. 1990. Myo-inositol and pros- women with significant neurological preventive measures for birth defects, taglandins reverse the glucose inhibition of neural tube fusion in cultured mouse symptoms, have been published. In understanding the genetic basis of the embryos. Diabetologia 33:593–596. both cases, a very slow clearance susceptibility to specific environmental Beck SL. 1999. Contributions of dam and (fourteen times slower than average) factor-induced birth defects may yield conceptus to differences in sensitivity to valproic acid among C57 black and SWV from blood, caused by mutations in critical clues that will ultimately to mice. Reprod Toxicol 13:353–360. CYP2C9 (the main enzyme responsible novel approaches that can be imple- Beemster P, Groenen P, Steegers-Theunissen R. for phenytoin metabolism) was respon- mented to prevent, preventable birth 2002. Involvement of inositol in reproduc- tion. Nutr Rev 60:80–87. sible [Brandolese et al., 2001; Kidd et al., defects. Pharmacogenomics is an Blanton SH, Henry RR, Yuan Q, Mulliken JB, 2001]. A very elegant study by Buehler emerging science aiming to identify Stal S, Finnell RH, Hecht JT. 2011. Folate et al. [1990] involved monitoring genetic polymorphisms that affect drug pathway and nonsyndromic cleft lip and palate. Birth Defects Res A Clin Mol Teratol 19 exposed to phenytoin efficacy and safety. Genome-wide asso- 91:50–60. monotherapy.In the amniocytes collect- ciation studies will screen numerous Blencowe H, Cousens S, Modell B, Lawn J. ed from these pregnancies at mid- genetic markers without a prior knowl- 2010. Folic acid to reduce neonatal mortal- ity from neural tube disorders. Int J gestation, they measured the activity of edge. Whole genome sequencing is the Epidemiol 3:i110–i121. epoxide hydrolase, an enzyme involved ultimate way to identify novel functional Blom HJ, Shaw GM, den Heijer M, Finnell RH. in phenytoin metabolism. If the enzyme mutations, especially the rare ones that 2006. Neural tube defects and folate: Case far from closed. Nat Rev Neurosci 7:724– activity was low (less than 30% of the may modify individual’s susceptibility to 731. standard), they predicted that the em- teratogens. Together with pharmaco- Botto LD, Yang Q. 2000. 5,10-Methylenetetra- bryo would be at higher risk of having and toxicogenomics research, these hydrofolate reductase gene variants and congenital anomalies: A HuGE review. Am the fetal hydantoin syndrome. In four genome-wide high throughput/high J Epidemiol 151:862–877. cases, significantly lower activity of information content assays will provide Botto LD, Mulinare J, Erickson JD. 2003b. Do epoxide hydrolase was found, and after essential information allowing physi- multivitamin or folic acid supplements reduce the risk for congenital heart defects? delivery, all four infants presented with cians to choose the ‘‘right’’ drug at the Evidence and gaps. Am J Med Genet Part A clinical symptoms of fetal hydantoin ‘‘right’’ dose for each patient in order 121A:95–101. syndrome (FSH). The remaining 15 to avoid any adverse drug reaction and Boyles AL, DeRoo LA, Lie RT, Taylor JA, Jugessur A, Murray JC, Wilcox AJ. 2010. fetuses with higher enzyme activity ultimately to make the personalized Maternal alcohol consumption, alcohol were not considered at risk, and all of medication possible. metabolism genes, and the risk of oral clefts: ARTICLE AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) 223

A population-based case–control study in defects in CD-1 mice. Toxicol Appl Phar- Gaily E, Kantola-Sorsa E, Hiilesmaa V, Isoaho M, Norway, 1996–2001. Am J Epidemiol macol 211:124–132. Matila R, Kotila M, Nylund T, Bardy A, 172:924–931. Dean JC, Hailey H, Moore SJ, Lloyd DJ, Turn- Kaaja E, Granstrom ML. 2004. Normal Brandolese R, Scordo MG, Spina E, Gusella M, penny PD, Little J. 2002. Long term health intelligence in children with prenatal expo- Padrini R. 2001. Severe phenytoin intox- and neurodevelopment in children exposed sure to carbamazepine. Neurology 62: ication in a subject homozygous for to antiepileptic drugs before birth. J Med 28–32. CYP2C9*3. Clin Pharmacol Ther 70: Genet 39:251–259. German J, Ehlers KH, Kowal A, De George FV, 391–394. Dean JC, Robertson Z, Reid V, Wang Q, Hailey Engle MA, Passarge E. 1970. Possible Brauer PR, Tierney BJ. 2004. Consequences of H, Moore S, Rasalam AD, Turnpenny P, teratogenicity of trimethadione and para- elevated homocysteine