ArticlePDF Available

Transcobalamin II Receptor Polymorphisms Are Associated with Increased Risk for Neural Tube Defects

Authors:

Abstract and Figures

Women who have low cobalamin (vitamin B(12)) levels are at increased risk for having children with neural tube defects (NTDs). The transcobalamin II receptor (TCblR) mediates uptake of cobalamin into cells. Inherited variants in the TCblR gene as NTD risk factors were evaluated. Case-control and family-based tests of association were used to screen common variation in TCblR as genetic risk factors for NTDs in a large Irish group. A confirmatory group of NTD triads was used to test positive findings. 2 tightly linked variants associated with NTDs in a recessive model were found: TCblR rs2336573 (G220R; p(corr)=0.0080, corrected for multiple hypothesis testing) and TCblR rs9426 (p(corr)=0.0279). These variants were also associated with NTDs in a family-based test before multiple test correction (log-linear analysis of a recessive model: rs2336573 (G220R; RR=6.59, p=0.0037) and rs9426 (RR=6.71, p=0.0035)). A copy number variant distal to TCblR and two previously unreported exonic insertion-deletion polymorphisms were described. TCblR rs2336573 (G220R) and TCblR rs9426 represent a significant risk factor in NTD cases in the Irish population. The homozygous risk genotype was not detected in nearly 1000 controls, indicating that this NTD risk factor may be of low frequency and high penetrance. 9 other variants are in perfect linkage disequilibrium with the associated single nucleotide polymorphisms. Additional work is required to identify the disease-causing variant. Our data suggest that variation in TCblR plays a role in NTD risk and that these variants may modulate cobalamin metabolism.
Content may be subject to copyright.
doi: 10.1136/jmg.2009.073775
published online June 24, 2010J Med Genet
F Pangilinan, A Mitchell, J VanderMeer, et al.
defects
associated with increased risk for neural tube
Transcobalamin II receptor polymorphisms are
http://jmg.bmj.com/content/early/2010/06/24/jmg.2009.073775.full.html
Updated information and services can be found at:
These include:
References http://jmg.bmj.com/content/early/2010/06/24/jmg.2009.073775.full.html#ref-list-1
This article cites 51 articles, 10 of which can be accessed free at:
P<P Published online June 24, 2010 in advance of the print journal.
service
Email alerting box at the top right corner of the online article.
Receive free email alerts when new articles cite this article. Sign up in the
Notes
articles must include the digital object identifier (DOIs) and date of initial publication.
priority; they are indexed by PubMed from initial publication. Citations to Advance online
prior to final publication). Advance online articles are citable and establish publication
yet appeared in the paper journal (edited, typeset versions may be posted when available
Advance online articles have been peer reviewed and accepted for publication but have not
http://jmg.bmj.com/cgi/reprintform
To order reprints of this article go to:
http://jmg.bmj.com/subscriptions go to: Journal of Medical GeneticsTo subscribe to
group.bmj.com on August 25, 2010 - Published by jmg.bmj.comDownloaded from
Transcobalamin II receptor polymorphisms are
associated with increased risk for neural tube defects
F Pangilinan,
1
A Mitchell,
1
J VanderMeer,
1
A M Molloy,
2
J Troendle,
3
M Conley,
3
P N Kirke,
4
M Sutton,
4
J M Sequeira,
5
E V Quadros,
5
J M Scott,
6
J L Mills,
3
L C Brody
1,7
ABSTRACT
Objective Women who have low cobalamin (vitamin B
12
)
levels are at increased risk for having children with neural
tube defects (NTDs). The transcobalamin II receptor(TCblR)
mediates uptake of cobalamin into cells. Inherited variants
in the TCblR gene as NTD risk factors were evaluated.
Methods Caseecontrol and family-based tests of
association were used to screen common variation in
TCblR as genetic risk factors for NTDs in a large Irish
group. A confirmatory group of NTD triads was used to
test positive findings.
Results 2 tightly linked variants associated with NTDs in
a recessive model were found: TCblR rs2336573 (G220R;
p
corr
¼0.0080, corrected for multiple hypothesis testing)
and TCblR rs9426 (p
corr
¼0.0279). These variants were
also associated with NTDs in a family-based test before
multiple test correction (log-linear analysis of a recessive
model: rs2336573 (G220R; RR¼6.59, p¼0.0037) and
rs9426 (RR¼6.71, p¼0.0035)). A copy number variant
distal to TCblR and two previously unreported exonic
insertionedeletion polymorphisms were described.
Conclusions TCblR rs2336573 (G220R) and TCblR
rs9426 represent a significant risk factor in NTD cases in
the Irish population. The homozygous risk genotype was
not detected in nearly 1000 controls, indicating that this
NTD risk factor may be of low frequency and high
penetrance. 9 other variants are in perfect linkage
disequilibrium with the associated single nucleotide
polymorphisms. Additional work is required to identify the
disease-causing variant. Our data suggest that variation in
TCblR plays a role in NTD risk and that these variants may
modulate cobalamin metabolism.
Neural tube defects (NTDs) are common birth
defects, affecting approximately 1 in 1000 preg-
nancies.
1 2
The neural tube closes during the
4 weeks of embryogenesis and gives rise to the
spinal cord and brain. Incomplete closure of
the neural tube causes a range of birth defects
including spina bida and anencephaly.
NTD aetiology is multifactorial and includes
environmental and genetic factors.
3 4
Periconcep-
tional folic acid supplementation in mothers can
reduce the risk of an NTD-affected pregnancy by
up to 70%.
56
Genetic variants relating to folate
metabolism have also been implicated in contrib-
uting to NTD risk. The most well studied of these
is the 677C/T variant in the methylene tetrahy-
drofolate reductase (MTHFR) gene (reviewed by
Botto and Yang
7
). In the European population, this
variant may account for 26% of NTDs.
8
Cobalamin (vitamin B
12
) plays an important role
in folate metabolism as a cofactor for methionine
synthase (MTR), the enzyme that catalyses the
transfer of the methyl group from 5-methyl tetra-
hydrofolate to homocysteine to produce methio-
nine. Compromised methionine synthase function
results in functional intracellular folate deciency
and a decit of methyl groups for methylation
reactions. Low maternal blood
9e17
or
amniotic uid
18e22
cobalamin levels have been
associated with NTD risk. Levels of the cobala-
minetranscobalamin II complex (holoTC) are also
reduced in mothers during
23
or after
24
an NTD
pregnancy.
Polymorphisms in cobalamin-related genes have
been investigated for NTD risk. Several transporter
proteins, receptors and converting enzymes are
required to ensure that dietary cobalamin is deliv-
ered in an active form to methionine synthase and
methylmalonyl coenzyme A mutase (MUT).
Transcobalamin II (TCN2) transports cobalamin
from the intestinal circulation into the bloodstream
and ultimately to target tissues.
25
TCN2 P259R
(rs1801198) was reported to be a maternal NTD
risk factor,
26
although other studies examined
multiple TCN2 variants without detecting an
effect.
24 27
Methionine synthase reductase (MTRR)
rereduces oxidised cobalamin, thereby maintaining
methionine synthase activity. Although some
studies found no association between NTDs and
variants in MTR or MTRR,
28e32
others observed
independent and/or joint effects.
16 33e37
Cubilin
(CUBN) is a receptor found on the luminal
epithelium of the intestine and the kidney that
binds the cobalamineintrinsic factor complex. An
intronic single nucleotide polymorphism (SNP,
rs1907362) in CUBN has been associated with
NTDs.
38
Uptake of the cobalaminetranscobalamin II
complex into cells and tissues occurs via binding to
the recently identied transcobalamin II receptor
(TCblR, also known as CD320 and 8D6).
39
Because
of TCblRsessential role in cobalamin bioavail-
ability, we evaluated its genetic variation for asso-
ciation with NTDs in a large Irish cohort. We report
a signicantly associated genetic risk factor for
NTDs in TCblR.
MATERIALS AND METHODS
Study populations
Recruitment of our Irish case and control popula-
tions has been previously described.
40e42
Our
complete Irish case population consists of 586
families with an NTD-affected child. This includes
442 full family triads (case, mother and father), 57
1
Molecular Pathogenesis
Section, Genome Technology
Branch, National Human
Genome Research Institute,
Bethesda, Maryland, USA
2
Department of Clinical
Medicine, Trinity College Dublin,
Dublin, Ireland
3
Division of Epidemiology,
Statistics and Prevention
Research, The Eunice Kennedy
Shriver National Institute of
Child Health and Human
Development, Department of
Health and Human Services,
National Institutes of Health,
Bethesda, Maryland, USA
4
Child Health Epidemiology Unit,
Health Research Board, Dublin,
Ireland
5
Departments of Medicine and
Cell Biology, The State
University of New York
Downstate Medical Center,
Brooklyn, New York, USA
6
Department of Biochemistry,
Trinity College Dublin, Dublin,
Ireland
7
Molecular Pathogenesis
Section, Genome Technology
Branch, National Human
Genome Research Institute,
Bethesda, Maryland, USA
Correspondence to
Lawrence C Brody, Molecular
Pathogenesis Section, Genome
Technology Branch, National
Human Genome Research
Institute, Room 5306, Building
50, 50 South Drive, MSC 8004,
Bethesda, MD 20892-8004,
USA; lbrody@helix.nih.gov
Received 6 October 2009
Revised 16 December 2009
Accepted 1 February 2010
Pangilinan F, Mitchell A, VanderMeer J, et al.J Med Genet (2010). doi:10.1136/jmg.2009.073775 1 of 9
Original article
JMG Online First, published on June 24, 2010 as 10.1136/jmg.2009.073775
Copyright Article author (or their employer) 2010. Produced by BMJ Publishing Group Ltd under licence.
group.bmj.com on August 25, 2010 - Published by jmg.bmj.comDownloaded from
caseemother pairs, 4 caseefather pairs, 44 cases only and 39
families with parents or single parent only. In total, there are 530
spina bida and 21 encephalocoele cases. Our control samples
(n¼999) were randomly selected from a bank of 56 049 blood
samples taken from women during their rst prenatal visit at
the three major maternity hospitals in Dublin between 1986 and
1990, after excluding samples from women whose current or
past pregnancies involved an NTD.
