Sie sind auf Seite 1von 20

N U TR IT ION RE S EA R CH 6 3 ( 20 1 9 ) 1– 2 0

Available online at www.sciencedirect.com

ScienceDirect
www.nrjournal.com

Review Article

Maternal risk factors and newborn infant vitamin


D status: a scoping literature review

Olusola F. Sotunde 1 , Alexandra Laliberte 1 , Hope A. Weiler⁎


School of Human Nutrition, McGill University Ste Anne de Bellevue, Québec, Canada H9X 3V9

ARTI CLE I NFO A BS TRACT

Article history: Low vitamin D (VitD) status is common among newborn infants, more so in temperate latitudes with
Received 18 September 2018 evidence that maternal VitD deficiency is a major risk factor given that the neonate relies solely on
Revised 19 November 2018 maternal-fetal transfer of VitD. This scoping review was conducted to provide an overview of the
Accepted 30 November 2018 latest evidence from studies regarding the impact of maternal risk factors on infant 25-
hydryoxyvitamin D [25(OH)D] concentrations with a focus on studies in Canada and the United
Keywords: States. Several maternal risk factors that contribute to low maternal-fetal 25(OH)D concentrations
Pregnancy have been reported over many decades, but no clear pattern has been established for multiethnic
Neonates populations. For example, darker skin pigmentation and ethnicity are common risk factors for low
Vitamin D status VitD status. Studies in predominantly white women showed that supplementation of VitD during
Risk factors pregnancy causes significant increases in maternal serum 25(OH)D which often improves cord
serum 25(OH)D values. In addition, VitD recommendations by health care professionals and
adherence to supplementation by pregnant women appear to positively influence maternal and
infant 25(OH)D concentrations. Conversely, winter season, obesity, lower socioeconomic status
including lifestyle factors (smoking), and use of medication pose risk for lower maternal-fetal
transfer of VitD. However, there is still a dearth of pertinent data on the relationship between some of
the maternal risk factors and newborn 25(OH)D concentrations, for instance, relationships between
gestational diabetes and neonatal VitD status. Additional research is required to determine if the
same target for 25(OH)D concentrations applies for pregnant women, neonates, and infants.
Crown Copyright © 2018 Published by Elsevier Inc. All rights reserved.

Article Outline

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Approach . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
3. Vitamin D metabolism in fetus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3.1. Consequences of vitamin D deficiency during pregnancy and infancy . . . . . . . . . . . . . . . . . . . . . . . . . 3

Abbreviations: 1,25(OH)2D, 1,25-dihydroxyvitamin D; 25(OH)D, 25-hydroxyvitamin D; BMI, body mass index; DRI, Dietary Reference
Intake; GDM, gestational diabetes mellitus; IOM, Institute of Medicine; MIREC, Maternal-Infant Research on Environmental Chemicals;
NHANES, National Health and Nutrition Examination Survey; PIH, pregnancy-induced hypertension; RDA, Recommended Dietary
Allowance; SES, socioeconomic status; VDBP, vitamin D–binding protein; VitD, vitamin D.
⁎ Corresponding author.
E-mail addresses: olusola.sotunde@mcgill.ca (O.F. Sotunde), alexandra.laliberte@mail.mcgill.ca (A. Laliberte), hope.weiler@mcgill.ca
(H.A. Weiler).
1
Authors contributed equally.

https://doi.org/10.1016/j.nutres.2018.11.011
0271-5317/Crown Copyright © 2018 Published by Elsevier Inc. All rights reserved.
2 N U TR IT ION RE S EA R CH 63 ( 20 1 9 ) 1 – 2 0

4. Maternal influence and risk factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3


4.1. Recommendations. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
4.1.1. Definitions and ranges of vitamin D . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
4.1.2. Dietary recommendations. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
4.2. Dietary intake and use of supplements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
4.2.1. Dietary intake . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
4.2.2. Use of supplements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
4.2.3. Population statistics on vitamin D supplementation of infants . . . . . . . . . . . . . . . . . . . . . . 12
4.3. Maternal vitamin D status . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
4.4. Sunlight exposure, ethnicity, and skin pigmentation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
4.5. Seasonal variations and clothing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
4.6. Professional opinion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
4.7. SES and education . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
4.8. Maternal morbidities/pregnancy complications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
4.8.1. Body mass index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
4.8.2. Hypertensive disease in pregnancy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
4.8.3. Gestational diabetes mellitus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
4.9. Lifestyle factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
4.9.1. Smoking status. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
4.9.2. Use of medications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
5. Knowledge gaps . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
6. Summary and conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
Acknowledgment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16

1. Introduction the production of 1,25(OH)2D during pregnancy, an approxi-


mate circulating 25(OH)D precursor concentration of 100 nmol/
As a result of high prevalence rates of vitamin D (VitD) deficiency L is reportedly required [13].
among children and the persistent incidence of rickets, several VitD status is not routinely tested in pregnant women or
upward revisions of recommended daily intakes of VitD have neonates because of its relatively high cost to health care.
occurred [1,2]. The National Academy of Science, formerly known Two pregnancy cohorts in Canada measured VitD status
as the Institute of Medicine (IOM), updated the VitD Recommended in infants, suggesting that approximately 24.4% of infants are
Dietary Allowance (RDA) in 2011 for pregnant and lactating born with a serum 25(OH)D concentration below 50 nmol/L
women to 600 IU. For infants, the Adequate Intake was increased [14], and 80% infants are born with 25(OH)D concentration
to 400 IU to achieve and maintain VitD serum/plasma concen- below 75 nmol/L [15]. In a US cohort study, 45.6% of black and
tration of at least 40 nmol/L of 25-hydroxyvitamin D [25(OH)D] [2] 9.7% of white infants are born with 25(OH)D below 37.5 nmol/L
and to prevent VitD deficiency and rickets [3]. VitD is of key using the 1997 IOM definition of VitD deficiency at the time of
importance for calcium and bone metabolism during pregnancy reporting [16]. According to the Canadian Paediatric Society,
and lactation for both the mother and neonate(s). VitD also has infants are at higher risk of VitD deficiency [<25 nmol/L of 25
nonskeletal effects, including its role in the development of (OH)D] if they are breast-fed, their mother has low VitD
healthy body composition [4-6] and improved birth outcomes [7]. status, their skin pigmentation is darker, and they live in
During pregnancy, VitD metabolism and calcium homoeostasis northern communities or are Aboriginal [17,18]. However, as
are regulated by many factors, including prolactin, placental maternal 25(OH)D concentration is not routinely known and
lactogen, osteoprotegerin, calcitonin, and estrogen [8,9]. Be- other previously identified risk factors are not fully eluci-
cause maternal-fetal transfer of VitD is the only source for the dated, some of the population at risk of low or deficient 25
fetus, a brief review of maternal physiology is warranted. (OH)D concentrations at birth might be unidentified. There-
Significant changes occur during pregnancy, starting with the fore, the main objective of this literature review is to shed
enhanced conversion of 25(OH)D to 1,25-dihydroxyvitamin D light on the various maternal factors that could impact infant
[1,25(OH)2D]. Free 1,25(OH)2D concentrations increase as does 25(OH)D concentrations.
vitamin D–binding protein (VDBP) [10]. In the first trimester,
maternal concentrations of 1,25(OH)2D double and continue to
increase to 3-fold the prepregnancy concentration [10]. This 2. Approach
increase in 1,25(OH)2D contributes to the increase in intestinal
calcium absorption to meet the fetal demand for mineralization Literature searches were conducted using PubMed and Google
of the skeleton [11]. An increase in VDBP observed in pregnancy Scholar databases for studies published before July 31, 2018.
is equally significant, as one of the major roles of VDBP is to The search terms included VitD, 25-hydroxyvitamin D, 25(OH)D,
bind and transport 25(OH)D and 1,25(OH)2D [12]. To optimize umbilical cord serum 25(OH)D, VitD deficiency, hypovitaminosis D,
N U TR IT ION RE S EA R CH 6 3 ( 20 1 9 ) 1– 2 0 3