during embryonic Lloyd D, Shaw D, Little J. 2008. A high methadione. Lancet 2:261–262. development and possible modes of action. frequency of the MTHFR 677C>T poly- Goldman AS, Baker L, Piddington R, Marx B, Curr Pharm Des 10:2719–2732. morphism in Scottish women with epilepsy: Herold R, Egler J. 1985. Hyperglycemia- Breen DP, Davenport RJ. 2006. Teratogenicity Possible role in pathogenesis. Seizure 17: induced teratogenesis is mediated by a of antiepileptic drugs. Br Med J 333:615– 269–275. functional deficiency of arachidonic acid. 616. Deen JL, Vos T, Huttly SR, Tulloch J. 1999. Proc Natl Acad Sci USA 82:8227–8231. Brender JD, Suarez L. 1990. Paternal occupation Injuries and noncommunicable diseases: Graham JMJr, Shaw GM. 2005. Gene-environ- and anencephaly. Am J Epidemiol 131:517– Emerging health problems of children in ment interactions in rare diseases that 521. developing countries. Bull World Health include common birth defects. Birth Brender J, Suarez L, Hendricks K, Baetz RA, Organ 77:518–524. Defects Res A Clin Mol Teratol 73:865– Larsen R. 2002. Parental occupation and Delgado-Escueta AV, Janz D. 1992. Consensus 867. neural tube defect-affected pregnancies guidelines: Preconception counseling, Grewal J, Carmichael SL, Ma C, Lammer EJ, among Mexican Americans. J Occup Envi- management, and care of the pregnant Shaw GM. 2008. Maternal periconceptional ron Med 44:650–656. woman with epilepsy. Neurology 42:149– smoking and alcohol consumption and Buehler BA, Delimont D, van Waes M, Finnell 160. risk for select congenital anomalies. Birth RH. 1990. Prenatal prediction of risk of the Dheen ST, Tay SS, Boran J, Ting LW, Kumar SD, Defects Res A Clin Mol Teratol 82:519– fetal hydantoin syndrome. N Engl J Med Fu J, Ling EA. 2009. Recent studies on 526. 322:1567–1572. neural tube defects in embryos of diabetic Groenen PM, Peer PG, Wevers RA, Swinkels Burren KA, Savery D, Massa V, Kok RM, Scott pregnancy: An overview. Curr Med Chem DW, Franke B, Mariman EC, Steegers- JM, Blom HJ, Copp AJ, Greene ND. 2008. 16:2345–2354. Theunissen RP. 2003. Maternal myo-inosi- Gene-environment interactions in the cau- Eriksson UJ, Borg LA. 1991. Protection by free tol, glucose, and status is associated with sation of neural tube defects: Folate defi- oxygen radical scavenging enzymes against the risk of offspring with spina bifida. Am J ciency increases susceptibility conferred by glucose-induced embryonic malformations Obstet Gynecol 189:1713–1719. loss of Pax3 function. Hum Mol Genet in vitro. Diabetologia 34:325–331. Hagay ZJ, Weiss Y, Zusman I, Peled-Kamar M, 17:3675–3685. Eriksson UJ, Siman CM. 1996. Pregnant diabetic Reece EA, Eriksson UJ, Groner Y. 1995. Campbell LR, Dayton DH, Sohal GS. 1986. rats fed the antioxidant butylated hydrox- Prevention of diabetes-associated embryop- Neural tube defects: A review of human and ytoluene show decreased occurrence of athy by overexpression of the free radical animal studies on the etiology of neural tube malformations in offspring. Diabetes 45: scavenger copper zinc superoxide dismutase defects. Teratology 34:171–187. 1497–1502. in transgenic mouse embryos. Am J Obstet Carmichael SL, Shaw GM, Selvin S, Schaffer DM. Eriksson UJ, Borg LA, Cederberg J, Nordstrand Gynecol 173:1036–1041. 2003. Diet quality and risk of neural tube H, Siman CM, Wentzel C, Wentzel P. 2000. Hall JL, Harris MJ, Juriloff DM. 1997. Effect defects. Med Hypotheses 60:351–355. Pathogenesis of diabetes-induced congenital of multifactorial genetic liability to exence- Chabra S, Gleason CA, Seidel K, Williams MA. malformations. Ups J Med Sci 105:53–84. phaly on the teratogenic effect of 2011. Rising prevalence of gastroschisis in Eriksson UJ, Cederberg J, Wentzel P. 2003. valproic acid in mice. Teratology 55:306– Washington State. J Toxicol Environ Health Congenital malformations in offspring of 313. A 74:336–345. diabetic mothers—Animal and human stud- Harris MJ, Juriloff DM. 2010. An update to the Collins JS, Atkinson KK, Dean JH, Best RG, ies. Rev Endocr Metab Disord 4:79–93. list of mouse with neural tube Stevenson RE. 2011. Long term mainte- Faiella A, Wernig M, Consalez GG, Hostick U, closure defects and advances toward a nance of neural tube defects prevention in a Hofmann C, Hustert E, Boncinelli E, complete genetic perspective of neural tube high prevalence state. J Pediatr (Epub). Balling R, Nadeau JH. 2000. A mouse closure. Birth Defects Res A Clin Mol Correa M, Manrique HM, Font L, Escrig MA, model for valproate teratogenicity: Parental Teratol 88:653–669. Aragon CM. 2008. Reduction in the effects, homeotic transformations, and Heilig CW, Saunders T, Brosius FCIII, Moley K, anxiolytic effects of ethanol by centrally altered HOX expression. Hum Mol Genet Heilig K, Baggs R, Guo L, Conner D. formed acetaldehyde: The role of catalase 9:227–236. 