An additional 367 case families were recruited in the UK
between 2001 and 2003, with the assistance of the UK Associ-
ation for Spina Bida and Hydrocephalus (ASBAH). This
cohort includes 258 full family triads, 66 caseemother pairs, 2
caseefather pairs, 38 caseeonly samples and 3 families with
parents only. There are 354 spina bida cases, 8 encephalocoele
cases and 2 cases with spina bida and encephalocoele. All the
UK families gave buccal swab samples collected using the
protocol of Meulenbelt et al
43
and completed questionnaires
detailing the cases NTD type, maternal pregnancy history and
family history of birth defects.
These cohorts are comparable in folic acid supplementation.
Of the Irish NTD mothers, 91 denitively answered whether
they were taking any folic acid or multivitamins in the month
before their last monthly period; 23 (25.3%) were on supple-
ments. Of the UK NTD mothers, 73 denitively answered the
same question; 23 (31.5%) were on supplements. Additionally,
the distributions of case birth years in the two cohorts were
similar and largely predated the recommendation of folate
supplementation by the United States Public Health Service in
1992. Case birth years in the Irish NTD cohort ranged from 1938
to 2003 with a median of 1981 and an SD of 11 years. Case birth
years in the UK NTD cohort ranged from 1926 to 2001 with
a median of 1978 and an SD of 15 years.
The sample collections were approved by the Health Research
Board Research Ethics Committee (Dublin, Ireland), the UK
Multi-Centre Research Ethics Committee (University of
Newcastle, UK) in collaboration with UK ASBAH and the
Institutional Review Board at the National Human Genome
Research Institute (Bethesda, Maryland, USA). Written consent
was obtained from all the participants.
Genomic DNA was extracted from all the blood samples and
buccal swabs using the QIAamp DNA Blood Mini Kit (Qiagen,
Valencia, CA, USA).
African American control DNA (HD100AA) and Caucasian
control DNA (HD200CAU) samples were purchased from the
Coriell Cell Repositories, Camden, NJ, USA.
SNP selection and discovery
To select a set of SNPs to capture common genetic variation in
TCblR, we evaluated SNPs genotyped by HapMap.
44
SNPs
within and up to 10 kilobases (kb) from the gene were consid-
ered. A minimum SNP set representing all such HapMap SNPs
was manually selected, while allowing exclusion of redundant
SNPs (r
2
>0.8).
Up to 18 Irish individuals, most of which were drawn from
NTD families, were screened for unreported polymorphisms.
Primers were designed to polymerase chain reaction
(PCR)eamplify exons 1 through 5; the resulting amplicons were
analysed via uorescent automated dideoxy DNA sequencing.
Genotyping
Multiplex ligation-dependent probe amplication (MLPA) was
used to estimate the relative copy number for the 14.8 kb copy
number variant (CNV) immediately downstream from TCblR.
Briey, probe pairs were designed to hybridise to test and control
loci (primer sequences available upon request). The multiplex
reaction contains probe pairs to detect ve test loci within the
CNV. The control loci included four single copy regions in
TCblR, and one single copy probe was placed in TCN2 for rela-
tive quantication. A locus in the androgen receptor was
included as an additional control to ensure that the method
detects copy number differences of the X chromosome.
The MLPA reaction was performed as directed with the
SALSA kit reagents obtained from MRC-Holland (Amsterdam,
The Netherlands) using at least 50 ng genomic DNA for each
sample. After hybridisation and ligation, products were PCR
amplied and analysed on an ABI 3100 Genetic Analyser
(Applied Biosystems, Foster City, CA, USA). Comparisons of
amplicon peak areas were used to estimate relative DNA quan-
tity scores for each locus. A real-time PCR assay using the
Applied Biosystems 7900HT Fast Real-Time PCR System
(Applied Biosystems) was used to independently estimate copy
number in a subset of samples with good correlation.
All other variants were genotyped by detection of allele-
specic extension products via matrix-assisted laser desorption/
ionisationetime of ight (MALDI-TOF) mass spectrometry
(Sequenom, San Diego, CA, USA). Primer sequences and assay
conditions are available upon request.
Genotyping quality was assessed in each sample population.
More than 10% of the Irish samples were repeated with $99%
concordance for all the variants. More than 8% of the UK
samples were repeated with $98% concordance for all the
variants. More than 7% of the Caucasian (Coriell) samples were
repeated with 100% concordance for all the variants. More than
10% of the African American (Coriell) samples were repeated
with 100% concordance for all but two variants (E88del and
rs9426, one discrepant sample each).
The mean genotyping call rates (ie, success rates) were 97%
for the Irish and UK samples. Call rates were at least 95% for all
the variants in all the groups with the exception of rs4147651 in
the Irish NTD cases (94%) and rs2232775 (Q8R) in the Irish
controls (93%). The mean genotyping call rates were 98% in the
Caucasian Coriell samples and 92% in the African American
Coriell samples. Call rates were at least 93% for all the variants
in both Coriell groups with the exception of rs250503 (87%) in
African Americans.
Only the NTD cases from Ireland were observed to deviate
from the HardyeWeinberg equilibrium (HWE, p<0.01): the
rs2232775 (Q8R) NTD cases (
c
2
¼15.4, p¼8.9310
05
), the
rs2336573 (G220R) NTD cases (
c
2
¼27.8, p¼1.3E
07
), the rs9426
NTD cases (
c
2
¼21.2, p¼7.2E
06
), and the E88del NTD cases
(
c
2
¼51.1, p¼8.7E
13
). These four SNPs were in HWE in all the
other genotyped groups. Additionally, the rst three SNPs are in
strong r
2
linkage disequilibrium (LD; r
2
$0.94), and similar
genotyping results were obtained with these independent
genotype assays. The E88del polymorphism has a low minor
allele frequency (MAF; p=0.02), which may account for its
failure to adhere to HWE in the NTD cases.
Discordant genotypes and triads exhibiting noneMendelian
inheritance for any single marker were excluded for that marker.
Twenty-four samples with discrepancies for >1 marker were
excluded from all the analyses.
Haplotype analysis
LD in the region was estimated using Haploview (http://www.
broad.mit.edu/mpg/haploview/).
45
Haplotype blocks were
dened based on D9values using the solid spine of LD option in
Haploview. Haplotype frequency estimates based on these block
denitions were then generated for Irish NTD cases, NTD
2 of 9 Pangilinan F, Mitchell A, VanderMeer J, et al.J Med Genet (2010). doi:10.1136/jmg.2009.073775
Original article
group.bmj.com on August 25, 2010 - Published by jmg.bmj.comDownloaded from
mothers and controls using PHASE V.2.1.
46 47
A permutation
test within PHASE V.2.1 was used to test whether haplotype
frequency distributions differed between the controls and the
NTD cases or the NTD mothers in the Irish cohort.
Statistical analyses
A logistic regression model, with the number of risk alleles as the
independent variable, was used as a primary test to evaluate
each polymorphism for NTD association. In this multiplicative
model, the odds ratio (OR) of two risk alleles is the square of the
OR of one risk allele. Additionally, one degree of freedom (DOF)
models of dominant and recessive genetic risk were applied via
logistic regression. These tests are not valid when any of the
genotypes cells are empty; in these cases, the Fisher exact test
was used in place of the logistic regression to test for genotypic
differences. These tests were used to perform NTD caseecontrol
comparisons and NTD motherecontrol comparisons in the Irish
NTD cases, the NTD mothers and the controls.
The (mother, father and case) triads were analysed by tting
multiplicative, dominant and recessive log-linear models with
one DOF to test for case effects and one DOF to test for direct
maternal effects.
48
The case term in the multiplicative log-linear
model provides comparable information to the transmission
disequilibrium test.
49
These analyses were modied to incorpo-
rate data from incomplete triads by using the expectation
maximisation algorithm.
50
These family-based tests were
applied to the Irish and UK NTD triads to test TCblR variants
for association with NTDs.
Correction for multiple tests was by permutation (99 999
random permutations). This method accounts for any LD
(non-independence) between evaluated SNPs. Multivariate
permuting of triads for log-linear analysis involved treating the
test as a one-sample test and permuting the hypothetical risk
allele. Permutations of the cases and the controls were inde-
pendent of permutations of triads, and the results were
combined by Bonferroni adjustment so that the resulting
adjusted p values accounted for all tests and all SNPs
while controlling the chance of any false positive (familywise
error) at 5%.
RESULTS
Genomic analysis
The TCblR gene spans approximately 6 kb on chromosome 19
(gure 1a). It is anked by a gene encoding a subunit of
ubiquinone, NDUAF7, and the LAG1 longevity assurance
homologue 4 (LASS4) gene, which encodes a ceramide synthase.
TCblR and NDUFA7 are in close proximity (approximately 3 kb)
and transcribed from the same strand. In contrast, LASS4 is
transcribed from the opposite strand and its 39terminus is
mapped approximately 40 kb away from the last exon of TCblR.
The reference human genomic sequence appears contiguous
through this region. Close examination of the genomic
sequencing used to build this region of the genome revealed that
the sequence in this interval had not been completed. The gap in
the sequence was annotated in GenBank (contig, NT_077812) as
including an undetermined number of an approximately 15 kb
repetitive element. During the course of this work, genome
annotation was expanded to include areas containing potential
segmental duplications. To ensure that we were capturing all
regional variability related to the TCblR gene, we rst asked if
this segmental duplication varied in copy number between
individuals. The CNVs have been implicated in disease and have
been shown to affect expression for nearby genes. We developed
an MLPA assay for this region and screened Caucasian and
African American samples available from the Coriell Cell
Repositories. We found that this region was polymorphic (ie,
this a bona de CNV) in the African American (MAF¼0.054)
and Caucasian (MAF¼0.021) samples tested. MLPA alone does
not denitively allow determination of copy number; however,
the most parsimonious model consistent with the MLPA-derived
data is that the common allele has two repeats, whereas the
minor allele has three repeats. The CNV was not found to be in
strong LD with any other marker (gure 1b). Although it could
serve as a candidate variant, we excluded it from further
consideration because of its low MAF and the relatively large
quantity of DNA consumed by the MLPA assay.