VitD synthesis, prenatal VitD exposure, Canada, United States, fed, with only 5% of the breast-fed children given VitD
pregnancy, fetus, infant, birthweight, infant length, rickets, bone supplementation during breast-feeding [28].
quality, supplementation, mother-newborn pair, maternal risk Furthermore, proxies for infant health have been reported
factors, maternal VitD status, obesity, smoking, skin pigmentation, to be influenced by maternal 25(OH)D concentrations [29-31].
ethnicity, SES, education, dietary intake, pre-pregnancy body mass A post hoc analysis of 2 pregnancy supplementation trials
index (BMI), sun exposure and screening tool. Articles retrieved reported a 59% lower risk of preterm birth in women with 25
were evaluated on their relevance to the topic of this review, and (OH)D concentrations ≥100 nmol/L compared to pregnant
studies on nonhumans, those that used nonblood measures of 25 women with ≤50 nmol/L (P = .02) [32]. Based on this finding,
(OH)D (placenta or amniotic fluid), and those published in non- a study of 1064 women was carried out at the Medical
English languages were excluded. Additional relevant studies University of South Carolina, USA, to investigate if a similar
were included as identified from reference lists of primary result would be observed in their general obstetrical popula-
papers, review articles, and meta-analyses. tion [33]. The study reported a 62% reduction in risk of
preterm birth in women with 25(OH)D ≥ 100 nmol/L com-
pared to those with ≤50 nmol/L (P < .0001) [33]. Robinson et al
3. Vitamin D metabolism in fetus [29] found a positive correlation between maternal 25(OH)D
concentrations and birthweight centiles. This finding corrob-
Fetal VitD requirements are higher in the latter half of orates other studies including meta-analyses, which reported
pregnancy because of bone growth and ossification and to a higher risk of having a small-for-gestational-age infant for
build VitD stores for use postnatally. Placental transfer of 25 women with VitD deficiency [30,31], whereas others observed
(OH)D is the major source of VitD for the developing fetus [19] that mean birthweight in infants of deficient mothers was
and crosses the hemochorial human placenta as suggested by 200 g lower than neonates born to women with sufficient VitD
positive correlations between cord blood 25(OH)D and mater- status [34]. Furthermore, a multicentered US cohort study
nal concentrations [20,21]. Therefore, sufficient maternal VitD reported that infants with maternal 25(OH)D < 30 nmol/L
stores are important to meet their own needs and those of the were smaller at birth compared to infants with maternal 25
fetus [18]. Nevertheless, limited research is available regard- (OH)D ≥ 30 nmol/L [35]. In contrast, an earlier Canadian study
ing fetal development and VitD metabolism. found that VitD-deficient [25(OH)D < 27.5 nmol/L] infants
were heavier and longer but had lower whole-body bone
mineral content relative to body weight compared to those
3.1. Consequences of vitamin D deficiency during pregnancy with higher concentrations, suggesting inadequate support
and infancy for bone mineralization when deficient [36]. Wheeze and
asthma in the offspring have also been reported as outcomes
VitD insufficiency has been associated with impaired growth associated with low VitD intake during pregnancy [37-39]. The
and bone development [22]. A cohort study of mother- Vitamin D Antenatal Asthma Reduction Trial reported a
offspring pairs showed the adverse effect of low maternal significantly decreased risk of asthma/recurrent wheeze in
VitD status on fetal femoral development, which is evident as children at 3 years of age for every 12.5-nmol/L increase in
splaying of the distal metaphysis of the femur and seen as maternal 25(OH)D (adjusted odds ratio [OR], 0.92; 95% confi-
early as 19 weeks of gestation [22]. Less optimal intrauterine dence interval [CI], 0.85-0.99; P = .02) [39]. Consequently, VitD
bone growth may be explained by impaired placental calcium deficiency during pregnancy is recognized for its immediate
exchange, which might be mediated by parathyroid-related and long-term implications to health of the offspring. Few
hormone peptide [23]. Congenital rickets is an example of studies exist on the prevalence of low VitD status in neonates
extremely adverse effect of low VitD stores in utero, resulting in Canada and United States [14-16,36,40-42] (Fig. 1).
in defective bone modeling [24] and a higher incidence of
neonatal morbidity [25]. Attributable to VitD deficiency,
postnatal rickets can be prevented with adequate nutritional 4. Maternal influence and risk factors
intake of VitD. Unfortunately, subclinical rickets is difficult to
detect early after birth, whereas the peak manifestation of Fetal health is related to the ability of the mother to provide
fulminant rickets is typically observed between 3 and adequate nutrients to their developing child [43], and nutri-
18 months of age [24]. tional status of women before and/or during pregnancy can
In Canada, the estimated annual incidence rate of rickets significantly influence maternal-fetal outcomes [44,45]. Sev-
is 2.9 cases per 100 000 occurring between 2 weeks and eral factors have been reported to influence VitD production
6 years of age. The highest rates of incidence occur among and stores in adults.
infants with darker skin pigmentation, from northern com-
munities, and with a lack of VitD supplementation [26]. The 4.1. Recommendations
US Centers for Disease Control and Prevention in 2001
identified 6 cases of nutritional rickets in infants who were 4.1.1. Definitions and ranges of vitamin D
breast-fed for over 6 months without receiving VitD supple- Serum 25(OH)D concentration is considered to be the best
ments [27]. A review of nutritional rickets case reports among indicator of VitD status because it reflects the exogenous VitD
US children <18 years identified 166 cases of rickets in 22 input from the diet and supplements plus endogenous
published studies between 1986 and 2003 [28]. Most (83%) of production [46]. Lack of consensus and definitions about
the children were African Americans and 96% were breast- describing VitD status has generated various terminologies and
4 N U TR IT ION RE S EA R CH 63 ( 20 1 9 ) 1 – 2 0

Fig. 1 – Prevalence of low vitamin D status in neonates in Canada and United States. Data to create this illustration for different regions
of Canada and the United States are indicated in the reference number in brackets from the literature cited in this review.

ranges that have created challenges in this field of research [47]. meets the needs of the population [46]. For the United States and
The different threshold levels used in different regions increase Canada, the 2011 Dietary Reference Intakes (DRIs) for VitD by the
the difficulty of comparing results [48]. Despite the variable IOM were set considering minimal sunlight exposure and for the
definitions over time, the value of 50 nmol/L is now dominant in purpose of maintaining bone health [46]. In the updated DRIs of
many recommendations (Table 1). For example, the IOM defined 2011, the Committee generated an RDA for pregnant and lactating
VitD sufficiency as concentrations of serum 25(OH)D ≥ 50 nmol/L women and Adequate Intakes for infants [3].
[3], but others reported that the target should be 75 nmol/L Health Canada recommends consumption of 2 cups of
[49,50]. A study on VitD status of infants showed that 50 nmol/L fluid milk or VitD-fortified soy beverages for all individuals
was enough for bone health [51], but the Endocrine Society sets aged over 2 years, including pregnant and lactating women
the target at 75 nmol/L to maximize bone health and muscle [46]. Two cups of fluid milk will provide 206-210 IU of VitD,
function [52]. At 75 nmol/L, it was also found to be associated whereas 2 cups of VitD-fortified soy beverages provide 172 IU
with fat mass and lean mass, suggesting that this concentration VitD [25]. Fluid milk is also fortified (discretionary) to provide
may be important for leaner body composition during infancy [4]. 100 IU VitD per cup in the United States [3]. However, because
The Canadian Paediatric Society defined VitD deficiency as of this vitamin's limited availability in food sources, reliance
<25 nmol/L [18], whereas the IOM, Health Canada, and Endocrine on supplementation is common for individuals to reach the
Society state that infants are at risk of VitD deficiency at serum 25 RDA. Various dosages of VitD in supplements can be found,
(OH)D < 30 nmol/L [3,46,52]. ranging from 200 to 1000 IU per tablet [3,25]. VitD
biofortification of foods by adding VitD and/or 25(OH)D to
4.1.2. Dietary recommendations livestock feeds to increase the VitD/25(OH)D content of the
Given the evidence on the role of VitD on maternal-fetal animal produce or by the use of UV radiation to enhance the
outcomes, specific recommendations have emerged to main- VitD content of foods like mushrooms and baker's yeast is an
tain maternal and infant VitD status within healthy target emerging means of food-based solutions to tackling VitD
ranges. In the absence of UVB exposure, maternal VitD status deficiency [53,54].
is entirely dependent on storage pools and all sources of For pregnant women, the need and effectiveness of taking
exogenous VitD, be it from natural food sources, fortified a VitD supplement in addition to the diet remain controver-
foods, or supplements. VitD from endogenous and exogenous sial in the literature, and most recommendations target other
sources is a prohormone which can be stored for later use [3]. nutrients, such as iron and folic acid [55-57]. In 1999, the
According to food fortification policy, VitD remains a vitamin, and United Nations International Children's Emergency Fund,
it is a topic of policy revisions to ensure that food supply better United Nations University, and World Health Organization
Table 1 – Vitamin D levels, definitions, and recommendations according to different worldwide organizations
Organizations Countries following Vitamin D levels and specifications a Categories DRIs (UL)
these thresholds (nmol/L)

Institutes of Medicine [46,143] United States, Canada Risk of deficiency: <30 Infants 0-6 mo 400 IU b (1000 IU)
Insufficiency: 30-50 Infants 6-12 mo 400 IU b (1500 IU)
Sufficient: ≥50 Pregnant and lactating women 600 IU b (4000 IU)
United Kingdom National United Kingdom Risk of deficiency: <30 Infants 0-6 mo Unspecified
Osteoporosis Society [144] Insufficiency: 30-50 Infants 6-12 mo Unspecified

N U TR IT ION RE S EA R CH 6 3 ( 20 1 9 ) 1– 2 0
Adequacy: ≥50 Pregnant and lactating women Unspecified
Endocrine Society [145] Poland, Hungary, Belarus, Deficiency: <50 Infants 0-6 mo Supplementation 400 IU/d (1000 IU)
Estonia, Czech Republic, Insufficiency: 50-75 Infants 6-12 mo 400-600 IU/d (1000 IU)
Ukraine Adequacy: 75-125 Pregnant and lactating women Supplementation 1500-2000 IU/d (4000 IU)
German Nutrition Society [146] German, Austria, Switzerland Optimal: ≥50 Infants 0-6 mo 800 IU/d
Infants 6-12 mo 800 IU/d
Pregnant and lactating women 800 IU/d
Nordic Nutrition Recommendations Denmark, Finland, Iceland, Insufficiency: <50 Infants 0-6 mo 400 IU/d
2012 [147] Sweden, Norway, the Faroe Adequacy: 50 Infants 6-12 mo 400 IU/d
Island, Greenland, Aaland Pregnant and lactating women 400 IU/d (4000 IU)
Health Council of the The Netherlands Adequacy for people aged between 4 and 70 y: ≥30 Infants 0-6 mo 400 IU/d b
Netherlands [148] Adequacy for people aged 70 y and over: ≥50 Infants 6-12 mo 400 IU/d b
Pregnant and lactating women 400 IU/d b
Australian and New Zealand Australia, New Zealand Mild deficiency: 25-50 Infants 0-6 mo Unspecified
Bone Mineral Society, Endocrine Moderate deficiency: 12.5-25 Infants 6-12 mo Unspecified
Society of Australia, and Severe deficiency: <12.5 Pregnant and lactating women Unspecified
Osteoporosis Australia [149,150]
Canadian Paediatric Society [18] Canada For pregnant/lactating women and infants Infants 0-6 mo 400-800 c IU/d (1000 IU/d)
Deficiency: <25 Infants 6-12 mo 400-800 c IU/d (1500 IU/d)
Insufficiency: 25-75 Pregnant and lactating women 600 IU/d (4000 IU/d)
Adequacy: 75-225

D: day; IU: international units; mo: months; UL: Tolerable Upper Intake Level.
a
For concentrations of serum 25(OH)D.
b
Adequate intakes.
c
Infants living in northern communities (north of 55° latitude) or with dark skin should get 800 IU/d between October and April.