2003. Glucose transporter-1-deficient mice inhibitors and acetaldehyde-sequestering Feldkamp ML, Alder SC, Carey JC. 2008. A case exhibit impaired development and deform- agents. Psychopharmacology (Berl) control population-based study investigating ities that are similar to diabetic embryopathy. 200:455–464. smoking as a risk factor for gastroschisis in Proc Natl Acad Sci USA 100:15613– Czeizel AE. 2000. Primary prevention of neural- Utah, 1997–2005. Birth Defects Res A Clin 15618. tube defects and some other major congen- Mol Teratol 82:768–775. Hendricks KA, Nuno OM, Suarez L, Larsen R. ital abnormalities: Recommendations for Fine EL, Horal M, Chang TI, Fortin G, Loeken 2001. Effects of hyperinsulinemia and the appropriate use of folic acid during MR. 1999. Evidence that elevated glucose obesity on risk of neural tube defects among pregnancy. Paediatr Drugs 2:437–449. causes altered gene expression, apoptosis, Mexican Americans. Epidemiology 12: Dansky LV, Finnell RH. 1991. Parental epilepsy, and neural tube defects in a mouse model 630–635. anticonvulsant drugs, and reproductive out- of diabetic pregnancy. Diabetes 48:2454– Hobbs CA, James SJ, Jernigan S, Melnyk S, Lu Y, come: Epidemiologic and experimental 2462. Malik S, Cleves MA. 2006. Congenital heart findings spanning three decades; 2: Human Finnell RH. 1991. Genetic differences in suscept- defects, maternal homocysteine, smoking, studies. Reprod Toxicol 5:301–335. ibility to anticonvulsant drug-induced and the 677 C>T polymorphism in the Davidson CM, Northrup H, King TM, Fletcher developmental defects. Pharmacol Toxicol methylenetetrahydrofolate reductase gene: JM, Townsend I, Tyerman GH, Au KS. 69:223–227. Evaluating gene–environment interactions. 2008. Genes in glucose metabolism and Finnell RH, Shaw GM, Lammer EJ, Brandl KL, Am J Obstet Gynecol 194:218–224. association with spina bifida. Reprod Sci Carmichael SL, Rosenquist TH. 2004. Hobbs CA, Cleves MA, Karim MA, Zhao W, 15:51–58. Gene-nutrient interactions: Importance of MacLeod SL. 2010. Maternal folate-related Dawson JE, Raymond AM, Winn LM. 2006. folates and retinoids during early embryo- gene environment interactions and congen- Folic acid and pantothenic acid protection genesis. Toxicol Appl Pharmacol 198: ital heart defects. Obstet Gynecol 116:316– against valproic acid-induced neural tube 75–85. 322. 224 AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) ARTICLE

Holmes LB, Harvey EA, Coull BA, Huntington thyrotropin in rat thyroid FRTL-5cells. Mol Lorente C, Cordier S, Goujard J, Ayme S, Bianchi KB, Khoshbin S, Hayes AM, Ryan LM. Endocrinol 10:801–812. F, Calzolari E, De Walle HE, Knill-Jones R. 2001. The teratogenicity of anticonvulsant Kaneko S, Battino D, Andermann E, Wada K, Kan 2000. Tobacco and alcohol use during drugs. N Engl J Med 344:1132–1138. R, Takeda A, Nakane Y, Ogawa Y, Avanzini pregnancy and risk of oral clefts. Occupa- Honein MA, Moore CA, Watkins ML. 2003. G, Fumarola C, Granata T, Molteni F, Pardi tional Exposure and Congenital Malforma- Subfertility and prepregnancy overweight/ G, Minotti L, Canger R, Dansky L, Oguni tion Working Group. Am J Public Health obesity: Possible interaction between these M, Lopes-Cendas I, Sherwin A, Ander- 90:415–419. risk factors in the etiology of congenital mann F, Seni MH, Okada M, Teranishi T. Lu W, Zhu H, Wen S, Laurent C, Shaw GM, renal anomalies. Birth Defects Res A Clin 1999. Congenital malformations due to Lammer EJ, Finnell RH. 2007. Screening Mol Teratol 67:572–577. antiepileptic drugs. Epilepsy Res 33:145–158. for novel PAX3 polymorphisms and risks of Hvas CL, Henriksen TB, Østergaard JR. 2000. Khandelwal M, Reece EA, Wu YK, Borenstein spina bifida. Birth Defects Res A Clin Mol Birth weight in offspring of women with M. 1998. Dietary myo-inositol therapy Teratol 79:45–49. epilepsy. Epidemiol Rev 22:275–282. in hyperglycemia-induced embryopathy. Lundberg YW, Wing MJ, Xiong W, Zhao J, Hwang SJ, Beaty TH, Panny SR, Street NA, Teratology 57:79–84. Finnell RH. 2003. Genetic dissection of Joseph JM, Gordon