SNP analysis and LD in the TCblR region
We selected 10 HapMap
44
polymorphisms to cover the region
within and anking TCblR. Additionally, while developing the
MLPA assay, we discovered a C/GA insertionedeletion (indel)
polymorphism in the 39UTR of exon 5 located 83 bases down-
stream from the stop codon. Lastly, to search for unreported
common variation, limited screening of TCblR coding regions
was performed by directly sequencing exons in at least 12 Irish
individuals. This revealed a three nucleotide insertionedeletion
(indel) polymorphism in exon 2, resulting in the presence or
absence of one of the three tandem glutamic acid (E) residues
starting at codon 86 (TCblR E88del).
These 12 polymorphisms and the CNV were typed in
Caucasians and African Americans to determine the LD struc-
ture in the region (gure 1b). Blocks dened by measures of D9
revealed that the upstream region and most TCblRs are in
a single block of LD (gure 1). In the Irish population, this block
encompassed all markers within TCblR. Most of the markers
examined are independently informative (r
2
<0.8). The excep-
tions consist of two sets of markers that are in high r
2
LD: (1)
rs2232775 (Q8R), rs2336573 (G220R) and rs9426 (r
2
$0.94) and
(2) rs250511 and rs173665 (r
2
¼0.99). A total of 12 markers were
genotyped in the Irish NTD cohort (table 1). The exon 5 indel
had the lowest MAF (0.001) and was excluded from further
study.
Evaluation of individual markers for association with NTDs
Each marker (n¼11) was tested for caseecontrol associations
with NTDs in the Irish population using three models of logistic
regression: multiplicative, dominant and recessive models.
Signicant associations are shown in table 2. Essentially, three
positive associations were found. First, the three highly linked
SNPs were found to be signicantly associated in a recessive
model of case effect (rs2232775 (Q8R), OR¼10.6 (95% con-
dence interval (CI) 1.28 to 88.56), p¼0.029; rs2336573 (G220R),
Fisher exact test, p¼0.0002; rs9426, Fisher exact test, p¼0.0006).
These SNPs share very high r
2
values and, as predicted, yield
similar association results. Second, TCblR E88del is also signi-
cantly associated with NTDs in a recessive model of case effect
(OR¼9.2 (95% CI 1.3 to 88.9), p¼0.04). Third, the highly linked
SNP pair (rs250511, OR¼0.7 (95% CI 0.56 to 0.99), p¼0.04;
rs173665, OR¼0.7 (95% CI 0.51 to 0.94), p¼0.017) is signi-
cantly associated with NTDs in a dominant model of case effect.
Additionally, rs173665 was associated when a multiplicative
model of case effect was applied (OR¼0.7 (95% CI 0.55 to 0.96),
p¼0.02). Upon correcting for multiple tests, two markers from
the highly linked trio remained signicant: rs2336573 (G220R;
p¼0.008) and rs9426 (p¼0.03), by Fisher exact test of a recessive
model in the cases versus the controls. No other comparisons
were signicant after adjusting for multiple tests.
Pangilinan F, Mitchell A, VanderMeer J, et al.J Med Genet (2010). doi:10.1136/jmg.2009.073775 3 of 9
Original article
group.bmj.com on August 25, 2010 - Published by jmg.bmj.comDownloaded from
Examining the same markers in the NTD mothers and the
controls revealed no signicant associations (data not shown).
A family-based test of association was also used to evaluate
these 11 markers for case and maternal NTD risk (table 3).
Multiplicative (data not shown), dominant and recessive models
of log-linear analysis were applied. Depending on the applied
model and the corresponding genotype frequencies, the test
failed to converge (no result obtained) for some SNPs. One
positive result was observed among the successfully tested SNPs.
All the three highly linked SNPs were signicantly associated
with a strong RR in log-linear analysis of the recessive model in
the cases (rs2232775 (Q8R), RR¼6.0, p¼0.0086; rs2336573
Figure 1 Genomic structure and LD in
the TCblR region. (A) Genomic structure
of the region including TCblR.
Orientation, relative size and distance of
LASS4 (53 kb), the 14.8 kb CNV, TCblR
(6 kb), NDUFA7 (10 kb) and RPS28
(0.9 kb) are shown. The 14.8 kb CNV is
shown in single copy, although two
copies are predicted to be present on
most chromosomes. (B) LD plots of
polymorphisms within and surrounding
TCblR. Genotyping data from the Irish
controls (n¼993), the Caucasian
Americans (n¼79) and the African
Americans (n¼83) were used to
construct D9and r
2
LD plots. Labels for
variants within TCblR are green.
Relative distances between markers are
shown with two exceptions: the CNV is
approximately 450 bases downstream
from TCblR, and rs250508 is
approximately 32 kb downstream from
TCblR.
4 of 9 Pangilinan F, Mitchell A, VanderMeer J, et al.J Med Genet (2010). doi:10.1136/jmg.2009.073775
Original article
group.bmj.com on August 25, 2010 - Published by jmg.bmj.comDownloaded from
(G220R), RR¼6.6, p¼0.0037; and rs9426, RR¼6.7, p¼0.0035).
However, these results did not withstand correction for multiple
tests (data not shown).
Further evidence was sought by evaluating TCblR variation for
NTD risk in an independent population. Seven SNPs were tested
in a UK cohort of NTD triads (table 3). Again, some tests failed
to converge, including the log-linear analysis of the recessive
model as applied to two of the associated SNPs (rs2232775
(Q8R) and rs2336573 (G220R)). The remaining models of SNPs
that were successfully tested were not found to be associated
with the case or maternal NTD risk in the UK population.
Haplotype analysis of TCblR variants for NTD risk
To test for TCblR haplotype association, a haplotype was
constructed based on the single block of LD in the Irish popu-
lation (gure 1b). rs2336573 (G220R) was retained to tag
rs2232775 (Q8R) and rs9426, which were not included in these
analyses. Similarly, rs173665 was retained as a tag for rs250511.
The resulting haplotype block is dened by the remaining seven
markers: rs4147651, rs7408841, rs2927707, E88del, rs2336573
(G220R), rs2227288 and rs173665. Haplotype frequencies were
estimated in the Irish controls, the NTD cases and the mothers
(table 4). Five haplotypes account for >90% of the variation
present in this haplotype block in the Irish population.
A permutation test did not detect differences in haplotype
frequencies between the NTD cases and the controls (p¼0.3) or
the NTD mothers and the controls (p¼1.0).
DISCUSSION
We evaluated the association between genetic variation in the
transcobalamin II receptor gene, TCblR, and NTD risk. When
considered singly, several independent (r
2
<0.8) TCblR
Table 1 Genotype distributions and allele frequencies in Irish controls
and NTD triads
Marker Controls NTD children NTD mothers NTD fathers
NDUFA7 rs4147651
GG 636 (0.66) 314 (0.63) 310 (0.62) 289 (0.64)
GA 290 (0.30) 167 (0.33) 175 (0.35) 140 (0.31)
AA 31 (0.03) 21 (0.04) 16 (0.03) 24 (0.05)
G 0.82 0.79 0.79 0.79
A 0.18 0.21 0.21 0.21
rs7408841
GG 377 (0.39) 210 (0.41) 223 (0.44) 193 (0.42)
GC 459 (0.48) 218 (0.43) 218 (0.43) 201 (0.44)
CC 129 (0.13) 81 (0.16) 63 (0.13) 61 (0.13)
G 0.63 0.63 0.66 0.65
C 0.37 0.37 0.34 0.35
TCblR rs2232775 Q8R
TT 846 (0.91) 479 (0.91) 479 (0.93) 423 (0.91)
TC 82 (0.09) 44 (0.08) 33 (0.06) 39 (0.08)
CC 0 (0.00) 6 (0.01) 2 (0.00) 3 (0.01)
T 0.95 0.95 0.96 0.95
C 0.05 0.05 0.04 0.05
TCblR rs2927707
TT 481 (0.50) 259 (0.51) 243 (0.48) 227 (0.50)
TC 409 (0.43) 212 (0.41) 212 (0.42) 189 (0.42)
CC 71 (0.07) 41 (0.08) 51 (0.10) 38 (0.08)
T 0.71 0.71 0.69 0.71
C 0.29 0.29 0.31 0.29
TCblR E88del
E/E 915 (0.96) 495 (0.95) 493 (0.96) 436 (0.56)
E/del 35 (0.04) 20 (0.04) 35 (0.04) 18 (0.38)
del/del 1 (0.00) 5 (0.01) 1 (0.00) 0 (0.06)
E 0.98 0.97 0.98 0.96
del 0.02 0.03 0.02 0.04
TCblR rs2336573 G220R
CC 889 (0.92) 464 (0.90) 468 (0.93) 417 (0.91)
CT 80 (0.08) 42 (0.08) 35 (0.07) 39 (0.08)
TT 0 (0.00) 8(0.02) 1 (0.00) 3 (0.01)
C 0.96 0.94 0.96 0.95
T 0.04 0.06 0.04 0.05
TCblR rs2227288
GG 780 (0.81) 417 (0.82) 404 (0.80) 364 (0.80)
GC 179 (0.19) 90 (0.18) 97 (0.19) 89 (0.20)
CC 6 (0.01) 4 (0.01) 6 (0.01) 2 (0.00)
G 0.90 0.91 0.89 0.90
C 0.10 0.10 0.11 0.10
TCblR rs250511
TT 802 (0.82) 453 (0.86) 428 (0.83) 392 (0.84)
TA 175 (0.18) 72 (0.14) 83 (0.16) 76 (0.16)
AA 7 (0.01) 4 (0.01) 6 (0.01) 0 (0.00)
T 0.90 0.92 0.91 0.84
A 0.10 0.08 0.09 0.16
TCblR exon 5 indel
del/del 982 (1.00) 508 (1.00) 504 (1.00) 456 (1.00)
del/ins 1 (0.00) 1 (0.00) 1 (0.00) 1 (0.00)
Continued
Table 1 Continued
Marker Controls NTD children NTD mothers NTD fathers
ins/ins 0 (0.00) 0 (0.00) 0 (0.00) 0 (0.00)
del 1.00 1.00 1.00 1.00
ins 0.00 0.00 0.00 0.00
TCblR rs9426
CC 882 (0.91) 470 (0.90) 473 (0.93) 425 (0.91)
CT 87 (0.09) 44 (0.08) 35 (0.07) 39 (0.08)
TT 0 (0.00) 7 (0.01) 1 (0.00) 3 (0.01)
C 0.95 0.94 0.96 0.95
T 0.05 0.06 0.04 0.05
rs173665
GG 790 (0.82) 445 (0.86) 421 (0.83) 389 (0.84)
GA 171 (0.18) 66 (0.13) 79 (0.16) 74 (0.16)
AA 8 (0.01) 4 (0.01) 6 (0.01) 1 (0.00)
G 0.90 0.93 0.91 0.92
A 0.10 0.07 0.09 0.08
rs250508
CC 304 (0.32) 176 (0.35) 166 (0.34) 140 (0.30)
CT 482 (0.50) 222 (0.44) 235 (0.48) 222 (0.47)
TT 169 (0.18) 107 (0.21) 87 (0.18) 107 (0.23)
C 0.57 0.57 0.58 0.54
T 0.43 0.43 0.42 0.46
Because of rounding, group frequencies may not sum to 1. Genotypes are expressed in n
(frequency); and alleles, in frequency.