5
6
Table 2 – Studies on maternal and neonatal risk factors for low neonatal vitamin D stores
Risk factors Authors, year Sample size and study location Intervention or assessment Outcomes

Maternal Dror et al, 2012 [151] 80 mother-newborn pairs Maternal venous blood collected at birth Maternal 25(OH)D concentrations was lower than
vitamin D (40 African Americans and admission and cord blood collected immediately umbilical cord concentrations (40.9 nmol/L vs
status 40 non–African Americans) postdelivery for serum 25(OH)D assays. 50.9 nmol/L, P < .001)
from Oakland, CA African American mothers have lower 25(OH)D
Dror et al, 2011 [42] 210 mother-newborn pairs Maternal venous blood collected at birth Maternal serum 25(OH)D was positively correlated
from Oakland, CA admission and cord blood collected immediately with cord blood 25(OH)D (r = 0.79, P < .0001).
postdelivery for serum 25(OH)D assays.
Nicolaidou 123 Healthy mother-newborn Mothers and cord blood measurement at Maternal 25(OH)D levels of 41.0 nmol/L vs umbilical
et al, 2006 [90] pairs from Greece delivery: serum 25(OH)D, calcium, phosphorus, blood concentrations of 51.0 nmol/L (P < .001).
alkaline phosphatase (ALP), PTH, Positive correlation was observed between maternal
osteocalcin, and calcitonin and infant 25(OH)D concentrations (r = 0.626, P < .001).
Sachan et al, 2005 [89] 207 urban and rural pregnant Mothers' measurements at delivery: Mean maternal serum 25(OH)D was 35.0 ± 23.3 nmol/L,
women from Lucknow, serum calcium, inorganic phosphorus, 25(OH)D, and cord blood 25(OH)D was 21.0 ± 14.3 nmol/L. PTH
Northern India alkaline phosphatase, and PTH. rose above the normal range when 25(OH)D

N U TR IT ION RE S EA R CH 63 ( 20 1 9 ) 1 – 2 0
Cord blood measurements: was <56.3 nmol/L.
alkaline phosphatase and 25(OH)D. Maternal serum 25(OH)D correlated positively
with cord blood 25(OH)D (r = 0.79, P < .001) and
negatively with PTH (r = 0.35, P < .001).
Bassir et al, 2001 [88] 50 mother-neonate pairs Mothers' measurements at delivery: 80% of the women had 25 (OH)D
from Tehran, Iran serum calcium, 25-OHD, PTH and concentrations <25 nmol/L.
phosphatase alkaline. The mean cord serum 25(OH)D concentration was
Cord blood measurements: serum very low (4.94 ± 9.4 nmol/l) and that of infants of
calcium, 25(OH)D and phosphatase mother with hypovitaminosis D was almost
alkaline levels. undetectable (1.2 ± 1.2 nmol/L).
Lee et al, 2007 [40] 40 healthy, mostly black Venous blood was drawn from the Of the mothers, 50% (n = 20) were vitamin D deficient,
(62.5%), mother-neonate mother and the infant to assess plasma 25(OH)D and most infants were vitamin D deficient (65%; n = 26).
pairs from Boston, MA concentration 24 to 48 h after birth. The mean 25(OH)D for black mothers and
infants = 41.1 ± 34.3 nmol/L and 26.6 ± 25.4 nmol/L,
respectively. The 25(OH)D for white mothers and
infants = 51.3 ± 47.6 nmol/L and 38.5 ± 35.2 nmol/L,
respectively.
Overall correlation r2 = 0.68, P < .001 between 25(OH)D
levels in infants and mothers.
Bodnar et al, 2007 [16] 400 mother-neonate pairs Mothers and cord blood measurements: At delivery, vitamin D deficiency and insufficiency
from Pittsburgh, PA Serum 25(OH)D at 4-21 wk of gestation, occurred in 29.2% and 54.1% of black women and 45.6%
at delivery, and in cord blood (n = 200 and 46.8% black neonates, respectively.
white and 200 black pregnant women) Five percent and 42.1% of white women and
9.7% and 56.4% of white neonates were
vitamin D deficient and insufficient, respectively.
Wang et al, 2016 [152] 102 mother-neonate Mothers and cord blood measurements at Prevalence of vitamin D insufficiency was 38.2%
pairs from China delivery: serum 25(OH)D. and 26.5% for mothers and neonates, respectively,
whereas 24.5% mothers and 64.7% neonates were
vitamin D deficient.
Neonatal serum 25(OH)D level differed
significantly between the groups of mothers
with different serum 25(OH)D levels (P < .001).
Risk factors Authors, year Sample size and study location Intervention or assessment

Overall correlations r = 0.914, P < .001 between


25(OH)D levels in infants and mothers.
Covered Cuhaci-Cakir and 83 Mother-infants Mothers and infants measurements: serum In summer, the rate of vitamin D deficiency was
clothing Demirel, 2014 [99], (3-4 mo old) from Ankara, Turkey 25(OH)D. calcium, inorganic phosphorus, higher in mothers who wore clothing that
and alkaline phosphatase. covered nearly all of the body (55%) than in
All infants were receiving 3 drops (400 IU) mothers whose clothing covered less of the
of vitamin D3 daily. body (13.6%) (P = .016).
In all groups of infants, regardless of seasonal
variation or mothers' clothing style, serum
25-OHD levels and serum concentrations of
Ca, P, and ALP were not statistically different.
Dijkstra 87 neonates of healthy Mothers and cord blood measurements: A higher prevalence of vitamin D deficiency
et al, 2007 [100] mothers with either dark skin serum 25(OH)D, alkaline phosphatase, PTH, was found in the newborn infants of mothers
and/or concealing clothing ionized calcium and phosphorus. at risk (63.3%) compared with the control
(risk group) or light skin group (15.8%). Within the risk group, the
(control group) from Rotterdam, neonates of mothers wearing veils were
Netherlands. identified to be at highest risk of vitamin D

N U TR IT ION RE S EA R CH 6 3 ( 20 1 9 ) 1– 2 0
deficiency with a prevalence of 90.9%.
25(OH)D values in pregnant women and
their newborn infants showed a positive
correlation (r = 0.88).
Vitamin D Hollis 350 pregnant women followed RCT, double-blinded, with 3 arms: One month prior delivery, mean 25(OH)D
supplementation/ et al, 2011 [69] from 12-16 wk of gestation (1) daily 400 IU of vitamin D (n = 111), concentration was 118 ± 34.9 nmol/L
RCTs until term from South (2) daily 2000 IU of vitamin D (n = 122), among the 4000 IU group vs 105.4 ± 35.7 nmol/L
Carolina, USA or (3) daily 4000 IU of vitamin D (n = 117). and 79.4 ± 34.3 nmol/L among the 2000 IU
Plasma 25(OH)D was measured at 2 time and the 400 IU groups, respectively (P < .0001).
points: (1) 1 mo prior delivery and (2) at delivery. Similar results were found at delivery: 111.0 ±
40.4 nmol/L for the 4000 IU vs
98.3 ± 34.2 nmol/L in the 2000 IU and
78.9 ± 36.5 nmol/L for the 400 IU dose
group (P < .0001)
82% women achieved 25(OH)D levels
≥80 nmol/L at delivery among the
4000 IU dose group compared to
70.8% in the 2000 IU dose groups
and 50% in the 400 IU (P < .0001).
Cord 25(OH)D was 66.3 ± 25.8 nmol/L
for the 4000 IU group vs 57.0 ± 24.5 nmol/L
in the 2000 IU and 45.5 ± 25.3 nmol/L
in the 400 IU group (P < .0001).
Abrams 49 neonates (27 Hispanic, Serum 25(OH)D and PTH were measured At baseline, mean serum cord 25(OH)D was 55.8 ±
et al, 2012 [73] 22 whites) at birth (cord) and at 3 mo of age. 23.5 nmol/L for white neonates and 41.0 ±
followed up from birth till All infants were on 400 IU/d of 16.3 nmol/L for Hispanic neonates (P = .03).
3 mo of age vitamin D supplements 38 infants returned for 3-mo follow-up (18
from Houston, TX whites and 18 Hispanic). Serum 25(OH)D
increased to 94.5 ± 19.0 nmol/L for white
infants and 78.0 ± 20.3 nmol/L for Hispanic
infants at 3 mo of age (P = .01).

(continued on next page)

7
8
Table 2
(continued)
Table 2 (continued)
Risk factors Authors, year Sample size and study location Intervention or assessment Outcomes

Wagner 257 pregnant women followed RCT, double-blinded with 2000 IU/d (n = 130) Overall maternal 25(OH)D change from
et al, 2013 [74] from 12 to 16 wk of gestation vs 4000 IU/d (n = 127). baseline was +14.1 ng/mL (SD12.7 P < .001).
until term from South Maternal serum total calcium, creatinine, and No statistically significant difference
Carolina, USA inorganic phosphorus were measured bimonthly between the 2000-IU and the 4000-IU
also from cord blood at delivery. Intact groups at last visit prior to delivery.
PTH was also measured. Mean cord blood 25(OH)D was 27.0 ±
13.3 ng/mL among the 4000-IU group
vs 22.1 ± 10.3 ng/mL in the 2000-IU group (P = .024).
Grant 260 pregnant women followed RCT, double-blinded, with 3 mother/infant arms: Significant differences between placebo
et al, 2014 [70], from 27 wk of gestation until (1) daily 1000/400 IU of vitamin D (n = 87), group and the lower-dose or the
term and their infant, from (2) daily 2000/800 IU of vitamin D (n = 86) or higher-dose group during gestation
birth to age 6 mo from (3) placebo/placebo (n = 87). (both P < .001) and in cord blood
Auckland, New Zealand Plasma 25(OH)D was measured at 4 time points: (P = .002 vs P < .001).

N U TR IT ION RE S EA R CH 63 ( 20 1 9 ) 1 – 2 0
(1) at enrolment; (2) at 36 wk of gestation; (3) in No significant differences observed
cord plasma at delivery; and (4) in infants between the intervention groups.
at 2, 4, and 6 mo of age. For infants, significant differences
between placebo and the lower-dose
or the higher-dose group were also
reported for infants at 2 mo (both
P < .001) and at 4 mo (P = .02 vs P < .001).
Hossain 200 pregnant women followed Open-label RCT allocated to 2 groups: At birth, maternal 25(OH)D was increased in
et al, 2014 from 20 wk of gestation until (A) routine care of 200 mg ferrous group B (18.3 ± 11 ng/dL vs 8.82 ± 11.84 ng/dL,
child birth rom Karachi, Pakistan sulfate and 600 mg calcium P = 0 .001) compared with group A (6.9 ±
daily (n = 100) and (B) vitD supplementation 7.0 ng/dL vs 6.32 ± 3.97 ng/dL, P = .06).
of 4000 IU/d + routine care of Neonatal 25OHD levels were significantly
200 mg ferrous sulfate higher in group B compared with group A
and 600 mg calcium daily (n = 100). (19.22 ± 12.19 ng/dL vs 6.27 ± 5.2 ng/dL).
Maternal serum 25(OH)D was collected Maternal and neonatal 25OHD concentrations
at baseline and delivery. Neonatal vitD were positively correlated (r = 0.83, P = .001).
status was assessed
from cord blood at birth or from
neonatal serum samples ≤48 h of birth.
Perumal 160 pregnant Bangladeshi RCT, double-blinded, with 2 arms: Mean 25(OH)D at delivery was 134 ± 31 nmol/L for
et al, 2015 [71], women followed from 26 to (1) weekly dose of 35 000 IU of the supplemented group vs 37 ± 18 nmol/L for the
29 wk of gestation until term vitamin D (n = 60) or (2) placebo (n = 55). placebo group (P < .001).
from Dhaka, Bangladesh Plasma 25(OH)D was measured at Mean 25(OH)D at 8 wk postpartum for the
3 time points: (1) at delivery including supplemented group was 62 ± 15 vs 38 ±
cord blood, (2) 8 wk 16 nmol/L for the placebo group (P < .001).
postpartum and, (3) 16 wk postpartum. The effect was attenuated at 16 wk after delivery
(55 ± 16 vs 47 ± 17 nmol/L; P = .06), respectively.
Sablok 180 pregnant women followed RCT, double-blinded, with 4 arms: All treated mothers showed an increase in 25(OH)D
et al, 2015 [72] from 14 to 20 wk of gestation (1) a total of 60 000 IU of vitamin D levels after supplementation, with median levels of
until delivery from Delhi, India (n = 28), (2) 240 000 IU 65 nmol/L postsupplementation compared to
of vitamin D (n = 27), (3) 480000 IU 35 nmol/L presupplementation and 24 nmol/L
of vitamin D (n = 53), and in the nonintervention group. Significance not reported.
Risk factors Authors, year Sample size and study location Intervention or assessment