S, McIntosh I, Franco- Kidd RS, Curry TB, Gallagher S, Edeki T, hyperthermia-induced neural tube defects mano CA. 1995. Association study of trans- Blaisdell J, Goldstein JA. 2001. Identifica- in mice. Birth Defects Res A Clin Mol forming growth factor alpha (TGF alpha) tion of a null allele of CYP2C9 in an Teratol 67:409–413. TaqI polymorphism and oral clefts: Indica- African-American exhibiting to Maeda Y, Akazawa S, Akazawa M, Takao Y, tion of gene–environment interaction in a phenytoin. Pharmacogenetics 11:803–808. Trocino RA, Takino H, Kawasaki E, Yokota population-based sample of infants with King GL, Loeken MR. 2004. Hyperglycemia- A, Okuno S, Nagataki S. 1993. Glucose birth defects. Am J Epidemiol 141:629– induced oxidative stress in diabetic compli- transporter gene expression in rat conceptus 636. cations. Histochem Cell Biol 122:333–338. during early organogenesis and exposure to Ingelman-Sundberg M, Sim SC. 2010. Intronic Kini U, Lee R, Jones A, Smith S, Ramsden S, insulin-induced hypoglycemic serum. Acta polymorphisms of cytochromes P450. Hum Fryer A, Clayton-Smith J. 2007. Influence Diabetol 30:73–78. 4:402–405. of the MTHFR genotype on the rate of Malik S, Cleves MA, Honein MA, Romitti PA, Jakubowski H, Perla-Kajan J, Finnell RH, malformations following exposure to anti- Botto LD, Yang S, Hobbs CA. 2008. Cabrera RM, Wang H, Gupta S, Kruger epileptic drugs in utero. Eur J Med Genet Maternal smoking and congenital heart WD, Kraus JP, Shih DM. 2009. Genetic 50:411–420. defects. Pediatrics 121:e810–e816. or nutritional disorders in homocysteine Kuciene R, Dulskiene V. 2010. Parental cigarette Malm H, Kajantie E, Kivirikko S, Kaariainen H, or folate metabolism increase protein smoking and the risk of congenital heart Peippo M, Somer M. 2002. Valproate N-homocysteinylation in mice. FASEB J septal defects. Medicina (Kaunas) 46:635– embryopathy in three sets of siblings: 23:1721–1727. 641. Further proof of hereditary susceptibility. Janssen PA, Rothman I, Schwartz SM. 1996. Kumar V, Bustin SA, McKay IA. 1995. Trans- Neurology 59:630–633. Congenital malformations in newborns of forming growth factor alpha. Cell Biol Int Matsumoto K, Akazawa S, Ishibashi M, Trocino women with established and gestational 19:373–388. RA, Matsuo H, Yamasaki H, Yamaguchi Y, diabetes in Washington State, 1984–91. Lammer EJ, Iovannisci DM, Tom L, Schultz K, Nagamatsu S, Nagataki S. 1995. Abundant Paediatr Perinat Epidemiol 10:52–63. Shaw GM. 2008. Gastroschisis: A gene– expression of GLUT1 and GLUT3 in rat Jauniaux E, Burton GJ. 2007. Morphological and environment model involving the VEGF- embryo during the early organogenesis biological effects of maternal exposure to NOS3 pathway. Am J Med Genet Part C period. Biochem Biophys Res Commun tobacco smoke on the feto-placental unit. 148C:213–218. 209:95–102. Early Hum Dev 83:699–706. Lemoine P, Harousseau H, Borteyru JP, Menuet Meador KJ, Pennell PB, Harden CL, Gordon JC, Johansson I, Ingelman-Sundberg M. 2010. JC. 1968. Les enfants des parents alcohol- Tomson T, Kaplan PW, Holmes GL, French Genetic polymorphism and toxicology— iques: Anomalies observees a propos de 127 JA, Hauser WA, Wells PG, Cramer JA. With emphasis on cytochrome P450. Tox- cas (The children of alcoholic parents: HOPE Work Group. 2008. Pregnancy icol Sci 120:1–13. Anomalies observed in 127 cases). Quest registries in epilepsy: A consensus statement Jones KL, Smith DW, Ulleland CN, Streissguth P. Medical 25:476–482. on health outcomes. Neurology 71:1109– 1973. Pattern of malformation in offspring Li R, Thorens B, Loeken MR. 2007. Expression 1117. of chronic alcoholic mothers. Lancet 1: of the gene encoding the high-Km glucose Meeker JD, Missmer SA, Cramer DW, Hauser R. 1267–1271. transporter 2 by the early postimplantation 2007. Maternal exposure to second-hand Jones KL, Lacro RV, Johnson KA, Adams J. 1989. mouse embryo is essential for neural tube tobacco smoke and pregnancy outcome Pattern of malformations in the children of defects associated with diabetic embryop- among couples undergoing assisted repro- women treated with carbamazepine during athy. Diabetologia 50:682–689. duction. Hum Reprod 22:337–345. pregnancy. N Engl J Med 320:1661–1666. Li ZW,Liu JM, Ren AG, Zhang L, Guo ZY,Li Z. Mills JL, Baker L, Goldman AS. 1979. Malforma- Joost HG, Thorens B. 2001. The extended 2008. Maternal passive smoking and the risk tions in infants of diabetic mothers occur GLUT-family of sugar/polyol transport of neural tube defects: A case–control study before the seventh gestational week. Impli- facilitators: Nomenclature, sequence char- in Shanxi province, China. Zhonghua Liu cations for treatment. Diabetes 28:292– acteristics, and potential function of its novel Xing Bing Xue Za Zhi 29:417–420. 