Pangilinan F, Mitchell A, VanderMeer J, et al.J Med Genet (2010). doi:10.1136/jmg.2009.073775 5 of 9
Original article
group.bmj.com on August 25, 2010 - Published by jmg.bmj.comDownloaded from
polymorphisms were implicated as NTD risk factors in the
Irish population. One pair of redundant SNPs (rs2336573
(G220R) and rs9426, r
2
¼1) remained signicantly associated
with NTDs even after rigorous correction for multiple tests. One
possible concern about the validity of this result is that the NTD
cases are not in HWE for these SNPs (see Materials and
methods), which can result from genotyping error. We consider
this unlikely because these two highly linked SNPs were geno-
typed with two independent assays, producing concordant
results. Moreover, these variants were also signicantly associ-
ated in an unadjusted recessive model of log-linear analysis that
does not use control data, eliminating the possible problem of
population stratication. Furthermore, this nding may not
have withstood correction because power is reduced when
examining a variant with a low MAF. Thus, we conclude
that the signal seen with the rs2336573 (G220R) and rs9426
variants represents a true genetic risk factor in TCblR in the Irish
population.
We sought to replicate this association in a sample of triads
from the UK. As this sample did not include population
controls, we were restricted to family-based tests of association.
The lack of individuals homozygous for the minor allele of
rs2336573 (G220R) precluded the use of the log-linear test for
a recessive case effect. Because testing of the Irish cohort
revealed a case effect, we also performed the transmission
disequilibrium test
49
but did not detect a case effect for either
rs2336573 G220R or rs2232775 Q8R in the UK cohort (data not
shown). The rs9426 SNP was not directly tested, but because of
its high LD with the other two markers, we would predict the
same result in the UK NTD cohort. This lack of replication may
be due to decreased power to detect an effect (approximately
525 Irish NTD triads vs approximately 345 UK NTD triads).
Alternatively, because of differences in dietary factors, prenatal
screening and/or greater ethnic heterogeneity, these variants
may not contribute to NTDs in the UK population.
Haplotype analysis of all the TCblR variants did not yield
further evidence of association; therefore, the identied SNP pair
exhibits the strongest signal for NTD risk. It is possible that
these variants do not alter gene function, and a yet unidentied
causal risk SNP may be linked to these two and reside on
a haplotype we have yet to test. In addition, these SNPs
(rs2336573 G220R and rs9426) have been typed in the HapMap
CEU (Caucasian) population and are in very high LD (r
2
¼1)
with ve additional TCblR SNPs: rs17160390 (intron 1),
rs2232783 (T149T), rs2232784 (S161S), rs2232785 (intron 3)
and rs2227289 (T279T). Additionally, there are four such
SNPs in introns of upstream genes: rs7249111, rs7250792,
rs2288414 in NDUFA7 and rs4147645 in ribosomal protein S28
(RPS28).
We aligned the TCblR protein sequences from multiple species
to determine whether any of these variants are in highly
conserved regions. The conservation track of the University of
CaliforniadSanta Cruz Genome Browser (http://genome.ucsc.
edu/)
51
displays syntenic regions of up to 17 species, and full
alignments for TCblR were obtained only for chimp, rhesus, dog,
cow, mouse, rat, tenrec and elephant. The only intronic SNP
with a non-zero phastCons conservation score is rs17160390
(TCblR intron 1); its signal is relatively low. In contrast,
rs2232783 (T149T) and rs2232784 (S161S) are the only other
SNPs in this set with phastCons conservation scores (0.742 and
0.677, respectively). These SNPs are part of the coding region for
the second low-density lipoprotein receptor class A (LDLRa)
domain in TCblR. As the name implies, these domains are shared
by proteins in the LDL receptor superfamily, which includes the
LDL receptor (LDLR), the LDL receptorerelated protein (LRP1),
megalin/LRP2, the apolipoprotein 2 receptor (LRP8) and the
very-lowedensity lipoprotein receptor (reviewed by May et al).
52
The LDLRa domains shared by these proteins contain cysteine-
rich regions of approximately 40 amino acids and are involved in
ligand binding. While rs2232783 (T149T) and rs2232784 (S161S)
fall into this well-conserved region, they are synonymous
coding SNPs and appear to be weak candidates for affecting
functionality.
Perhaps, the most likely causal candidate SNP is rs2336573
(G220R), the nonsynonymous coding SNP directly tested in this
study. This amino acid substitution replaces a glycine with
arginine and is predicted to reside 10 residues from the trans-
membrane and cytoplasmic domains found at the carboxyl
terminus of the protein. We aligned and examined the TCblR
orthologs from 23 mammalian species. This position contains
glycine in 14 species. The other species contain glutamic acid,
arginine, alanine or tryptophan at this residue. The moderate
conservation of this amino acid implies functional tolerance of
the rs2336573 (G220R) variant in TCblR. While such speculation
is intriguing, functional analyses of these individual variants will
be required to determine which of these NTD-associated SNPs
may be the direct contributor to NTD risk.
Strengths of the current study include a large, homogenous
Irish population and a large UK population for replication. One
limitation was the lack of UK populationebased controls, which
prevented replication of the same caseecontrol analyses that
identied rs2336573 (G220R) and rs9426 as NTD risk SNPs in
the Irish group. Thus, replication studies in independent cohorts
are required to determine whether these TCblR polymorphisms
contribute to NTDs in other populations. Additionally, the lack
of metabolite measurements in either population prevented
testing whether the risk genotype inuences circulating cobal-
amin levels.
Table 2 Caseecontrol logistic regression of significantly associated TCblR variants in the Irish population
Model
Logistic regression Fisher exact test
ORy(95% CI) p Value corrected p Value p Value Corrected p value
rs2232775 (Q8R) Recessive 10.6 (1.28 to 88.56) 0.0288 0.3747
E88del Recessive 9.2 (1.08 to 79.16) 0.0428 0.6887
rs2336573 (G220R) Recessive FTC 0.0002 0.0080
rs250511 Dominant 0.7 (0.55 to 0.99) 0.0422 1.0000
rs9426 Recessive FTC 0.0006 0.0279
rs173665 Dominant 0.7 (0.51 to 0.94) 0.0170 0.7217
rs173665 Multiplicative 0.7 (0.55 to 0.96) 0.0249 1.0000
FTC, failed to converge.
yOR for the recessive model is the odds of disease with two copies divided by the odds of disease with zero or one copy; OR for the dominant model is the odds of disease with one or two
copies divided by odds of the disease with zero copies.
OR for the multiplicative model is the OR for disease for each copy of the allele.
6 of 9 Pangilinan F, Mitchell A, VanderMeer J, et al.J Med Genet (2010). doi:10.1136/jmg.2009.073775
Original article
group.bmj.com on August 25, 2010 - Published by jmg.bmj.comDownloaded from
In summary, we identied a highly linked SNP pair (rs2336573
(G220R) and rs9426) in TCblR that is signicantly associated in
a recessive model of NTD case risk. The risk genotype (minor
allele homozygote) was undetected in the large control group
(n¼979), contributing to the large RR (RRz6) observed in NTD
cases. As a complex disease, multiple genetic risk factors are
expected to contribute to the development of NTDs. Because of
the relative rarity of the TCblR 2336573 (G220R) and TCblR
rs9426 risk genotypes (approximately 1% in the NTD cases), we
lack the power to perform meaningful interaction analyses of
these genotypes with other established NTD risk factors such as
MTHFR 677C/T. Its low frequency also means that the risk
genotype does not contribute greatly to the total genetic risk
for NTDs in the general population. However, for those few
individuals carrying the risk genotype, risk is increased approx-
imately sixfold.