(4) nonintervention (n = 60). Fewer infants from nonintervention group


Plasma 25(OH)D was measured at had serum 25(OH)D greater than 50 nmol/L
2 time points: (1) at enrolment and (2) compared to infants in the intervention
at delivery including cord blood. groups (14% vs 46.2%; P < .001).
Prepregnancy Bodnar 400 mother-neonate pairs Mothers and cord blood measurements: Compared with women with BMI <25, pregravid
and pregnancy et al, 2007 [41] from Pittsburgh, PA serum 25(OH)D at 4-21 wk of obese women with BMI ≥ 30 had lower adjusted
BMI gestation, predelivery, and at delivery. mean serum 25(OH)D concentrations at 4-22 wk
(56.5 vs. 62.7 nmol/L; P < .05) and a higher
prevalence of vitamin D deficiency (61 vs. 36%; P < .01).
Vitamin D concentration of neonates born to
obese mothers was lower than neonates of
lean mothers (adjusted mean, 50.1 vs.
56.3 nmol/L; P < .05).
An increase in BMI from 22 to 34 was associated
with 2-fold (95% CI, 1.2-3.6) and 2.1-fold (1.2-3.8)
increases in the odds of midpregnancy and
neonatal vitamin D deficiency, respectively.
Women with prepregnancy BMI ≥35 kg/m2 had

N U TR IT ION RE S EA R CH 6 3 ( 20 1 9 ) 1– 2 0
Woolcott 1635 pregnant women from Serum maternal blood assessed for
et al, 2016 [14] Quebec City and Halifax, Canada 25(OH)D at 15 wk of gestation and 8.2 nmol/L (4.0-12.3) serum 25(OH)D less than
neonatal cord blood at delivery women with prepregnancy BMI <25 kg/m2
Cord blood 25(OH)D was lower by 6.1 nmol/L
(0.5-12.8) with maternal BMI 30-<35 compared
to BMI <25 kg/m2
Josefson 360 white mother-neonate pairs Serum maternal and cord blood Maternal serum 25(OH)D was lower by
et al, 2016 [112] from Chicago, IL; Cleveland, OH; were assessed for 25(OH)D 0.40 ng/ml for BMI higher by 1 kg/m2
and Toronto, Ontario, Canada (P < .001) after adjusting for other covariates.
Cord blood 25-OHD was lower by 0.26 ng/ml for
maternal BMI higher by 1 kg/m2 (P < .004).
Smoking/ Diaz-Gomez 61 mother-neonate pairs 25(OH)D, 1,25 (OH)2D, calcium, Newborns of mothers who smoked had significantly l
cigarette smoke et al, 2007 [139], (n = 32 smoking women vs phosphorus, and alkaline ower serum 25(OH)D (14.2 ± 6.2 ng/mL vs 22.3 ±
exposure n = 29 nonsmoking women) phosphatase were assessed 11.3 ng/mL, P = .009).
from Canary Island, Spain from blood samples of mothers
during the last trimester of
pregnancy. Infants were
assessed at 2 to 3 d old.
Banihosseini 108 mother-neonate pair Mothers and cord blood No significant difference in the mean 25(OH)D
et al, 2013 [153], (n = 54 exposed to cigarette measurement at delivery: 25(OH)D, PTH, in maternal serum in exposed and nonexposed group.
smoke vs n = 54 nonexposed calcium, and alkaline phosphatase. No significant difference in the mean
women) from Tehran, Iran. concentration of 25(OH)D in cord serum of
exposed and nonexposed group.
Significant correlation between maternal
and umbilical cord serum 25(OH)D in both
exposed and nonexposed subjects
(r = 0.824, P < .001).
Gangat 68 newborns from 25(OH)D levels were measured from African American infants had lower 25(OH)D
et al, 2012 [96], New Orleans, LA blood samples collected levels compared with white infants (43 ±
from cord blood, at 2 and 4 mo of age 2.8 nmol/L vs 69.2 ± 4.2 nmol/L, P < .001)
respectively, at birth.
No significant correlation was seen between

9
(continued on next page)
10
Table 2
(continued)
Table 2 (continued)
Risk factors Authors, year Sample size and study location Intervention or assessment Outcomes

25(OH)D cord blood values and levels at


2 mo and 4 mo.
Skin Dror 80 mother-newborn pair Maternal venous blood collected at labor African American mothers have lower
pigmentation/ et al, 2012 [151] (40 African Americans admission and cord blood collected 25(OH)D compared to non–African American
ethnicity and 40 non–African immediately postdelivery for mothers (69.1 ± 26.1 vs 82.3 ± 30.3 nmol/L, P = .04).
Americans) from Oakland, CA serum 25(OH)D assays. Also, African American neonates have lower
cord 25(OH)D compared to non–African
American neonates (36.0 vs 48.2 nmol/L P = .004)
Dror et al, 2011 [42] 210 mother-newborn pair Maternal venous blood collected at African American mothers have lower
(107 African Americans and labor admission and cord 25(OH)D compared to non–African American
93 non–African Americans) blood collected immediately mothers (69.2 ± 32.4 vs 78.7 ± 34.5 nmol/L, P = .046).
from Oakland, CA postdelivery for serum 25(OH)D assays. Also, African American neonates have
lower cord 25(OH)D compared to non–African

N U TR IT ION RE S EA R CH 63 ( 20 1 9 ) 1 – 2 0
American neonates (39.1 ± 20.5 vs 48.7 ±
24.7 nmol/L P = .001)
Abrams 49 neonates (27 Hispanic, Serum 25(OH)D and PTH were At baseline, mean serum cord 25(OH)D was
et al, 2012 [73], 22 whites) followed up from measured at birth (cord) lower for Hispanic neonates compared to white
birth till 3 mo of age from and at 3 mo of age. neonates (41.0 ± 16.3 vs 55.8 ± 23.5 nmol/L, P = .03).
Houston, TX All infants were on 400 IU/d Also at 3-mo follow-up (38 infants returned
of vitamin D supplements for 3-mo follow-up; 18 whites and 18 Hispanic),
serum 25(OH)D remained lower for Hispanic
infants compared to white infants (78.0 ±
20.3 nmol/L vs 94.5 ± 19.0 nmol/L; P = .01).
Seasonal Gangat 68 newborns from 25(OH)D levels were measured Mean 25(OH)D levels were lower in the winter
variation et al, 2012 [96] New Orleans, LA from blood samples collected from (44.1 ± 4.9 nmol/L, n = 12) and summer
cord blood, at 2 and 4 mo of age (52.2 ± 3.6 nmol/L, n = 24) and higher in the
spring (61.0 ± 9.7 nmol/L, n = 12) and fall
(72.0 ± 6.1 nmol/L, n = 20), P < .05).
Dror et al, 2011 [42] 210 mother-newborn pair Maternal venous blood collected Prevalence of serum 25(OH)D < 75 nmol/L was
(107 African Americans at labor admission and cord blood highest in African American mothers who
and 93 non–African collected immediately postdelivery delivered in late winter/early spring (78.6%)
Americans) from Oakland, CA for serum 25(OH)D assays. and lowest in non–African American
mothers who delivered in late
summer/early fall (36.4%).
Dijkstra 87 neonates of healthy Mothers and cord blood measurements The mean serum 25(OH)D values did not
et al, 2007 [100] mothers with either dark at delivery: serum 25(OH)D, alkaline differ significantly between the seasons
skin and/or concealing phosphatase, PTH, ionized calcium, of birth in both groups.
clothing (risk group) or and phosphorus.
light skin (control group)
from Rotterdam, Netherlands.
Cuhaci-Cakir and F 83 Mother-infants (3-4 mo old) Serum 25(OH)D. calcium, inorganic In summer, the rate of vitamin D deficiency was
Demirel, 2014 [99], from Ankara, Turkey phosphorus, and alkaline phosphatase higher in mothers who wore clothing that
were measured from both mothers covered nearly all of the body (55%) than in
and infants blood samples. mothers whose clothing covered less of the
body (13.6%) (P = .016).
N U TR IT ION RE S EA R CH 6 3 ( 20 1 9 ) 1– 2 0 11

released a proposition about the composition of a multinutrient


supplement providing the daily recommended allowance of
vitamins, including 200 IU of VitD for pregnant women [58].
differences based on their clothing style (P > .05).

of Ca, P, and ALP were not statistically different.


serum 25-OHD levels and serum concentrations
seasonal variation or mothers' clothing style, Today, several organizations, including Health Canada and the
insufficiency, and there were no significant

American Dietetic Association, encourage women to take a


In the winter, nearly half of both groups of

prenatal multivitamin daily during their pregnancy [18,55,59-


mothers had vitamin D deficiency or

In all groups of infants, regardless of

61]. Most prenatal multivitamins contain 400 IU of VitD, although


a panel of experts specifically suggests a daily dose of 600 IU
Intervention or assessment

supplemental VitD for women during pregnancy to prevent


nutritional rickets in infants [62]. To maintain maternal 25(OH)D
concentrations in the targeted range, intake exceeding 600 IU/d
may be needed [1,18,63].
D: day; CA: California; IL: Illinois; IU: international units; LA: Louisiana; MA: Massachusetts; Mo: months; OH: Ohio; PA: Pennsylvania; TX: Texas; Wk: weeks.

4.2. Dietary intake and use of supplements

4.2.1. Dietary intake


In North America, fortified dairy and dietary supplements are
the main dietary source of VitD [3,64]. The Maternal-Infant
Research on Environmental Chemicals (MIREC) study across
Canada identified milk (52.3%), fish (all species) (9.1%), and
yogurt (8.6%) as the 3 major food components that contrib-
uted to dietary VitD intakes [65]. Aghajafari et al [66] observed
a significant increase of 168 IU in VitD intake from diet and
Sample size and study location

All infants were receiving 3 drops

supplements from the first to the third trimester among


pregnant women in Alberta. However, 44% of the pregnant
(400 IU) of vitamin D3 daily.

women did not meet the VitD DRI of 600 IU/d because of
limited VitD from food and beverages [66]. Furthermore, the
US National Health and Nutrition Examination Survey
(NHANES) data showed that 66% of the pregnant women
studied did not take VitD supplements, whereas 19% took
supplements containing <400 IU/d [67]. The authors con-
cluded that current US VitD supplementation recommenda-
tions of 400 IU/d might not be adequate for many pregnant
women and their infants [67].