293. members (review). Mol Membr Biol 18: Lie RT, Wilcox AJ, Taylor J, Gjessing HK, Mitanchez D. 2010. Foetal and neonatal compli- 247–256. Saugstad OD, Aabyholm F, Vindenes H. cations in gestational diabetes: Perinatal Joost HG, Bell GI, Best JD, Birnbaum MJ, 2008. Maternal smoking and oral clefts: mortality, congenital malformations, macro- Charron MJ, Chen YT, Doege H, James The role of detoxification pathway genes. somia, shoulder dystocia, birth injuries, DE, Lodish HF, Moley KH, Moley JF, Epidemiology 19:606–615. neonatal complications. Diabetes Metab Mueckler M, Rogers S, Schurmann A, Little J, Cardy A, Arslan MT, Gilmour M, Mossey 36:617–627. Seino S, Thorens B. 2002. Nomenclature PA. 2004. Smoking and orofacial clefts: A Morgan SC, Relaix F, Sandell LL, Loeken MR. of the GLUT/SLC2A family of sugar/ United Kingdom-based case–control study. 2008. Oxidative stress during diabetic preg- polyol transport facilitators. Am J Physiol Cleft Palate Craniofac J 41:381–386. nancy disrupts cardiac neural crest migration Endocrinol Metab 282:E974–E976. Loeken MR. 2005. Current perspectives on the and causes outflow tract defects. Birth Kallen K. 1998. Maternal smoking, body mass causes of neural tube defects resulting from Defects Res A Clin Mol Teratol 82:453–463. index, and neural tube defects. Am J diabetic pregnancy. Am J Med Genet Part C Mortensen JT, Olsen J, Larsen H, Bendsen J, Obel Epidemiol 147:1103–1111. 135C:77–87. C, Sorensen HT. 2003. Psychomotor devel- Kambe F, Nomura Y, Okamoto T, Seo H. 1996. Loeken MR. 2006. Advances in understanding opment in children exposed in utero to Redox regulation of thyroid-transcription the molecular causes of diabetes-induced , antidepressants, neurolep- factors, Pax-8 and TTF-1, is involved in birth defects. J Soc Gynecol Investig 13: tics, and anti-epileptics. Eur J Epidemiol their increased DNA-binding activities by 2–10. 18:769–771. ARTICLE AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) 225

Murray JC. 2002. Gene/environment causes of Prescott NJ, Winter RM, Malcolm S. 2001. Shaw GM, Todoroff K, Schaffer DM, Selvin S. cleft lip and/or palate. Clin Genet 61:248– Nonsyndromic cleft lip and palate: Complex 2000. Maternal height and prepregnancy 256. genetics and environmental effects. Ann body mass index as risk factors for selected Myrianthopoulos NC, Melnick M. 1987. Studies Hum Genet 65:505–515. congenital anomalies. Paediatr Perinat in neural tube defects. I: Epidemiologic and Quadros EV, Regec AL, Khan KM, Quadros E, Epidemiol 14:234–239. etiologic aspects. Am J Med Genet 26:783– Rothenberg SP. 1999. Transcobalamin II Shaw GM, Quach T, Nelson V, Carmichael SL, 796. synthesized in the intestinal villi facilitates Schaffer DM, Selvin S, Yang W. 2003. Naruse I, Collins MD, Scott WJJr. 1988. Strain transfer of cobalamin to the portal blood. Neural tube defects associated with maternal differences in the teratogenicity induced Am J Physiol 277:G161–G166. periconceptional dietary intake of simple by sodium valproate in cultured mouse Ray JG, Thompson MD, Vermeulen MJ, Meier sugars and glycemic index. Am J Clin Nutr embryos. Teratology 38:87–96. C, Wyatt PR, Wong PY, Summers AM, 78:972–978. Obican SG, Finnell RH, Mills JL, Shaw GM, Farrell SA, Cole DE. 2007. Metabolic Shaw GM, Carmichael SL, Vollset SE, Yang W, Scialli AR. 2010. Folic acid in early syndrome features and risk of neural tube Finnell RH, Blom H, Midttun O, Ueland pregnancy: A public health success story. defects. BMC Pregnancy Childbirth 7:21. PM. 2009. Mid-pregnancy cotinine and FASEB J 24:4167–4174. Reece EA, Homko CJ. 1993. Diabetes-related risks of orofacial clefts and neural tube Ohtsuki S, Kikkawa T, Hori S, Terasaki T. 2006. complications of pregnancy. J Natl Med defects. J Pediatr 154:17–19. Modulation and compensation of the Assoc 85:537–545. Shea AK, Steiner M. 2008. Cigarette smoking mRNA expression of energy related trans- Reece EA, Gabrielli S, Abdalla M. 1988. The during pregnancy. Tob Res 10: porters in the brain of glucose transporter prevention of diabetes-associated birth 267–278. 