Finding a genetic risk factor in the receptor required for
cobalamin bioavailability further establishes the role of cobal-
amin in the development of NTDs. This raises the possibility
that women with the risk variant may need additional dietary
cobalamin to provide sufcient intracellular cobalamin for the
developing embryo at the time of neural tube closure. There may
also be individuals with these polymorphisms in the general
population who need dietary supplements to maintain normal
Table 3 Dominant and recessive models of log-linear analysis of TCblR variants
Variant
Republic of Ireland UK
Dominant
RRy
Dominant
p value
Recessive
RR
Recessive
p value
Dominant
RR
Dominant p
value
Recessive
RR
Recessive
p value
rs4147651
Case 1.1048 0.4329 0.7598 0.3496 FTC FTC 0.6830 0.3386
Mother 1.0342 0.5795 0.5868 0.1055 0.9765 0.5303 1.0637 0.8976
rs7408841
Case FTC FTC 1.3588 0.0824 1.2350 0.2127 1.0259 0.9091
Mother FTC FTC 0.8482 0.4135 1.1464 0.3378 0.8033 0.3594
rs2232775
Case FTC FTC 6.000 0.0086 0.7965 0.4081 FTC FTC
Mother FTC FTC 0.4567 0.3357 0.8394 0.1738 FTC FTC
rs2927707
Case 1.0561 0.6148 0.7133 0.1091 0.9483 0.7146 0.9659 0.9014
Mother FTC FTC 1.2461 0.3263 0.9582 0.6013 0.8331 0.4988
E88del
Case 1.3405 0.7462 FTC FTC ND ND ND ND
Mother FTC FTC FTC FTC ND ND ND ND
rs2336573
Case FTC FTC 6.5871 0.0037 0.7064 0.2550 FTC FTC
Mother FTC FTC 0.2469 0.1858 0.7518 0.1386 FTC FTC
rs2227288
Case 1.0210 0.8971 0.6856 0.4932 0.8320 0.3962 0.4258 0.2315
Mother FTC FTC 2.6990 0.2006 0.8055 0.5434 0.4428 0.3369
rs250511
Case 0.7980 0.2625 FTC FTC ND ND ND ND
Mother FTC FTC FTC FTC ND ND ND ND
rs9426
Case FTC FTC 6.7127 0.0035 ND ND ND ND
Mother FTC FTC 0.2471 0.1656 ND ND ND ND
rs173665
Case 0.7253 0.1313 FTC FTC 0.9480 0.8021 0.7332 0.7084
Mother FTC FTC FTC FTC FTC FTC 2.4150 0.4509
rs250508
Case 0.8428 0.1861 1.2416 0.1621 ND ND ND ND
Mother 0.9832 0.6401 0.7748 0.1239 ND ND ND ND
ND, not done.
None of the reported p values are <0.05 upon correction for multiple tests (permutation).
yRR for the recessive model is the risk of disease with 2 copies divided by risk of disease with 0 or 1 copy; RR for the Dominant Model is the risk of disease with 1 or 2 copies divided by risk of
disease with 0 copies.
Pangilinan F, Mitchell A, VanderMeer J, et al.J Med Genet (2010). doi:10.1136/jmg.2009.073775 7 of 9
Original article
group.bmj.com on August 25, 2010 - Published by jmg.bmj.comDownloaded from
cobalamin homeostasis. The reported NTD risk SNPs are prime
candidates to investigate in any disease state inuenced by
cobalamin metabolism.
Acknowledgements These studies would not be possible without the participation
of the affected families and their recruitment by the Irish Association of Spina Bifida
and Hydrocephalus and the Irish Public Health Nurses in Ireland, and by the UK
ASBAH. The authors would like to thank Amanda Samuels and David Bernard for the
technical assistance in screening TCblR coding regions for polymorphisms.
Funding The authors acknowledge the research support from the intramural research
programmes of the National Human Genome Research Institute, the Eunice Kennedy
Shriver National Institute of Child Health and Human Development, the National
Institutes of Health, the Department of Health and Human Services and the Health
Research Board, Ireland. EVQ and JMS are supported by the National Institutes of
Health grant DK064732.
Competing interests None.
Patient consent Obtained.
Ethics approval This study was conducted with the approval of the National
Institutes of Health, Bethesda, Maryland, USA; the Health Research Board, Dublin,
Ireland, and the Multi-Center Research Ethics Committee, University of Newcastle,
UK.
Provenance and peer review Not commissioned; externally peer reviewed.
REFERENCES
1. Anon. Congenital malformations worldwide: a report from the international
clearinghouse for birth defects monitoring systems. Amsterdam: Elsevier Science
Publishers, 1991.
2. Busby A, Abramsky L, Dolk H, Armstrong B, Addor MC, Anneren G, Armstrong N,
Baguette A, Barisic I, Berghold A, Bianca S, Braz P, Calzolari E, Christiansen M,
Cocchi G, Daltveit AK, De Walle H, Edwards G, Gatt M, Gener B, Gillerot Y, Gjergja R,
Goujard J, Haeusler M, Latos-Bielenska A, McDonnell R, Neville A, Olars B, Portillo I,
Ritvanen A, Robert-Gnansia E, Rosch C, Scarano G, Steinbicker V. Preventing neural
tube defects in Europe: a missed opportunity. Reprod Toxicol 2005;20:393e402.
3. Cowchock S, Ainbender E, Prescott G, Crandall B, Lau L, Heller R, Muir WA, Kloza E,
Feigelson M, Mennuti M, Cederquist L. The recurrence risk for neural tube defects in
the United States: a collaborative study. Am J Med Genet 1980;5:309e14.
4. Seller MJ. Recurrence risks for neural tube defects in a genetic counseling clinic
population. J Med Genet 1981;18:245e8.
5. Anon. Prevention of neural tube defects: results of the medical research council
vitamin study. MRC vitamin study research group. Lancet 1991;338:131e7.
6. Czeizel AE, Dudas I. Prevention of the first occurrence of neural-tube defects by
periconceptional vitamin supplementation. N Engl J Med 1992;327:1832e5.
7. Botto LD, Yang Q. 5,10-Methylenetetrahydrofolate reductase gene variants and
congenital anomalies: a HuGE review. Am J Epidemiol 2000;151:862e77.
8. Kirke PN, Mills JL, Molloy AM, Brody LC, O’Leary VB, Daly L, Murray S, Conley M,
Mayne PD, Smith O, Scott JM. Impact of the MTHFR C677T polymorphism on risk of
neural tube defects: caseecontrol study. BMJ 2004;328:1535e6.
9. Gaber KR, Farag MK, Soliman SE, El-Bassyouni HT, El-Kamah G. Maternal vitamin
B12 and the risk of fetal neural tube defects in Egyptian patients. Clin Lab
2007;53:69e75.
10. Hao L, Liu M, Liu X, Chen X, Tang Y, Li Z. [Relationship between folate, vitamin B12,
total plasma homocysteine and mutation of reductase]. Zhonghua Yu Fang Yi Xue Za
Zhi 2000;34:22e4.
11. Kirke PN, Molloy AM, Daly LE, Burke H, Weir DG, Scott JM. Maternal plasma folate
and vitamin B12 are independent risk factors for neural tube defects. Q J Med
1993;86:703e8.
12. Wright ME. A case-control study of maternal nutrition and neural tube defects in
Northern Ireland. Midwifery 1995;11:146e52.
13. Zhang T, Xin R, Gu X, Wang F, Pei L, Lin L, Chen G, Wu J, Zheng X. Maternal serum
vitamin B12, folate and homocysteine and the risk of neural tube defects in the
offspring in a high-risk area of China. Public Health Nutr 2009;12:680e6.
14. Suarez L, Hendricks K, Felkner M, Gunter E. Maternal serum B12 levels and risk for
neural tube defects in a TexaseMexico border population. Ann Epidemiol
2003;13:81e8.
15. Groenen PM, van Rooij IA, Peer PG, Gooskens RH, Zielhuis GA, Steegers-Theunissen
RP. Marginal maternal vitamin B12 status increases the risk of offspring with spina
bifida. Am J Obstet Gynecol 2004;191:11e17.
16. Candito M, Rivet R, Herbeth B, Boisson C, Rudigoz RC, Luton D, Journel H, Oury JF,
Roux F, Saura R, Vernhet I, Gaucherand P, Muller F, Guidicelli B, Heckenroth H,
Poulain P, Blayau M, Francannet C, Roszyk L, Brustie C, Staccini P, Gerard P,
Fillion-Emery N, Gueant-Rodriguez RM, Van Obberghen E, Gueant JL. Nutritional and
genetic determinants of vitamin B and homocysteine metabolisms in neural tube
defects: a multicenter case-control study. Am J Med Genet A 2008;146A:1128e33.
17. Molloy AM, Kirke PN, Troendle JF, Burke H, Sutton M, Brody LC, Scott JM, Mills JL.
Maternal vitamin B12 status and risk of neural tube defects in a population with high
neural tube defect prevalence and no folic acid fortification. Pediatrics
2009;123:917e23.
18. Gardiki-Kouidou P, Seller MJ. Amniotic fluid folate, vitamin B12 and
transcobalamins in neural tube defects. Clin Genet 1988;33:441e8.
19. Dawson EB, Evans DR, Van Hook JW. Amniotic fluid B12 and folate levels
associated with neural tube defects. Am J Perinatol 1998;15:511e14.
20. Economides DL, Ferguson J, Mackenzie IZ, Darley J, Ware II, Holmes-Siedle M.
Folate and vitamin B12 concentrations in maternal and fetal blood, and amniotic fluid
in second trimester pregnancies complicated by neural tube defects. Br J Obstet
Gynaecol 1992;99:23e5.
21. Steen MT, Boddie AM, Fisher AJ, Macmahon W, Saxe D, Sullivan KM, Dembure PP,
Elsas LJ. Neural-tube defects are associated with low concentrations of cobalamin
(vitamin B12) in amniotic fluid. Prenat Diagn 1998;18:545e55.
22. Weekes EW, Tamura T, Davis RO, Birch R, Vaughn WH, Franklin JC, Barganier C,
Cosper P, Finley SC, Finley WH. Nutrient levels in amniotic fluid from women with
normal and neural tube defect pregnancies. Biol Neonate 1992;61:226e31.
23. Ray JG, Wyatt PR, Thompson MD, Vermeulen MJ, Meier C, Wong PY, Farrell SA,
Cole DE. Vitamin B12 and the risk of neural tube defects in a folic-acidefortified
population. Epidemiology 2007;18:362e6.