4.2.2. Use of supplements

4.2.2.1. Vitamin D supplementation. Routine supplementa-


tion of pregnant women has been considered as a strategy to
increase newborn stores of VitD. Several questions remain
Authors, year

about the optimal period to take supplemental VitD and how


much maternal supplementation is enough to support
maternal and fetal stores. Few studies have focused specifi-
cally on VitD during pregnancy. A subsample of pregnant
women (n = 1186) from the MIREC study reported that VitD
supplemental intake accounted for a median of 60% of total
VitD intake [65]. A study carried out in Oakland, CA, showed
significant correlations between average maternal VitD intake
(food and supplement) with maternal and cord serum 25(OH)
Risk factors

D (r = 0.26 and r = 0.33, P < .001, respectively) [42]. Overall,


there is good support for maternal supplementation as a
strategy to support neonatal 25(OH)D concentrations [68].
Table 2 summarizes the main outcomes of VitD intervention
studies on pregnant women and neonates [69-75]. A meta-
analysis [68] on supplemental VitD during pregnancy showed a
mean difference of 54.7 nmol/L of serum 25(OH)D concentration
among participants who received a supplement compared to
placebo/control groups. Daily supplementation allowed women
to reach higher 25(OH)D concentrations at term compared to a
single oral dose of VitD. On the other hand, the main responses
12 N U TR IT ION RE S EA R CH 63 ( 20 1 9 ) 1 – 2 0

were highly heterogeneous, which could be explained by and postpartum women [84]. Conversely, perceived health
different doses, inception points, and methods to assess serum benefits [85], a recommendation by a health care professional,
25(OH)D used in the analyzed trials [68]. The effect of maternal and blood test results [85] were reported to enhance adher-
VitD supplementation on neonates has also been shown through ence to supplement use. The brand, dosage, cost, and safety
randomized controlled trials [69,70,72]. Gangat et al reported that of supplements were also factors considered when buying a
neonates whose mothers received placebo during pregnancy had supplement [85]. Based on the recommendation for taking
significantly lower cord blood 25(OH)D compared to those whose VitD by a health professional being a facilitator, Awker et al
mothers received 400 and 800 IU/d (32.5 vs 60.0 and 65.0 nmol/L; [86] thus looked at the impact of a clinic-wide VitD protocol on
P < .001, respectively) [70]. Similarly, Hollis et al reported that pregnant women and reported that women were more likely
cord 25(OH)D was 45.5 ± 25.3 nmol/L for neonates born to to take VitD supplements when it was prescribed.
mothers who received 400 IU/d vs 57.0 ± 24.5 nmol/L in the
2000-IU/d and 66.3 ± 25.8 nmol/L in the 4000-IU/d groups 4.2.3. Population statistics on vitamin D supplementation of
(P < .0001) [69]. Overall, in a recent meta-analysis, maternal VitD infants
supplement use was associated with a mean difference of Despite the international recommendation to provide a daily
34 nmol/L (95% CI, 25-42) in cord blood [76]. VitD supplement to breast-fed infants, putting it into practice
There is a general consensus for breast-fed infants to be seems to be a challenge. A survey of 16 389 US infants aged 1-
supplemented with 400 IU VitD/d because of low VitD content 10.5 months showed that <8% of the infants received oral
of human milk [1,17,46]. Despite the recommendation, the VitD supplementation [80]. One-month-old infants had the
NHANES study reported an 8.7% (SE 1.3%) prevalence of VitD lowest VitD supplementation prevalence (3.9%), with the
supplement use in US infants aged 0-11 months [77], whereas highest prevalence among the 5-month-old infants (7.3%)
a review on hypovitaminosis D in the United States reported [80]. Furthermore, the 2009-2012 NHANES study suggested
that the most (78%) severely VitD-deficient infants were those that less than 1 in 5 breast-fed infants met the VitD
breast-fed in winter who did not receive VitD supplementa- recommendation of 400 IU/d compared to nearly 1 in 3 non–
tion [47]. Formula-fed infants were not included in the breast-fed infants [78].
recommended daily supplementation of 400 IU because According to the results of the 2009-2010 Canadian
infant formulas provide 400 IU of VitD/L [46,78]. However, to Community Health Survey, 74.1% of breast-fed infants re-
reach the requirements, formula-fed infants need to consume ceived the recommended intake of VitD on a given day [83].
1 L/d [79], which is not typical [80]. The percentages of individuals who gave VitD containing
supplements were lower among mothers with lower income
4.2.2.2. Adherence to supplementation. Adherence to sup- (67.3% from the lowest quintile vs 83.0% from the highest
plementation is another factor that can impact both maternal quintile) and education (less than secondary graduation,
and infant VitD stores. The 2015 Canadian Community Health 63.7% vs postsecondary graduation, 77.5%). As previously
Survey reported that only 42.7% (Coefficient of variation, 2.7%) of discussed, ethnic background was also highlighted in this
women ≥19 years used VitD supplements [81]. Sociodemographic survey [83]. Years since immigration was also important,
factors including racial disparities contribute to differences in where 66.4% of recent immigrants of <5 years gave their
adherence to supplementation. For instance, black women infant a VitD supplement compared to 75.6% of those who
were found to adhere significantly less to prenatal supplemen- immigrated ≥5 years ago [83]. More married or common-law
tation compared to white women (72% vs 79%; P ≤ .01) [82]. mothers gave a VitD supplement to their breast-fed infants
Similarly, a US national cohort study reported that women compared to single, divorced, separated, or widowed mothers
of ethnic minority and lower economic status were more (75.4% vs 65.8%). On a regional level in Canada, the province of
likely not to use a daily VitD supplement (OR [95% CI] of Ontario had a significantly lower percentage of breast-fed
2.7 [2.1-3.6] and 2.8 [2.3-3.5] for non-Hispanic blacks and infants on VitD supplementation (68.9%) than the national
Hispanics, respectively), which could influence the concentra- average (74.1%), whereas the Prairies region had the highest
tions of serum 25(OH)D [67]. level (82.8%). In the Quebec region, the percentage was 74.5%
There were also differences in use of VitD supplements in which is close to the Canadian average where about 3 of 4
infancy based on race/ethnicity in Canada [83]. A lower breast-feeding mothers followed the Health Canada recom-
percentage of black mothers (57.5%) reportedly provided mendation to provide their infants with a supplement
their breast-fed infants with VitD supplements compared to containing VitD [83].
73.7% of Asian and 75.9% of white mothers [83]. The NHANES
study equally reported significant differences in VitD supple- 4.3. Maternal vitamin D status
ment use among children from age 0 to 18 years based on
race/ethnicity [77], with 15.5% of non-Hispanic blacks using Low VitD status during pregnancy is a major issue identified
VitD a supplement compared to 18.6% and 31.0% of Mexican in a number of studies (Table 2). The NHANES data reported
Americans and non-Hispanic whites, respectively [77]. that 33% (95% CI, 28-40) of the pregnant women studied had
There are other barriers and facilitators of adherence to 25(OH)D < 50 nmol/L and 7% (95% CI, 4-10) had 25(OH)
supplementation. For example, women's beliefs about sever- D < 25 nmol/L [67]. The study also showed that minority
ity of nutrient deficiencies, low perceptions of susceptibility race/ethnicity, lower socioeconomic status (SES), younger age,
to develop deficiencies, and limited knowledge about micro- pregnancy, and later trimester were independently associated
nutrient deficiencies other than iron were reported barriers to with lack of VitD supplementation. There is a lack of national
initiation of supplement use among prepregnant, pregnant, surveys regarding VitD status of pregnant women in Canada.
N U TR IT ION RE S EA R CH 6 3 ( 20 1 9 ) 1– 2 0 13