1-deficient mice. Biol Pharm Bull 29:1587– defects. Semin Perinatol 12:292–301. Shepard TH, Tanimura T, Robkin MA. 1970. 1591. Reece EA, Wiznitzer A, Homko CJ, Hagay Z, Energy metabolism in early mammalian Ottman R. 1990. An epidemiologic approach Wu YK. 1996a. Synchronization of the embryos. Symp Soc Dev Biol 29:42–58. to gene–environment interaction. Genet factors critical for diabetic teratogenesis: An Shi M, Christensen K, Weinberg CR, Romitti P, Epidemiol 7:177–185. in vitro model. Am J Obstet Gynecol 174: Bathum L, Lozada A, Morris RW,Lovett M, Ou CY,Stevenson RE, Brown VK, Schwartz CE, 1284–1288. Murray JC. 2007. Orofacial cleft risk is Allen WP, Khoury MJ, Rozen R, Oakley Reece EA, Wu YK, Wiznitzer A, Homko C, Yao increased with maternal smoking and spe- GPJr, Adams MJJr. 1996. 5,10-Methylene- J, Borenstein M, Sloskey G. 1996b. Dietary cific detoxification-gene variants. Am J tetrahydrofolate reductase genetic polymor- polyunsaturated fatty acid prevents malfor- Hum Genet 80:76–90. phism as a risk factor for neural tube defects. mations in offspring of diabetic rats. Am J Shi M, Wehby GL, Murray JC. 2008. Review on Am J Med Genet 63:610–614. Obstet Gynecol 175:818–823. genetic variants and maternal smoking in Ozerol E, Ozerol I, Gokdeniz R, Temel I, Akyol Roy M, Leclerc D, Wu Q, Gupta S, Kruger WD, the etiology of oral clefts and other birth O. 2004. Effect of smoking on serum Rozen R. 2008. Valproic acid increases defects. Birth Defects Res C Embryo Today concentrations of total homocysteine, folate, expression of methylenetetrahydrofolate 84:16–29. vitamin B12, and nitric oxide in pregnancy: reductase (MTHFR) and induces lower Sivan E, Reece EA, Wu YK, Homko CJ, A preliminary study. Fetal Diagn Ther 19: teratogenicity in MTHFR deficiency. J Cell Polansky M, Borenstein M. 1996. Dietary 145–148. Biochem 105:467–476. vitamin E prophylaxis and diabetic embry- Padmanabhan R, Shafiullah MM. 2003. Amelio- Salbaum JM, Kappen C. 2010. Neural tube defect opathy: Morphologic and biochemical anal- ration of sodium valproate-induced neural genes and maternal diabetes during preg- ysis. Am J Obstet Gynecol 175:793–799. tube defects in mouse fetuses by maternal nancy. Birth Defects Res A Clin Mol Teratol Smithells RW, Sheppard S, Schorah CJ, Seller MJ, folic acid supplementation during gestation. 88:601–611. Nevin NC, Harris R, Read AP, Fielding Congenit Anom (Kyoto) 43:29–40. Savona-Ventura C, Gatt M. 2004. Embryonal DW. 1980. Possible prevention of neural- Pani L, Horal M, Loeken MR. 2002a. Poly- risks in gestational diabetes mellitus. Early tube defects by periconceptional vitamin morphic susceptibility to the molecular Hum Dev 79:59–63. supplementation. Lancet 1:339–340. causes of neural tube defects during diabetic Sever LE. 1995. Looking for causes of neural tube Spiegelstein O, Chatterjie N, Alexander G, embryopathy. Diabetes 51:2871–2874. defects: Where does the environment fit in? Finnell RH. 2003. Teratogenicity of Pani L, Horal M, Loeken MR. 2002b. Rescue Environ Health Perspect 103:165–171. valproate conjugates with anticonvulsant of neural tube defects in Pax-3-deficient Shaw GM, Jensvold NG, Wasserman CR, activity in mice. Epilepsy Res 57:145–152. embryos by p53 loss of function: Implica- Lammer EJ. 1994. Epidemiologic character- Spiegelstein O, Gould A, Wlodarczyk B, Tsie M, tions for Pax-3-dependent development istics of phenotypically distinct neural tube Lu X, Le C, Troen A, Selhub J, Piedrahita and tumorigenesis. Genes Dev 16:676– defects among 0.7 million California births, JA, Salbaum JM, Kappen C, Melnyk S, 680. 1983–1987. Teratology 49:143–149. James J, Finnell RH. 2005. Developmental Parle-McDermott A, Pangilinan F,Mills JL, Kirke Shaw GM, Velie EM, Schaffer D. 1996. Risk consequences of in utero sodium arsenate PN, Gibney ER, Troendle J, O’Leary VB, of neural tube defect-affected pregnancies exposure in mice with folate transport Molloy AM, Conley M, Scott JM, Brody among obese women. JAMA 275:1093– deficiencies. Toxicol Appl Pharmacol 203: LC. 2007. The 19-bp deletion polymor- 1096. 