24. Afman LA, Lievers KJ, van der Put NM, Trijbels FJ, Blom HJ. Single nucleotide
polymorphisms in the transcobalamin gene: relationship with transcobalamin
concentrations and risk for neural tube defects. Eur J Hum Genet 2002;10:433e8.
25. Quadros EV, Regec AL, Khan KM, Quadros E, Rothenberg SP. Transcobalamin II
synthesized in the intestinal villi facilitates transfer of cobalamin to the portal blood.
Am J Physiol 1999;277:G161e6.
26. Pietrzyk JJ, Bik-Multanowski M. 776C>G polymorphism of the transcobalamin II
gene as a risk factor for spina bifida. Mol Genet Metab 2003;80:364.
27. Swanson DA, Pangilinan F, Mills JL, Kirke PN, Conley M, Weiler A, Frey T, Parle-
McDermott A, O’Leary VB, Seltzer RR, Moynihan KA, Molloy AM, Burke H, Scott JM,
Brody LC. Evaluation of transcobalamin II polymorphisms as neural tube defect risk
factors in an Irish population. Birth Defects Res A Clin Mol Teratol 2005;73:239e44.
28. De Marco P, Merello E, Calevo MG, Mascelli S, Raso A, Cama A, Capra V.
Evaluation of a methylenetetrahydrofolate-dehydrogenase 1958G>A polymorphism
for neural tube defect risk. J Hum Genet 2006;51:98e103.
29. O’Leary VB, Mills JL, Pangilinan F, Kirke PN, Cox C, Conley M, Weiler A, Peng K,
Shane B, Scott JM, Parle-McDermott A, Molloy AM, Brody LC. Analysis of
methionine synthase reductase polymorphisms for neural tube defects risk
association. Mol Genet Metab 2005;85:220e7.
30. Boyles AL, Billups AV, Deak KL, Siegel DG, Mehltretter L, Slifer SH, Bassuk AG,
Kessler JA, Reed MC, Nijhout HF, George TM, Enterline DS, Gilbert JR, Speer MC.
Neural tube defects and folate pathway genes: family-based association tests of
gene-gene and gene-environment interactions. Environ Health Perspect
2006;114:1547e52.
31. Dunlevy LP, Chitty LS, Burren KA, Doudney K, Stojilkovic-Mikic T, Stanier P, Scott R,
Copp AJ, Greene ND. Abnormal folate metabolism in foetuses affected by neural
tube defects. Brain 2007;130:1043e9.
32. Brouns R, Ursem N, Lindemans J, Hop W, Pluijm S, Steegers E, Steegers-
Theunissen R. Polymorphisms in genes related to folate and cobalamin metabolism
and the associations with complex birth defects. Prenat Diagn 2008;28:485e93.
33. Pietrzyk JJ, Bik-Multanowski M, Sanak M, Twardowska M. Polymorphisms of the
5,10-methylenetetrahydrofolate and the methionine synthase reductase genes as
independent risk factors for spina bifida. J Appl Genet 2003;44:111e13.
34. van der Linden IJ, den Heijer M, Afman LA, Gellekink H, Vermeulen SH, Kluijtmans
LA, Blom HJ. The methionine synthase reductase 66A>G polymorphism is
a maternal risk factor for spina bifida. J Mol Med 2006;84:1047e54.
35. Doolin MT, Barbaux S, McDonnell M, Hoess K, Whitehead AS, Mitchell LE.
Maternal genetic effects, exerted by genes involved in homocysteine remethylation,
influence the risk of spina bifida. Am J Hum Genet 2002;71:1222e6.
36. Zhu H, Wicker NJ, Shaw GM, Lammer EJ, Hendricks K, Suarez L, Canfield M, Finnell
RH. Homocysteine remethylation enzyme polymorphisms and increased risks for
neural tube defects. Mol Genet Metab 2003;78:216e21.
37. Gueant-Rodriguez RM, Rendeli C, Namour B, Venuti L, Romano A, Anello G, Bosco P,
Debard R, Gerard P, Viola M, Salvaggio E, Gueant JL. Transcobalamin and methionine
Table 4 Haplotype frequency estimates of the TCblR D9block
Haplotype Controls NTD cases NTD mothers
GCTdelCGG 0.33 0.32 0.30
GGTdelCGG 0.24 0.24 0.24
AGCdelCGG 0.19 0.21 0.21
GGTdelCCG 0.10 0.10 0.11
GGCdelCGA 0.10 0.07 0.09
GCTdelTGG 0.02 0.03 0.02
GCTinsTGG 0.02 0.03 0.01
GGCdelCGG 0.01 0.01 0.01
p Value 0.3y1.0y
The haplotype block consists of the following markers: rs4147651, rs7408841, rs2927707,
E88del, rs2336573 (G220R), rs2227288 and rs173665.
yResult of the permutation test for significant differences in haplotype frequencies in the
NTD cases versus the controls or the NTD mothers versus the controls.
8 of 9 Pangilinan F, Mitchell A, VanderMeer J, et al.J Med Genet (2010). doi:10.1136/jmg.2009.073775
Original article
group.bmj.com on August 25, 2010 - Published by jmg.bmj.comDownloaded from
synthase reductase mutated polymorphisms aggravate the risk of neural tube defects
in humans. Neurosci Lett 2003;344:189e92.
38. Franke B, Vermeulen SH, Steegers-Theunissen RP, Coenen MJ, Schijvenaars MM,
Scheffer H, den Heijer M, Blom HJ. An association study of 45 folate-related genes in
spina bifida: Involvement of cubilin (CUBN) and tRNA aspartic acid methyltransferase
1 (TRDMT1). Birth Defects Res A Clin Mol Teratol 2009;85:216e26.
39. Quadros EV, Nakayama Y, Sequeira JM. The protein and the gene encoding the
receptor for the cellular uptake of transcobalamin-bound cobalamin. Blood
2009;113:186e92.
40. Shields DC, Kirke PN, Mills JL, Ramsbottom D, Molloy AM, Burke H, Weir DG, Scott
JM, Whitehead AS. The “thermolabile” variant of methylenetetrahydrofolate
reductase and neural tube defects: an evaluation of genetic risk and the relative
importance of the genotypes of the embryo and the mother. Am J Hum Genet
1999;64:1045e55.
41. Brody LC, Conley M, Cox C, Kirke PN, McKeever MP, Mills JL, Molloy AM, O’Leary
VB, Parle-McDermott A, Scott JM, Swanson DA. A polymorphism, R653Q, in the
trifunctional enzyme methylenetetrahydrofolate dehydrogenase/
methenyltetrahydrofolate cyclohydrolase/formyltetrahydrofolate synthetase is
a maternal genetic risk factor for neural tube defects: report of the birth defects
research group. Am J Hum Genet 2002;71:1207e15.
42. Pangilinan F, Geiler K, Dolle J, Troendle J, Swanson DA, Molloy AM, Sutton M,
Conley M, Kirke PN, Scott JM, Mills JL, Brody LC. Construction of a high resolution
linkage disequilibrium map to evaluate common genetic variation in TP53 and neural
tube defect risk in an Irish population. Am J Med Genet A 2008;146A:2617e25.
43. Meulenbelt I, Droog S, Trommelen GJ, Boomsma DI, Slagboom PE. High-yield
noninvasive human genomic DNA isolation method for genetic studies in
geographically dispersed families and populations. Am J Hum Genet
1995;57:1252e4.
44. Anon. The International HapMap Project. Nature 2003;426:789e96.
45. Barrett JC, Fry B, Maller J, Daly MJ. Haploview: analysis and visualization of LD and
haplotype maps. Bioinformatics 2005;21:263e5.
46. Stephens M, Smith NJ, Donnelly P. A new statistical method for haplotype
reconstruction from population data. Am J Hum Genet 2001;68:978e89.
47. Stephens M, Donnelly P. A comparison of Bayesian methods for haplotype
reconstruction from population genotype data. Am J Hum Genet 2003;73:1162e9.
48. Weinberg CR, Wilcox AJ, Lie RT. A log-linear approach to case-parent-triad data:
assessing effects of disease genes that act either directly or through maternal effects
and that may be subject to parental imprinting. Am J Hum Genet 1998;62:969e78.
49. Spielman RS, McGinnis RE, Ewens WJ. Transmission test for linkage disequilibrium:
the insulin gene region and insulin-dependent diabetes mellitus (IDDM). Am J Hum
Genet 1993;52:506e16.
50. Dempster A, Laird NM, Rubin DB. Maximum likelihood from incomplete data via EM
algorithm. J R Stat Soc Series B Stat Methodol 1977;39:1e38.
51. Kent WJ, Sugnet CW, Furey TS, Roskin KM, Pringle TH, Zahler AM, Haussler D. The
human genome browser at UCSC. Genome Res 2002;12:996e1006.
52. May P, Woldt E, Matz RL, Boucher P. The LDL receptor-related protein (LRP) family:
an old family of proteins with new physiological functions. Ann Med
2007;39:219e28.
Pangilinan F, Mitchell A, VanderMeer J, et al.J Med Genet (2010). doi:10.1136/jmg.2009.073775 9 of 9
Original article
group.bmj.com on August 25, 2010 - Published by jmg.bmj.comDownloaded from
... All five newborns were homozygous for CD320 p.E88del. Concurrently, this allele was also nominally associated with risk of an NTD (Pangilinan et al., 2010). The identification of this allele in independent studies bolsters the case for it being a functional variant, or closely linked to such a variant, with potential clinical consequences. ...
... An independent sample was selected to perform a replication study of the nominally significant association of CD320 p.E88del with NTD risk (Pangilinan et al., 2010). This is a population-based, case-control sam- LGC Genomics (Herts, UK) performed genotyping of CD320 p.E88del using KASP genotyping chemistry. ...
... As CD320 p.E88del was previously found to be nominally associated with NTDs in the Irish population (Pangilinan et al., 2010), we performed a replication study by genotyping this variant in an NTD cohort from New York State (Table 4). We did not observe any significant differences between cases and controls in any race/ethnicity, although power is low considering the low MAF of this variant and lower sample size of the replication cohort (less than half). ...