However, a study on 467 pregnant women reported that 31.5% black, Indian, and Asian people [92]. Different assays for VDBP
had 25(OH)D concentrations <50 nmol/L (mean concentra- often have different affinities for different VDBP epitopes,
tions: 26.3 ± 9.4 nmol/L) [87]. Studies from different countries which might make results inconsistent. This reinforces the
equally observed significant and strong correlations between need for further research on genetic disparities between
infant 25(OH)D concentrations and maternal values during ethnicity for VitD production and concentrations.
gestation [16,88], at delivery (cord blood) [75,88-90], and 24- In a recent meta-analysis, Martin et al [95] reported a high
48 hours after birth [40,75] (Table 2). prevalence of VitD deficiency in dark-skinned individuals,
Maternal VitD deficiency is commonplace in temperate including pregnant women. Correlations between cord blood
latitudes and is a major risk factor for low 25(OH)D concen- 25(OH)D concentrations and skin pigmentation among black
trations in infancy [18]. Adequate VitD status during preg- and white infants showed significantly lower VitD concen-
nancy is known to be important for fetal skeletal trations for infants with darker skin pigmentation compared
development, general growth, and even tooth enamel devel- to the white group (43 ± 2.8 nmol/L vs 69.2 ± 4.2 nmol/L,
opment [91] and is a preventive measure against adverse respectively; P < .001) [96]. The development of skin colora-
outcomes, but it is not a guarantee that neonates will have tion begins at the embryo stage and continues after birth.
sufficient concentrations at birth [16,90]. A target of 75 nmol/L Prior to 32 weeks of gestation, black and white infants exhibit
serum 25(OH)D for pregnant women as discussed earlier a similar pattern of skin pigmentation, but skin color of black
[49,50] might be more beneficial, as it is well established that infants becomes darker, whereas white ones retain their light
neonatal 25(OH)D is usually lower than maternal concentra- skin color [97]. The skin of newborns is not fully pigmented
tions (Table 2). Therefore, the importance of achieving because of the relatively low production of melanin during
sufficient VitD status in mothers as a result of intake from the fetal and neonatal periods but will mature during the 6
all sources needs consideration. months following birth [98]. Considering these physiological
changes, assessing skin pigmentation of newborns is likely
4.4. Sunlight exposure, ethnicity, and skin pigmentation not the best practice to determine their risk of low VitD status,
and maternal natural skin coloration should be considered
Several factors influence the amount of ultraviolet exposure instead.
available for the photosynthesis of VitD, including skin
pigmentation, sunshine exposure, seasonal variation, geo- 4.5. Seasonal variations and clothing
graphical location, and ultraviolet protection (clothing, sun-
screen). It was reported in adults (30-39 years) that those with The effect of seasonal variations on maternal VitD status
lighter skin pigmentation who expose their body to sunlight results in 25(OH)D concentrations being lowest in winter in
for 10-15 minutes during summertime will generate between North America [87,96]. In a cohort of pregnant women living
10 000 and 20 000 IU of VitD, whereas dark-skinned individ- in Toronto, Canada, prevalence rates of VitD deficiency were
uals need about 5-10 times more exposure to synthesize the 31.3%, 29.9%, 17.0%, and 21.8% in winter, spring, summer, and
same amount of VitD [1,92]. The degree of melanin content fall seasons, respectively (P = .006) [87]. Gangat et al [96]
affects VitD production by absorbing ultraviolet rays. The equally reported lowest cord blood 25(OH)D concentrations
more melanin the skin contains, the less ultraviolet beta rays (nmol/L) in winter (44.1 ± 4.9) compared to summer (52.2 ±
are available to activate endogenous VitD production [93]. 3.6), spring (61.0 ± 9.7), and fall (72.0 ± 6.1) (P < .05) in South-
Thus, individuals with darker skin pigmentation ranging from ern Louisiana, USA.
dark (skin type IV) to black (skin type VI), such as black or Clothing is also a factor limiting the exposure of skin to
West Indian individuals, require substantially more sunlight solar radiation and varies according to the season. Although
exposure for the synthesis of VitD. there seems to be scarcity of data on effects of clothing style
In the literature, ethnic background seems to be another risk on maternal VitD status in North America, such studies have
factor leading to low neonatal VitD status, mostly related to low been carried out outside North America [72,99,100]. Cuhaci-
maternal serum 25(OH)D concentrations. A study reported lower Cakir and Demirel investigated the VitD status of Turkish
VitD values among black women (46.3 ± 21.0 nmol/L) compared mothers and their 3- to 4-month-old infants according to
to Hispanics (65.3 ± 18.8 nmol/L) and whites (73.8 ± 36.0 nmol/L; season and mother's clothing style [99]. In summer, signifi-
P < .001) at first trimester [74]. Dror et al also reported signifi- cantly lower serum 25(OH)D concentrations were found in
cantly lower mean serum 25(OH)D in African American women covered mothers (hair, arms, and legs completely covered)
(69.2 ± 32.4 nmol/L) and infants (39.1 ± 20.5 nmol/L) compared to compared to mothers not wearing covered clothing
non–African American women (78.7 ± 34.5 nmol/L; P = .046) and (34.5 nmol/L vs 58.5 nmol/L, respectively; P < .001). During
infants (48.7 ± 24.7 nmol/L); P < .001); the study however found the winter season, 25% of mothers in both groups were VitD
skin pigmentation to be a more sensitive index of VitD status deficient [<37.5 nmol/L 25(OH)D], and another 25% were VitD
than racial origin [42]. insufficient (<50 nmol/L). Very low rates of VitD deficiency
One study tried to genetically explain these VitD race- were found in all infant groups as assessed at 3-4 months of
related disparities between black and white individuals and age as a result of daily VitD supplementation since birth [99].
found that levels of VDBP were lower in black people than in However, an earlier study in Rotterdam, Amsterdam, reported
whites (168 ± 3 μg/mL vs 337 ± 5 μg/mL, respectively; P < .001) a higher prevalence of VitD deficiency among neonates of
[94]. Variation in VDBP levels may in part be responsible for veiled mothers or mothers with dark skin in association with
racial differences in 25(OH)D concentrations [94]. However, low (12.5%) use of VitD supplements [100]. The practice of
previous findings reported similar VDBP levels among white, purdah—the custom of fully covering women with clothing—
14 N U TR IT ION RE S EA R CH 63 ( 20 1 9 ) 1 – 2 0

might be a major contributing factor to VitD deficiency observed than 3000 adults reported an inverse association between
in Iranian pregnant mothers and their neonates [88]. Overall, VitD and waist circumference, with the prevalence of VitD
wintertime is the major limiting factor in the production of VitD; deficiency 3-fold higher in adults with high subcutaneous
however, even during summer months, clothing and avoidance adipose tissue and visceral adipose tissue compared to those
of the sun can also reduce its production. with low subcutaneous adipose tissue and visceral adipose
tissue (15.8 vs 5.1%, P < .001) [111]. Because VitD deficiency
4.6. Professional opinion might mediate adverse pregnancy outcomes related to
prepregnancy BMI, Bodnar et al [41] investigated the relation-
As reported previously, there are important divergent opinions of ship between serum 25(OH)D and prepregnancy BMI in a
health professionals about VitD supplementation. Previous pregnancy cohort. A negative correlation was found between
studies reported that few pediatric health care providers prepregnancy BMI and 25(OH)D concentrations at term
recommend VitD for breast-fed infants [101-103] despite the (r = −0.19; P < .001) and in cord blood 25(OH)D concentrations
World Health Organization recommendation to provide a VitD (r = −0.18; P < .0001). An increase in prepregnancy BMI from 22
supplement to children if exposure to sunlight is not possible to 34 kg/m2 was associated with 2.1-fold (95% CI, 1.2-3.8)
[104]. Reasons for these include belief that a recommendation of increase in the odds of neonatal VitD deficiency [41]. Another
supplementation might discourage mothers from breast-feeding pregnancy cohort reported that women with prepregnancy BMI
[103], assumption that human milk contains adequate VitD ≥35 kg/m2 had 8.2 nmol/L (4.0-12.3) serum 25(OH)D less than
[101,102,105] and belief that infants were sufficiently exposed to women with prepregnancy BMI <25 kg/m2 [14]. Similarly, in a
sunlight [102,105]. Importantly, health care professional opinions multicenter study with mother-neonate pairs at 28 weeks’
can affect maternal behaviors, possibly leading to low neonatal gestation, higher BMI in pregnancy was associated with
25(OH)D concentrations. lower maternal 25(OH)D concentrations. However, results
did not show a significant relationship between neonatal
4.7. SES and education serum 25(OH)D concentrations and maternal BMI [112].
Nevertheless, BMI cutoff should not be used for pregnant
SES was another factor reported in the literature that could cohorts [113], and this could be an important limitation in
influence 25(OH)D concentrations of pregnant women. In studies examining maternal obesity in pregnancy and serum
their secondary analysis of the US NHANES 2001-2006, Ginde 25(OH)D concentrations.
et al [67] pointed out different characteristics of nonpregnant Despite the dearth of data about direct relationships
and pregnant women in relation to their VitD status. Higher between maternal VitD status and weight gain during
serum 25(OH)D concentrations were associated with higher pregnancy, 25(OH)D is inversely associated with adiposity
SES, the use of a VitD supplement, longer duration of the and VitD deficiency strongly linked with variation in subcu-
supplementation, and greater outdoor physical activity. In a taneous and visceral adiposity [111]. However, the possibility
multiethnic Canadian population, despite the high levels of that behaviors related to maternal body weight and maternal
education and SES of the pregnant women studied, VitD serum 25(OH)D concentrations interact to regulate infant 25
insufficiency remained common, with 24% of women with (OH)D concentrations requires further investigation.
serum 25(OH)D concentrations <50 nmol/L [106]. The MIREC
study reported a higher proportion of women with lower VitD 4.8.2. Hypertensive disease in pregnancy
intake to have less than a university degree with family Pregnancy-induced hypertension (PIH), also known as preeclamp-
income less than CA $60 000, which is below the Canadian sia and its advanced stage eclampsia, is a pregnancy-specific
median family income of $86 000 [65,107]. Thus, SES and syndrome associated with adverse pregnancy outcomes. The
educational levels should be considered when educating biological cause of preeclampsia involves a number of processes
mothers, notably because of the low adherence to maternal that may be affected by VitD directly or indirectly; this includes
and neonatal supplementation reported among populations placental implantation, abnormal angiogenesis, immune dys-
with lower SES and education [67,83]. function, excessive inflammation, renin-angiotensin-aldosterone
system, and hypertension [114-117].
4.8. Maternal morbidities/pregnancy complications Bodnar et al [118] demonstrated that maternal VitD
deficiency in early pregnancy was a strong, independent risk
It is estimated that low VitD status accounts for about 10% of factor for preeclampsia. The study observed a 5-fold increase
pregnancy complications in Canada [108]. A combined dataset of in the odds of preeclampsia using the 1997 IOM definition of
2 US randomized clinical trials of VitD supplementation during 25(OH)D < 37.5 nmol/L [118]. Also, 2 more recent case-
pregnancy concluded that maternal delivery 25(OH)D was controlled studies demonstrated that maternal VitD defi-
inversely associated with pregnancy comorbidities, with fewer ciency in early pregnancy [119] and midpregnancy [120] is an
events as 25(OH)D increased [109]. As discussed earlier, independent risk factor for preeclampsia even after adjusting
hypovitaminosis D has been associated with a number of health for multiple covariates [119]. Rostami et al showed a 9%
problems in offspring and potentially with maternal morbidities. decrease in preeclampsia risk among pregnant women
supplemented with VitD compared to those who did not
4.8.1. Body mass index receive supplements [121]. Also, the Vitamin D Antenatal
Obesity has been reported to be another risk factor for Asthma Reduction Trial reported a significantly lower risk of
deficient levels of VitD because of the sequestration of VitD preeclampsia among women with 25(OH)D ≥ 75 nmol/L com-
in adipose tissue [110]. The Framingham Heart Study on more pared to women with 25(OH)D < 75 nmol/L in both early and
N U TR IT ION RE S EA R CH 6 3 ( 20 1 9 ) 1– 2 0 15