18–26. phism in intron-1 of dihydrofolate reductase Shaw GM, Todoroff K, Velie EM, Lammer EJ. Styrud J, Eriksson UJ. 1992. Development of (DHFR) may decrease rather than increase 1998a. Maternal illness, including fever and rat embryos in culture media containing risk for spina bifida in the Irish population. medication use as risk factors for neural tube different concentrations of normal and Am J Med Genet Part A 143A:1174–1180. defects. Teratology 57:1–7. diabetic rat serum. Teratology 46:473– Pavlinkova G, Salbaum JM, Kappen C. 2009. Shaw GM, Wasserman CR, Murray JC, Lammer 483. Maternal diabetes alters transcriptional pro- EJ. 1998b. Infant TGF-alpha genotype, Suarez L, Felkner M, Brender JD, Canfield M, grams in the developing embryo. BMC orofacial clefts, and maternal periconcep- Hendricks K. 2008. Maternal exposures to Genomics 10:274. tional multivitamin use. Cleft Palate Cra- cigarette smoke, alcohol, and street drugs Pennell PB. 2006. 2005 AES annual course: niofac J 35:366–370. and neural tube defect occurrence in Evidence used to treat women with epilepsy. Shaw GM, Todoroff K, Schaffer DM, Selvin S. offspring. Matern Child Health J 12:394– Epilepsia 47:46–453. 1999a. Periconceptional nutrient intake and 401. Perucca E. 2005. Birth defects after prenatal risk for neural tube defect-affected pregnan- Suarez L, Ramadhani T, Felkner M, Canfield exposure to antiepileptic drugs. Lancet cies. Epidemiology 10:711–7116. MA, Brender JD, Romitti PA, Sun L. 2011. Neurol 4:781–786. Shaw GM, Wasserman CR, O’Malley CD, Maternal smoking, passive tobacco smoke, Phelan SA, Ito M, Loeken MR. 1997. Neural Nelson V, Jackson RJ. 1999b. Maternal and neural tube defects. Birth Defects Res A tube defects in embryos of diabetic mice: pesticide exposure from multiple sources Clin Mol Teratol 91:29–33. Role of the Pax-3 gene and apoptosis. and selected congenital anomalies. Epidemi- Suzuki N, Svensson K, Eriksson UJ. 1996. High Diabetes 46:1189–1197. ology 10:60–66. glucose concentration inhibits migration 226 AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) ARTICLE

of rat cranial neural crest cells in vitro. Volcik KA, Shaw GM, Lammer EJ, Zhu H, Wiltse J. 2005. Mode of action: Inhibition of Diabetologia 39:401–411. Finnell RH. 2003a. Evaluation of infant histone deacetylase, altering WNT-depend- Takao Y, Akazawa S, Matsumoto K, Takino H, methylenetetrahydrofolate reductase geno- ent gene expression, and regulation of beta- Akazawa M, Trocino RA, Maeda Y, Okuno type, maternal vitamin use, and risk of high catenin—Developmental effects of valproic S, Kawasaki E, Uotani S, et al. 1993. versus low level spina bifida defects. Birth acid. Crit Rev Toxicol 35:727–738. Glucose transporter gene expression in rat Defects Res A Clin Mol Teratol 67:154– Wlodarczyk B, Spiegelstein O, Gelineau-van conceptus during high glucose culture. 157. Waes J, Vorce RL, Lu X, Le CX, Finnell Diabetologia 36:696–706. Volcik KA, Shaw GM, Zhu H, Lammer EJ, RH. 2001. Arsenic-induced congenital Tanimura T, Shepard TH. 1970. Glucose metab- Laurent C, Finnell RH. 2003b. Associations malformations in genetically susceptible olism by rat embryos in vitro. Proc Soc Exp between polymorphisms within the thymi- folate binding protein-2 knockout mice. Biol Med 135:51–54. dylate synthase gene and spina bifida. Birth Toxicol Appl Pharmacol 177:238–246. Torfs CP, Christianson RE, Iovannisci DM, Defects Res A Clin Mol Teratol 67:924– Wlodarczyk BJ, Cabrera RM, Hill DS, Bozinov Shaw GM, Lammer EJ. 2006. Selected gene 928. D, Zhu H, Finnell RH. 2006a. Arsenic- polymorphisms and their interaction with Vurucu S, Demirkaya E, Kul M, Unay B, Gul D, induced gene expression changes in the maternal smoking, as risk factors for gastro- Akin R, Gokcay E. 2008. Evaluation of the neural tube of folate transport defective schisis. Birth Defects Res A Clin Mol relationship between C677T variants of mouse embryos. Neurotoxicology 27:547– Teratol 76:723–730. methylenetetrahydrofolate reductase gene 557. Trocino RA, Akazawa S, Takino H, Takao Y, and hyperhomocysteinemia in children Wlodarczyk BJ, Tang LS, Triplett A, Aleman F, Matsumoto K, Maeda Y,Okuno S, Nagataki receiving antiepileptic drug therapy. Prog Finnell RH. 2006b. Spontaneous neural S. 1994. Cellular-tissue localization and Neuropsychopharmacol Biol Psychiatry tube defects in splotch mice supplemented regulation of the GLUT-1 protein in both 32:844–848. with selected micronutrients. Toxicol Appl the embryo and the visceral yolk sac from Waller DK, Mills JL, Simpson JL, Cunningham Pharmacol 213:55–63. normal and experimental diabetic rats dur- GC, Conley MR, Lassman MR, Rhoads Wu X, Bishopric NH, Discher DJ, Murphy BJ, ing the early postimplantation period. GG. 1994. Are obese women at higher risk Webster KA. 1996. Physical and functional Endocrinology 134:869–878. for producing