Article
The biological and clinical significance of the p.E88del variant in the transcobalamin receptor, CD320, is unknown. This allele is annotated in ClinVar as likely benign, pathogenic, and of uncertain significance. To determine functional consequence and clinical relevance of this allele, we employed cell culture and genetic association studies. Fibroblasts from 16 CD320 p.E88del homozygotes exhibited reduced binding and uptake of cobalamin. Complete ascertainment of newborns with transiently elevated C3 (propionylcarnitine) in New York State demonstrated that homozygosity for CD320 p.E88del was over‐represented (7/348, p < 6 × 10−5). Using population data, we estimate that ~85% of the p.E88del homozygotes born in the same period did not have elevated C3, suggesting that cobalamin metabolism in the majority of these infants with this genotype is unaffected. Clinical follow‐up of 4/9 homozygous individuals uncovered neuropsychological findings, mostly in speech and language development. None of these nine individuals exhibited perturbation of cobalamin metabolism beyond the newborn stage even during periods of acute illness. Newborns homozygous for this allele in the absence of other factors are at low risk of requiring clinical intervention, although more studies are required to clarify the natural history of various CD320 variants across patient populations.
... The high LD with the other We next used a second UK cohort to assess whether a similar significant TDT association would be replicated. This cohort has been previously described (Pangilinan et al., 2010 ...
... It is increasingly recognized that vitamin B 12 deficiency and genetic variants in the transcobalamin II receptor, which is required to transport vitamin B 12 into peripheral tissues (94), interact to increase the risk for folate-associated pathologies (149), and this may also be true for cerebral pathologies. Brain vitamin B 12 levels decrease with age and are also decreased in individuals with schizophrenia or autism (157). ...
Article
It is increasingly recognized that tissue-specific nutrient deficiencies can exist in the absence of whole-body deficiency and that these deficiencies may result from disease or disease-related physiological processes. Brain and central nervous system tissues require adequate nutrient levels to function. Many nutrients are concentrated in the cerebrospinal fluid relative to the serum in healthy individuals, and other nutrients resist depletion in the presence of whole-body nutrient depletion. The endothelial, epithelial, and arachnoid brain barriers work in concert to selectively transport, concentrate, and maintain levels of the specific nutrients required by the brain while also blocking the passage of blood-borne toxins and pathogens to brain and central nervous system tissues. These barriers preserve nutrient levels within the brain and actively concentrate nutrients within the cerebrospinal fluid and brain. The roles of physical and energetic barriers, including the blood–brain and blood–nerve barriers, in maintaining brain nutrient levels in health and disease are discussed. Expected final online publication date for the Annual Review of Nutrition Volume 39 is August 21, 2019. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
... For example, a mouse model creating a null mutation at the Mtr locus causes embryonic lethality between embryonic day (E) 4.5 and E8.5 (26) while targeted mutation of the Mut gene results in the perinatal death of homozygous KO mice (27,28). Human genetic studies found that a three-base pair (bp) deletion in the CD320 receptor protein, p.Glu88del (CD320 c.262_264delGAG), is associated with risk of NTDs in an Irish population (29) and correlates with elevated plasma acylcarnitine at birth but otherwise may be asymptomatic (30). A mouse containing a gene-trapped KO allele of Cd320 results in a nonlethal phenotype with moderately increased concentrations of plasma Hcy and MMA, reduced concentrations of serum cobalamin and greatly reduced cobalamin levels in the central nervous system (31,32). ...
Article
In humans, poor nutrition, malabsorption and variation in cobalamin (vitamin B12) metabolic genes are associated with hematological, neurological and developmental pathologies. Cobalamin is transported from blood into tissues via the transcobalamin (TC) receptor encoded by the CD320 gene. We created mice carrying a targeted deletion of the mouse ortholog, Cd320. Knockout (KO) mice lacking this TC receptor have elevated levels of plasma methylmalonic acid and homocysteine but are otherwise healthy, viable, fertile and not anemic. To challenge the Cd320 KO mice we maintained them on a vitamin B12-deficient diet. After 5 weeks on this diet, reproductive failure develops in Cd320 KO females but not males. In vitro, homozygous Cd320 KO embryos from cobalamin-deficient Cd320 KO dams develop normally to embryonic day (E) 3.5, while in vivo, few uterine decidual implantation sites are observed at E7.5, suggesting that embryos perish around the time of implantation. Dietary restriction of vitamin B12 induces a severe macrocytic anemia in Cd320 KO mice after 10-12 months while control mice on this diet are anemia-free up to 2 years. Despite the severe anemia, cobalamin-deficient KO mice do not exhibit obvious neurological symptoms. Our results with Cd320 KO mice suggest that an alternative mechanism exists for mice to transport cobalamin independent of the Cd320 encoded receptor. Our findings with deficient diet are consistent with historical and epidemiological data suggesting that low vitamin B12 levels in humans are associated with infertility and developmental abnormalities. Our Cd320 KO mouse model is an ideal model system for studying vitamin B12 deficiency.
Chapter
Vitamin B12 (cobalamin, Cbl) is a cofactor for just two enzymes, while various forms of folate are coenzymes for a wide range of enzymes. Metabolism of both vitamins involve complex absorption and transport systems and multiple intracellular conversions to active cofactor forms. Methylmalonic aciduria and associated clinical abnormalities are features of defects in synthesis of the adenosylcobalamin cofactor form, while homocystinuria with megaloblastic anaemia and neurological disorders are clinical features of patients with defects methylcobalamin synthesis, with both of these found to various degrees of severity in combined intracellular defects and disorders of Cbl absorption and transport. The most common inherited folate disorder, severe MTHFR deficiency, presents with homocystinuria and neurological abnormalities. In this chapter, clinical presentation, metabolic derangement, genetics, diagnostic test and treatment and prognosis are described for defects in cobalamin and folate metabolism.
Chapter
Cobalamin (vitamin B12) is required for activity of the enzymes methylmalonyl-CoA mutase and methionine synthase in human cells. Inborn errors affecting cobalamin uptake or metabolism are characterized by accumulation of the substrates for these enzymes, methylmalonic acid and homocysteine, in blood and urine. Inborn errors affecting synthesis of the adenosylcobalamin coenzyme required by methylmalonyl-CoA mutase (cblA and cblB) result in isolated methylmalonic aciduria; inborn errors affecting synthesis of the methylcobalamin coenzyme required by methionine synthase (cblE and cblG) result in isolated homocystinuria. Combined methylmalonic aciduria and homocystinuria is seen in patients with impaired intestinal cobalamin absorption (intrinsic factor deficiency, Imerslund-Gräsbeck syndrome) and with defects affecting synthesis of both cobalamin coenzymes (cblC, cblD, cblF and cblJ). A series of disorders caused by pathogenic variant mutations affecting gene regulators (transcription factors) of the MMACHC gene have recently been described (HCFC1 [cblX disorder] and deficiencies of THAP11, and ZNF143 [the cblK disorder]).
Article
We report seven cases from our clinic with transcobalamin receptor deficiency (TCRD). None of our cases have experienced health issues or metabolic decompensation. All have experienced typical growth and development throughout childhood, with our oldest case now 10 years old. Every case has had normalization of initial biochemical abnormalities following parenteral hydroxocobalamin administration. Several cases had trace elevations of methylmalonic acid throughout childhood, all which normalized without further hydroxocobalamin administration. Population data from our state's newborn screening program suggest the incidence of TCRD is comparable to other metabolic disorders associated with elevations of C3 acylcarnitine including propionic academia, isolated methylmalonic academia and combined methylmalonic academia and hyperhomocysteinemia due to cobalamin metabolism disorders. Based on the generally benign nature of this condition, we assert that TCRD may be considered an incidental finding on newborn screen. However, additional long-term data are needed to ascertain the long term outcomes of children identified with TCRD.
Chapter
Folate and cobalamin (vitamin B12) are B group vitamins that play interacting roles in cellular metabolism. Hereditary folate malabsorption results in a specific deficiency of folate transport across the intestine and across the blood–brain barrier. Glutamate formiminotransferase deficiency has been variably described as having phenotypes ranging from severe neurologic disease to benign excretion of formiminoglutamate. Methylenetetrahydrofolate reductase deficiency is the most common and best‐characterized inborn error of folate metabolism. Transcobalamin deficiency is associated with megaloblastic anemia and pancytopenia, and with immunologic, gastrointestinal, and mental disorders. Prenatal therapy has been attempted for several fetuses with cobalamin‐responsive methylmalonic aciduria using vitamin B12 or OHCbl, administered intramuscularly or orally to the mother. Prenatal therapy was also performed on a fetus with the cblE disorder, with 1 mg of hydroxocobalamin administered intramuscularly weekly to the mother.
Article
Neural tube defects (NTDs) are birth defects that arise during embryogenesis when normal neural tube closure fails to occur. According to the World Health Organization, NTDs are detected annually in approximately 300,000 neonates worldwide. The exact etiology of NTDs remains complex and poorly understood. It is generally agreed that most NTD cases are of multifactorial origin, having a combination of multiple genes and a number of environmental risk factors. The role of folic acid, vitamin B12 deficiency, genetics and other risk factors, in the etiology of NTDs, have also been extensively studied. This knowledge synthesis brings together different types of evidence to update the role of vitamin B12 deficiency, genetics and other risk factors, in the etiology of NTDs. Following a PubMed search and screening for relevant articles, we included 40 studies in our review (30 case control studies, 3 cross‐sectional studies, 5 cohort studies, and 2 case reports). The available data showed that vitamin B12 levels were decreased in mothers and infants in NTD groups compared to control groups. Holo‐transcobalamin, the active form of vitamin B12, was also found in lower levels in mothers with NTD‐affected infants. Several studies reported elevated homocysteine levels in mothers and infants in NTD groups. Additionally, numerous studies reported links between genetic variants and increased NTD risk. These genes include GIF, LRP2, CUBN, TCb1R, MTHFR, and others. Several maternal factors have also been linked with significant NTD risk such as BMI, maternal diet, air pollutants, low maternal age, and many others. The majority of studies on NTDs have focused on the role of folic acid, hence there is a need for well‐designed studies on the role of other risk factors like vitamin B12 deficiency in the etiology of NTDs.