late pregnancy (adjusted OR, 0.28; CI, 0.08-0.96; P = .04) [122]. A cohort study on 184 pregnant women with GDM reported
Furthermore, another recent VitD supplementation trial that neonates born to mothers with 25(OH)D < 50 nmol/L had
reported fewer preeclampsia events (1.2% vs 7.4%, P < .05) more adverse outcomes compared to neonates born to
and fewer cases of intrauterine growth restriction (9.6% vs mothers with 25(OH)D > 50 nmol/L [137]. Specifically, neo-
22.2, P = .03) among pregnant women supplemented with nates born to mothers with 25(OH)D < 50 nmol/L were more
4000 IU/d VitD compared to those supplemented with 400 IU/d frequently small-for-gestational-age (17.3% vs 5.9%, P = .017),
[123]. In another Iranian study, pregnant women with previous had higher incidences of hospitalization in intensive care
history of preeclampsia and baseline 25(OH)D ≥ 25 ng/ml units (32% vs 19%, P = .048), and had higher incidence of
(n = 142) were randomized into intervention (50 000 IU cho- hypoglycemia (17.3% vs 7.1%, P = .039) compared to their
lecalciferol once every 2 weeks; n = 70) and control (placebo; counterparts [137]. Also Asemi et al reported a lower
n = 72) groups [124]. The study reported a 1.94 (95% CI, 1.02- incidence of hyperbilirubinemia among neonates born to
3.71) higher risk of preeclampsia for control group compared mothers supplemented with VitD compared to the placebo
to the intervention group (P = .036) [124]. Wei et al reported group (27.3% vs 60.9% P = .02) [138]. To date, there is not
that pregnant women with 25(OH)D < 50 nmol/L have an in- enough conclusive evidence to incorporate into specific
creased risk of developing preeclampsia (OR, 2.09 [1.50-2.90]) recommendations of VitD supplementation for pregnant
[125]. Higher odds (OR, 1.79 [1.25-2.58]) of developing pre- women with GDM to support optimal maternal and neonatal
eclampsia when 25(OH)D concentration is <75 nmol/L were outcomes. However, it is important to ensure that maternal
also reported in another meta-analysis [31]. To the best of our 25(OH)D concentrations are at least sufficient, especially in
knowledge, no study has reported on the associations the presence of pregnancy complications with potential to
between PIH and neonatal 25(OH)D concentrations at birth. negatively impact maternal-fetal transfer.
Furthermore, recommendations for the prevention of pre-
eclampsia do not specifically include a targeted intake of 4.9. Lifestyle factors
VitD to threshold of VitD status at this time [126].
4.9.1. Smoking status
4.8.3. Gestational diabetes mellitus The mechanism by which smoking could be associated with
Gestational diabetes mellitus (GDM) is a temporary condition of decreased circulating concentrations of VitD, especially dur-
pregnancy which has been associated with a high risk of future ing pregnancy, remains unclear [139]. Diaz-Gomez et al [139]
type 2 diabetes mellitus [127]. A potential mechanism to explain observed that even if no significant differences were deter-
the relationship between VitD insufficiency and the risk of mined between maternal groups, neonates of smoking
GDM includes the direct effect of VitD on β-cell function, as low mothers had significantly lower serum 25(OH)D concentra-
serum 25(OH)D decreases β-cell function and insulin sensitivity tions than neonates of nonsmokers (35.5 ± 15.5 nmol/L vs
[128]. Studies have shown inconsistent associations between 55.8 ± 28.3 nmol/L, respectively; P = .009). Neonates of
maternal VitD status and glycemia [129-131] with a shortage of smoking mothers were also significantly smaller than neo-
evidence from high-latitude regions like Canada or the northern nates of nonsmoking mothers (2938 ± 465 g vs 3358 ± 456 g,
states of the USA [87]. However, 2 studies reported associations P = .001) [139]. Dodds and colleagues showed a relationship
between VitD insufficiency in early pregnancy and increased risk between smoking in pregnancy and VitD deficiency (OR [95%
of developing GDM over the course of pregnancy [132,133]. CI]) of 3.73 (1.95-7.14) [134]. It is unclear if lower maternal and/
Furthermore, another study reported inverse associations be- or neonatal 25(OH)D concentrations are caused by smoke or
tween VitD status and GDM among women who smoke during other co-associating factors such as supplement use, sun
pregnancy [134]. exposure, and others. However, chemicals from second-hand
A trial in 120 pregnant women from Yazd, Iran, reported a smoke can be passed to the fetus through the placenta [140],
decrease in insulin resistance among women supplemented and nicotine has the effect of decreasing blood flow to the
with 50 000 IU VitD every 2 weeks compared to pregnant women fetus [141]. This could potentially decrease the transfer of 25
on 200 IU/d (Homeostatic model assessment of insulin resistance (OH)D from mother to child.
(HOMA-IR); 0.7 ± 1.04 vs 1.46 ± 1.69, P = .02) [135]. Another trial in
60 women with GDM in Iran reported better maternal and 4.9.2. Use of medications
neonatal outcomes in women supplemented with 1000 mg Several drugs can interfere with VitD metabolism, including
calcium per day plus 2 doses of 50 000 IU cholecalciferol (1 at antiepileptic drugs, glucocorticoids, bisphosphonates, antiretro-
baseline and another at day 21 of the trial) compared to women in viral drugs, antiestrogen drugs, and antihypertensive drugs [142].
the placebo group [136]. Specifically, there were lower cesarean As adequate 25(OH)D concentrations are important for maternal
sections (23·3% vs 63·3%, P = 0·002), maternal hospitalizations (0 and neonatal stores and health, the use of these drugs during
vs 13·3%, P = 0·03), macrosomia (0 vs 13·3%, P = 0·03), pregnancy can be critical. Unfortunately, there is scarcity of data
hyperbilirubinemia (20·0% vs 56·7%, P = 0·03), and neonatal on this topic, and further investigation on the subject is required.
hospitalizations (20·0% vs 56·7%, P = 0·03) in the supplemented
compared to placebo group [136].
A meta-analysis reported that pregnant women with 25 5. Knowledge gaps
(OH)D < 50 nmol/L have an increased risk of developing GDM
(OR 1.38 [1.12-1.70]) [125], whereas another reported slightly Several knowledge gaps are highlighted in this review; for
higher odds of GDM when 25(OH)D concentration is <75 nmol/L instance, data are available on the relationship between
(OR 1.49 [1.18-1.89]) [31]. maternal risk factors and their relative importance. However,
16 N U TR IT ION RE S EA R CH 63 ( 20 1 9 ) 1 – 2 0

knowledge gaps exist between some maternal risk factors and [4] Hazell TJ, Gallo S, Berzina I, Vanstone CA, Rodd C, Weiler
neonatal VitD status. Addressing the following research gaps HA. Plasma 25-hydroxyvitamin D, more so than its epimer,
has a linear relationship to leaner body composition across
will be of value:
infancy in healthy term infants. Appl Physiol Nutr Metab
2014;39:1137–43.
• The impact of pregnancy comorbidities like GDM and PIH [5] Crozier SR, Harvey NC, Inskip HM, Godfrey KM, Cooper C,
on neonatal VitD status. Robinson SM, et al. Maternal vitamin D status in pregnancy
• The use of medications that interfere with VitD metabolism is associated with adiposity in the offspring: findings from
in pregnancy and the effects on maternal-fetal VitD status. the Southampton Women's Survey. Am J Clin Nutr 2012;96:
57–63.
• Explore the genetic disparities among different ethnicities
[6] Harvey NC, Moon RJ, Sayer AA, Ntani G, Davies JH, Javaid
for VitD production and concentrations.
MK, et al. Maternal antenatal vitamin D status and offspring
• Identify the confounding contributors to the high preva- muscle development: findings from the Southampton
lence of VitD deficiency among pregnant women who are Women's survey. J Clin Endocrinol Metab 2014;99:330–7.
newly immigrated to the temperate latitudes. [7] Hollis BW, Wagner CL. Vitamin D and pregnancy: skeletal
• The total maternal VitD intake that is consistent with effects, nonskeletal effects, and birth outcomes. Calcif
healthy targets for neonatal VitD status. Tissue Int 2013;92:128–39.
[8] Kovacs CS, Kronenberg HM. Maternal-fetal calcium and
bone metabolism during pregnancy, puerperium, and lac-
tation. Endocr Rev 1997;18:832–72.
6. Summary and conclusion [9] Mulligan ML, Felton SK, Riek AE, Bernal-Mizrachi C. Impli-
cations of vitamin D deficiency in pregnancy and lactation.
According to the scientific evidence summarized in this Am J Obstet Gynecol 2010;202:429.e1-9.
[10] Wagner CL, Taylor SN, Johnson DD, Hollis BW. The role of
literature review, major maternal risk factors related to low
vitamin D in pregnancy and lactation: emerging concepts.
25(OH)D concentrations in North American newborns are low
Womens Health (Lond) 2012;8:323–40.
maternal 25(OH)D concentrations, inadequate VitD supple- [11] Kovacs CS. Maternal mineral and bone metabolism during
ment use, and intermediate or dark skin pigmentation. pregnancy, lactation, and post-weaning recovery. Physiol
Although black ethnicity came out as a major risk factor for Rev 2016;96:449–547.
low 25(OH)D concentrations, it was difficult to tease out the [12] White P, Cooke N. The multifunctional properties and
extent of the contribution of genetics, clothing style, SES, and characteristics of vitamin D-binding protein. Trends
Endocrinol Metab 2000;11:320–7.
cultural practices. Moderate risk factors include low SES and
[13] Institute of Medicine (US) Committee to Review Dietary
low education levels, GDM, PIH, seasonal variations, high
Reference Intakes for Vitamin D and Calcium. Overview of
maternal BMI, smoking or exposure to smoke, and no vitamin D. In: Ross AC, Taylor CL, Yaktine AL, Del Valle HB,
recommendation of VitD supplementation. Living in the editors. Dietary reference intakes for calcium and vitamin D.
temperate zone limits sun exposure because of winter Washington (DC); 2011.
months and latitude, which can reduce the production of [14] Woolcott CG, Giguère Y, Weiler HA, Spencer A, Forest J-C,
endogenous VitD synthesis. In Canada and the United States, Armson BA, et al. Determinants of vitamin D status in
pregnant women and neonates. Can J Public Health 2016;
VitD testing is not routinely undertaken during pregnancy or
107:410–6.
at birth because of its relatively high cost compared to [15] Aghajafari F, Field CJ, Kaplan BJ, Maggiore JA, O'Beirne M,
supplementation, but it is pertinent that health care providers Hanley DA, et al. The high prevalence of vitamin D
know the risk factors for low VitD status, including the insufficiency in cord blood in Calgary, Alberta (APrON-D
importance of supplementation and adherence. Study). J Obstet Gynaecol Can 2017;39:347-53.e1.
[16] Bodnar LM, Simhan HN, Powers RW, Frank MP, Cooperstein
E, Roberts JM. High prevalence of vitamin D insufficiency in
black and white pregnant women residing in the northern
Acknowledgment United States and their neonates. J Nutr 2007;137:447–52.
[17] Canadian Paediatric Society. Vitamin D: caring for kids.
This research did not receive any specific grant from funding http://www.caringforkids.cps.ca/handouts/vitamin_d; 2012,
Accessed date: 16 May 2016.
agencies in the public, commercial, or not-for-profit sectors.
[18] Godel JC, Irvine J, Onyett H, Saylor K, Schroter H, Young M,
The authors declare no conflict of interest. Wong S. Vitamin D supplementation: recommendations for
Canadian mothers and infants. Paediatr Child Health 2007;
12:583–98.
REFERENCES [19] Dror DK, Allen LH. Vitamin D inadequacy in pregnancy: biology,
outcomes, and interventions. Nutr Rev 2010;68:465–77.
[20] Jacquemyn Y, Ajaji M, Karepouan N. Vitamin D levels in
[1] Wagner CL, Greer FR. Prevention of rickets and vitamin D maternal serum and umbilical cord blood in a multi-ethnic
deficiency in infants, children, and adolescents. Pediatrics population in Antwerp, Belgium. Facts Views Vis Obgyn
2008;122:1142–52. 2013;5:3–5.
[2] Ross AC, Manson JE, Abrams SA, Aloia JF, Brannon PM, [21] Godang K, Froslie KF, Henriksen T, Qvigstad E, Bollerslev J.
Clinton SK, et al. The 2011 dietary reference intakes for Seasonal variation in maternal and umbilical cord 25(OH)
calcium and vitamin D: what dietetics practitioners need to vitamin D and their associations with neonatal adiposity.
know. J Am Diet Assoc 2011;111:524–7. Eur J Endocrinol 2014;170:609–17.
[3] Ross AC, Taylor CL, Yaktine AL, Valle HBD. Dietary reference [22] Mahon P, Harvey N, Crozier S, Inskip H, Robinson S, Arden N,
intakes for calcium and vitamin D. National Academies et al. Low maternal vitamin D status and fetal bone
Press; 2011. development: cohort study. J Bone Miner Res 2010;25:14–9.
N U TR IT ION RE S EA R CH 6 3 ( 20 1 9 ) 1– 2 0 17