malformed offspring? Am J sensitivity of zinc finger transcription factors van der Linden IJ, den Heijer M, Afman LA, Obstet Gynecol 170:541–548. to redox change. Mol Cell Biol 16:1035– Gellekink H, Vermeulen SH, Kluijtmans Waller DK, Shaw GM, Rasmussen SA, Hobbs 1046. LA, Blom HJ. 2006. The methionine CA, Canfield MA, Siega-Riz AM, Gallaway Zabihi S, Loeken MR. 2010. Understanding synthase reductase 66A>G polymorphism MS, Correa A. 2007. Prepregnancy obesity diabetic teratogenesis: Where are we now is a maternal risk factor for spina bifida. J as a risk factor for structural birth defects. and where are we going? Birth Defects Res Mol Med 84:1047–1054. Arch Pediatr Adolesc Med 161:745–750. A Clin Mol Teratol 88:779–790. van der Linden IJ, Nguyen U, Heil SG, Franke B, Watkins ML, Botto LD. 2001. Maternal prepreg- Zeiger JS, Beaty TH, Liang KY. 2005. Oral clefts, Vloet S, Gellekink H, den Heijer M, Blom nancy weight and congenital heart defects in maternal smoking, and TGFA: A meta- HJ. 2007. Variation and expression of offspring. Epidemiology 12:439–446. analysis of gene–environment interaction. dihydrofolate reductase (DHFR) in relation Watkins ML, Scanlon KS, Mulinare J, Khoury MJ. Cleft Palate Craniofac J 42:58–63. to spina bifida. Mol Genet Metab 91:98– 1996. Is maternal obesity a risk factor for Zhao Z, Reece EA. 2005. Experimental mech- 103. anencephaly and spina bifida? Epidemiology anisms of diabetic embryopathy and van der Put NM, Steegers-Theunissen RP, Frosst 7:507–512. strategies for developing therapeutic inter- P, Trijbels FJ, Eskes TK, van den Heuvel LP, Watkins ML, Rasmussen SA, Honein MA, Botto ventions. J Soc Gynecol Investig 12:549– Mariman EC, den Heyer M, Rozen R, LD, Moore CA. 2003. Maternal obesity and 557. Blom HJ. 1995. Mutated methylenetetrahy- risk for birth defects. Pediatrics 111:1152– Zhu H, Junker WM, Finnell RH, Brown S, Shaw drofolate reductase as a risk factor for spina 1158. GM, Lammer EJ, Canfield M, Hendricks K. bifida. Lancet 346:1070–1071. Wentzel P, Thunberg L, Eriksson UJ. 1997. 2003. Lack of association between ZIC2 and van Rooij IA, Wegerif MJ, Roelofs HM, Peters Teratogenic effect of diabetic serum is ZIC3 genes and the risk of neural tube WH, Kuijpers-Jagtman AM, Zielhuis GA, prevented by supplementation of superoxide defects (NTDs) in Hispanic populations. Merkus HM, Steegers-Theunissen RP. dismutase and N-acetylcysteine in rat Am J Med Genet Part A 116A:414– 2001. Smoking, genetic polymorphisms in embryo culture. Diabetologia 40:7–14. 415. biotransformation enzymes, and nonsyn- Wentzel P, Wentzel CR, Gareskog MB, Eriksson Zhu H, Curry S, Wen S, Wicker NJ, Shaw GM, dromic oral clefting: A gene–environment UJ. 2001. Induction of embryonic dysmor- Lammer EJ, Yang W, Jafarov T, Finnell RH. interaction. Epidemiology 12:502–507. phogenesis by high glucose concentration, 2005. Are the betaine-homocysteine meth- Veiby G, Daltveit AK, Engelsen BA, Gilhus NE. disturbed inositol metabolism, and inhibited yltransferase (BHMT and BHMT2) genes 2009. Pregnancy, delivery, and outcome for protein kinase C activity. Teratology 63: risk factors for spina bifida and orofacial the child in maternal epilepsy. Epilepsia 193–201. clefts? Am J Med Genet Part A 135A:274– 50:2130–2139. Werler MM, Louik C, Shapiro S, Mitchell AA. 277. Venners SA, Wang X, Chen C, Wang L, Chen D, 1996. Prepregnant weight in relation to risk Zhu H, Yang W, Lu W, Zhang J, Shaw GM, Guang W, Huang A, Ryan L, O’Connor J, of neural tube defects. JAMA 275:1089– Lammer EJ, Finnell RH. 2006. A known Lasley B, Overstreet J, Wilcox A, Xu X. 1092. functional polymorphism (Ile120Val) of 2004. Paternal smoking and pregnancy loss: White FM, Cohen FG, Sherman G, McCurdy R. the human PCMT1 gene and risk of A prospective study using a biomarker of 1988. Chemicals, birth defects and stillbirths spina bifida. Mol Genet Metab 87:66–70. pregnancy. Am J Epidemiol 159:993–1001. in New Brunswick: Associations with Zhu H, Enaw JO, Ma C, Shaw GM, Lammer EJ, Vinten J, Adab N, Kini U, Gorry J, Gregg J, Baker agricultural activity. CMAJ 138:117–124. Finnell RH. 2007. Association between GA. 2005. Neuropsychological effects of Wilson JG, Warkany J. 1965. Teratology: Princi- CFL1 gene polymorphisms and spina bifida exposure to anticonvulsant medication in ples and techniques. Chicago: University of risk in a California population. BMC Med utero. Neurology 64:949–954. Chicago Press. 279 p. Genet 8:12.