Article
Very few cases of craniorachischisis (CRN) with concomitant omphalocele (OMP) in the setting of trisomy 18 are reported in literature. Solitary midline closure defects are estimated to be more prevalent in trisomy 18 compared to the general population. Neurulation defect comparisons include anencephaly 0–2% versus 0.0206%, spina bifida 1–3% versus 0.0350%, and encephalocele 0–2% versus 0.0082% [Parker et al. (2010); Birth Defects Research. Part A: Clinical and Molecular Teratology, 88:1008–1016; Springett et al. (2015); American Journal of Medical Genetics. Part A, 167A:3062–3069]. The solitary anterior malformation OMP has been reported as high as 6% with trisomy 18 [Springett et al. (2015); American Journal of Medical Genetics. Part A, 167A:3062–3069]. We report the third published case of CRN with concomitant OMP observed in a likely trisomy 18 fetus that screened positive by noninvasive prenatal screening. Furthermore, we review and analyze the current literature to augment understanding of the genetic basis for anterior and posterior closure defects such as CRN and OMP. Although the current genetic lexicon lacks any definitive association with the simultaneous defects presented, previous research elucidated various genes related to anterior or posterior closure interruption individually. By consolidating current research, the authors advance knowledge of interconnected genetic pathology and direct future genetic mapping efforts.
Article
Full-text available
The goal of the International HapMap Project is to determine the common patterns of DNA sequence variation in the human genome and to make this information freely available in the public domain. An international consortium is developing a map of these patterns across the genome by determining the genotypes of one million or more sequence variants, their frequencies and the degree of association between them, in DNA samples from populations with ancestry from parts of Africa, Asia and Europe. The HapMap will allow the discovery of sequence variants that affect common disease, will facilitate development of diagnostic tools, and will enhance our ability to choose targets for therapeutic intervention.
Article
This study was designed to identify the cellular component of the intestinal villus where transcobalamin II (TCII) is synthesized, because this protein provides an essential function in the intestinal absorption of vitamin B-12 (cobalamin, Cbl). When a segment of proximal or distal small intestine of the guinea pig is cultured in medium containing [Co-57]Cbl, TCII[Co-57]Cbl appears within 15 min. Northern blot analysis of RNA from both proximal and distal small intestine identified the TCII transcript. In situ hybridization of the distal ileum with S-35-labeled TCII antisense transcript localized grains predominantly in crypts and in the lower third and central core of the villi. Grains were also evident at the base of the enterocytes in close apposition with the vascular network, whereas few grains appeared in the apical region of the columnar cells. This study provides evidence that TCII is constitutively expressed in the intestinal villi where vascular endothelium is abundant. In the distal ileum, where the intrinsic factor (IF) receptor is expressed, after uptake of IF-Cbl and the subsequent binding of free Cbl to TCII synthesized in the villi, the TCII-Cbl complex enters the microcirculation and passes into the portal blood.
Article
Each year, more than 4500 pregnancies in the European Union are affected by neural tube defects (NTD). Unambiguous evidence of the effectiveness of periconceptional folic acid in preventing the majority of neural tube defects has been available since 1991. We report on trends in the total prevalence of neural tube defects up to 2002, in the context of a survey in 18 European countries of periconceptional folic acid supplementation (PFAS) policies and their implementation. EUROCAT is a network of population-based registries in Europe collaborating in the epidemiological surveillance of congenital anomalies. Representatives from 18 participating countries provided information about policy, health education campaigns and surveys of PFAS uptake. The yearly total prevalence of neural tube defects including livebirths, stillbirths and terminations of pregnancy was calculated from 1980 to 2002 for 34 registries, with UK and Ireland estimated separately from the rest of Europe. A meta-analysis of changes in NTD total prevalence between 1989-1991 and 2000-2002 according to PFAS policy was undertaken for 24 registries. By 2005, 13 countries had a government recommendation that women planning a pregnancy should take 0.4mg folic acid supplement daily, accompanied in 7 countries by government-led health education initiatives. In the UK and Ireland, countries with PFAS policy, there was a 30% decline in NTD total prevalence (95% CI 16-42%) but it was difficult to distinguish this from the pre-existing strong decline. In other European countries with PFAS policy, there was virtually no decline in NTD total prevalence whether a policy was in place by 1999 (2%, 95% CI 28% reduction to 32% increase) or not (8%, 95% CI 26% reduction to 16% increase). The potential for preventing NTDs by periconceptional folic acid supplementation is still far from being fulfilled in Europe. Only a public health policy including folic acid fortification of staple foods is likely to result in large-scale prevention of NTDs.
Article
A randomised double-blind prevention trial with a factorial design was conducted at 33 centres in seven countries to determine whether supplementation with folic acid (one of the vitamins in the B group) or a mixture of seven other vitamins (A,D,B1,B2,B6,C and nicotinamide) around the time of conception can prevent neural tube defects (anencephaly, spina bifida, encephalocele). A total of 1817 women at high risk of having a pregnancy with a neural tube defect, because of a previous affected pregnancy, were allocated at random to one of four groups--namely, folic acid, other vitamins, both, or neither. 1195 had a completed pregnancy in which the fetus or infant was known to have or not have a neural tube defect; 27 of these had a known neural tube defect, 6 in the folic acid groups and 21 in the two other groups, a 72% protective effect (relative risk 0.28, 95% confidence interval 0.12-0.71). The other vitamins showed no significant protective effect (relative risk 0.80, 95% Cl 0.32-1.72). There was no demonstrable harm from the folic acid supplementation, though the ability of the study to detect rare or slight adverse effects was limited. Folic acid supplementation starting before pregnancy can now be firmly recommended for all women who have had an affected pregnancy, and public health measures should be taken to ensure that the diet of all women who may bear children contains an adequate amount of folic acid.
Article
We analyzed nutrient levels in amniotic fluid obtained during the second trimester of normal, uncomplicated pregnancies from 221 women who delivered apparently healthy infants and from 8 with neural tube defect (NTD) pregnancies. Folate was measured by microbiological assay, vitamin B12 by a radiobinding method, and zinc, copper and iron by atomic absorption spectrophotometry. We found that the mean amniotic fluid nutrient levels of normal pregnancies were 24.7nmol/l for folate, 600pmol/l for vitamin B12, and 1.7, 1.9, and 9.0 μmol/l for zinc, copper and iron, respectively. Amniotic fluid folate, zinc, copper and iron levels of NTD pregnancies were similar to those found during normal pregnancy, however, vitamin B12 levels were markedly lower than those of normal pregnancies.
Article
Spina bifida is a class of neural tube defects, which are congenital malformations of the central nervous system with a prevalence of 0.5 to 12 per 1000 births globally. In this article we attempt to identify genes related to folate and its metabolic pathways that are involved in the etiology of spina bifida. We selected 50 folate metabolism–related genes and genotyped polymorphisms in those genes. Eighty-seven polymorphisms in 45 genes passed quality controls. Associations with spina bifida were investigated in 180 patients and 190 controls. For those polymorphisms that were nominally associated with spina bifida risk, the relation with serum and red blood cell folate, vitamin B12, and homocysteine was evaluated in controls. A polymorphism in CUBN was significantly associated with decreased spina bifida risk, after correction for multiple testing, and was related to increased vitamin B12 (p = 0.039) and red blood cell folate (p = 0.001). The CUBN gene encodes the intrinsic factor–cobalamin receptor (or cubilin), a peripheral membrane protein that acts as a receptor for intrinsic factor–vitamin B12 complexes. Vitamin B12 is an important cofactor in the folate metabolism, and low B12 status in mothers has been linked to neural tube defects in children. Other interesting findings include nominally significant associations with polymorphisms in TRDMT1, ALDH1L1, SARDH, and SLCA19A1 (RFC1). Our study indicates interesting new candidate genes and functional pathways for further study and confirms earlier findings. None of the genes CUBN, TRDMT1, ALDH1L1, or SARDH have been investigated previously for association with spina bifida. Birth Defects Research (Part A) 2009.
Article
S ummary A broadly applicable algorithm for computing maximum likelihood estimates from incomplete data is presented at various levels of generality. Theory showing the monotone behaviour of the likelihood and convergence of the algorithm is derived. Many examples are sketched, including missing value situations, applications to grouped, censored or truncated data, finite mixture models, variance component estimation, hyperparameter estimation, iteratively reweighted least squares and factor analysis.
Article
We describe a log-linear method for analysis of case-parent-triad data, based on maximum likelihood with stratification on parental mating type. The method leads to estimates of association parameters, such as relative risks, for a single allele, and also to likelihood ratio chi2 tests (LRTs) of linkage disequilibrium. Hardy-Weinberg equilibrium need not be assumed. Our simulations suggest that the LRT has power similar to that of the chi2 "score" test proposed by Schaid and Sommer and that both can outperform the transmission/disequilibrium test (TDT), although the TDT can perform better under an additive model of inheritance. Because a restricted version of the LRT is asymptotically equivalent to the TDT, the proposed test can be regarded as a generalization of the TDT. The method that we describe generalizes easily to accommodate maternal effects on risk and, in fact, produces powerful and orthogonal tests of the contribution of fetal versus maternal genetic factors. We further generalize the model to allow for effects of parental imprinting. Imprinting effects can be fitted by a simple, iterative procedure that relies on the expectation-maximization algorithm and that uses standard statistical software for the maximization steps. Simulations reveal that LRT tests for detection of imprinting have very good operating characteristics. When a single allele is under study, the proposed method can yield powerful tests for detection of linkage disequilibrium and is applicable to a broader array of causal scenarios than is the TDT.