[23] Tobias JH, Cooper C. Perspective: PTH/PTHrP activity and the [41] Bodnar LM, Catov JM, Roberts JM, Simhan HN. Prepregnancy
programming of skeletal development in utero. J Bone Miner obesity predicts poor vitamin D status in mothers and their
Res 2004;19:177–82. neonates. J Nutr 2007;137:2437–42.
[24] Balasubramanian S. Vitamin D deficiency in breastfed [42] Dror DK, King JC, Durand DJ, Allen LH. Association of
infants & the need for routine vitamin D supplementation. modifiable and nonmodifiable factors with vitamin D status
Indian J Med Res 2011;133:250–2. in pregnant women and neonates in Oakland, CA. J Am Diet
[25] Weiler H. Vitamin D: the current state in Canada. https:// Assoc 2011;111:111–6.
www.cfdr.ca/Downloads/CCFN-docs/Watching-Brief-on- [43] Martin-Gronert MS, Ozanne SE. Maternal nutrition during
Vitamin-D—final—Aug3-08-revA.aspx; 2008, Accessed date: pregnancy and health of the offspring. Biochem Soc Trans
12 March 2016. 2006;34:779–82.
[26] Ward LM, Gaboury I, Ladhani M, Zlotkin S. Vitamin D- [44] Rush D. Nutrition and maternal mortality in the developing
deficiency rickets among children in Canada. CMAJ 2007; world. Am J Clin Nutr 2000;72:212S-40S.
177:161–6. [45] Bhutta ZA, Das JK, Rizvi A, Gaffey MF, Walker N, Horton S,
[27] CDC. Centers for Disease Control and Prevention identifies et al. Evidence-based interventions for improvement of
nutritional deficiencies among young children. https:// maternal and child nutrition: what can be done and at what
www.cdc.gov/media/pressrel/r010330.htm; 2001, Accessed cost? Lancet 2013;382:452–77.
date: 5 May 2017. [46] Health Canada. Vitamin D and calcium: updated dietary
[28] Weisberg P, Scanlon KS, Li R, Cogswell ME. Nutritional reference intakes. http://www.hc-sc.gc.ca/fn-an/nutrition/
rickets among children in the United States: review of cases vitamin/vita-d-eng.php#a10; 2012, Accessed date: 17 May 2017.
reported between 1986 and 2003. Am J Clin Nutr 2004;80 [47] Rovner AJ, O'Brien KO. Hypovitaminosis D among healthy
[1697S-705S]. children in the United States: a review of the current
[29] Robinson CJ, Wagner CL, Hollis BW, Baatz JE, Johnson DD. evidence. Arch Pediatr Adolesc Med 2008;162:513–9.
Maternal vitamin D and fetal growth in early-onset severe [48] Spiro A, Buttriss JL. Vitamin D: An overview of vitamin D
preeclampsia. Am J Obstet Gynecol 2011;204:556.e1-4. status and intake in Europe. Nutr Bull 2014;39:322–50.
[30] Leffelaar ER, Vrijkotte TG, van Eijsden M. Maternal early [49] Dawson-Hughes B, Heaney RP, Holick MF, Lips P, Meunier PJ,
pregnancy vitamin D status in relation to fetal and neonatal Vieth R. Estimates of optimal vitamin D status. Osteoporos
growth: results of the multi-ethnic Amsterdam born children Int 2005;16:713–6.
and their development cohort. Br J Nutr 2010;104:108–17. [50] Holick MF. Vitamin D status: measurement, interpretation,
[31] Aghajafari F, Nagulesapillai T, Ronksley PE, Tough SC, and clinical application. Ann Epidemiol 2009;19:73–8.
O'Beirne M, Rabi DM. Association between maternal serum [51] Gallo S, Hazell T, Vanstone CA, Agellon S, Jones G, L'Abbe M,
25-hydroxyvitamin D level and pregnancy and neonatal et al. Vitamin D supplementation in breastfed infants from
outcomes: systematic review and meta-analysis of obser- Montreal, Canada: 25-hydroxyvitamin D and bone health
vational studies. BMJ 2013;346:f1169. effects from a follow-up study at 3 years of age. Osteoporos
[32] Wagner C, Baggerly C, McDonnell S, Baggerly K, French C, Int 2016;27:2459–66.
Baggerly L, et al. Post-hoc analysis of vitamin D status and [52] Holick MF, Binkley NC, Bischoff-Ferrari HA, Gordon CM, Hanley
reduced risk of preterm birth in two vitamin D pregnancy DA, Heaney RP, et al. Evaluation, treatment, and prevention of
cohorts compared with South Carolina March of Dimes vitamin D deficiency: an Endocrine Society clinical practice
2009-2011 rates. J Steroid Biochem Mol Biol 2016;155:245–51. guideline. J Clin Endocrinol Metab 2011;96:1911–30.
[33] McDonnell SL, Baggerly KA, Baggerly CA, Aliano JL, French [53] Hayes A, Cashman KD. Food-based solutions for vitamin D
CB, Baggerly LL, et al. Maternal 25 (OH) D concentrations≥ 40 deficiency: putting policy into practice and the key role for
ng/mL associated with 60% lower preterm birth risk among research. Proc Nutr Soc 2017;76:54–63.
general obstetrical patients at an urban medical center. [54] Pilz S, März W, Cashman KD, Kiely ME, Whiting SJ, Holick
PLoS One 2017;12:e0180483. MF, et al. Rationale and plan for vitamin D food fortification:
[34] Bowyer L, Catling-Paull C, Diamond T, Homer C, Davis G, a review and guidance paper. Front Endocrinol 2018;9:373.
Craig ME. Vitamin D, PTH and calcium levels in pregnant [55] Health Canada. Prenatal nutrition guidelines for health
women and their neonates. Clin Endocrinol 2009;70:372–7. professionals—background on Canada's food guide. http://
[35] Eckhardt CL, Gernand AD, Roth DE, Bodnar LM. Maternal www.hc-sc.gc.ca/fn-an/pubs/nutrition/guide-prenatal-eng.
vitamin D status and infant anthropometry in a US multi- php; 2010, Accessed date: 13 May 2016.
centre cohort study. Ann Hum Biol 2015;42:215–22. [56] World Health Organization. Guideline: daily iron and folic
[36] Weiler H, Fitzpatrick-Wong S, Veitch R, Kovacs H, acid supplementation in pregnant women. http://apps.who.
Schellenberg J, McCloy U, et al. Vitamin D deficiency and int/iris/bitstream/10665/77770/1/9789241501996_eng.pdf;
whole-body and femur bone mass relative to weight in 2012, Accessed date: 13 March 2017.
healthy newborns. CMAJ 2005;172:757–61. [57] National Institute of Diabetes and Digestive and Kidney
[37] Camargo Jr CA, Rifas-Shiman SL, Litonjua AA, Rich-Edwards Diseases. Fit for two—tips for pregnancy. https://www.
JW, Weiss ST, Gold DR, et al. Maternal intake of vitamin D niddk.nih.gov/health-information/health-topics/weight-
during pregnancy and risk of recurrent wheeze in children control/tips-for-two-pregnancy/Pages/fit-for-two.aspx;
at 3 y of age. Am J Clin Nutr 2007;85:788–95. 2013, Accessed date: 12 June 2016.
[38] Erkkola M, Nwaru BI, Viljakainen HT. Maternal vitamin D [58] UNICEF, World Health Organization, United Nations
during pregnancy and its relation to immune-mediated University. Composition of a multi-micronutrient supple-
diseases in the offspring. Vitam Horm 2011;86:239–60. ment to be used in pilot programmes among pregnant
[39] Wolsk HM, Harshfield BJ, Laranjo N, Carey VJ, O'Connor G, women in developing countries: report of a United Nations
Sandel M, et al. Vitamin D supplementation in pregnancy, Children's Fund (UNICEF), World Health Organization (WHO),
prenatal 25 (OH) D levels, race, and subsequent asthma or United Nations University workshop; 1999. http://www.who.
recurrent wheeze in offspring: secondary analyses from the int/iris/handle/10665/75358, Accessed date: 13 March 2018.
Vitamin D Antenatal Asthma Reduction Trial. J Allergy Clin [59] Wood CL, Cheetham TD. Vitamin D: increasing supplement
Immunol 2017;140:1423-9.e5. use among at-risk groups (NICE guideline PH56). Arch Dis
[40] Lee JM, Smith JR, Philipp BL, Chen TC, Mathieu J, Holick MF. Child Educ Pract Ed 2016;101:43–5.
Vitamin D deficiency in a healthy group of mothers and [60] Office of Kids and Families. Having a baby. Australia: New
newborn infants. Clin Pediatr (Phila) 2007;46:42–4. South Wales Government; 2012.
18 N U TR IT ION RE S EA R CH 63 ( 20 1 9 ) 1 – 2 0

[61] Kaiser L, Allen LH. Position of the American Dietetic [79] American Academy of Pediatrics. A supplement of 400 IU
Association: nutrition and lifestyle for a healthy pregnancy per day of vitamin D is recommended for all breastfed
outcome. J Am Diet Assoc 2008;108:553–61. infants. https://www.cdc.gov/breastfeeding/
[62] Munns CF, Shaw N, Kiely M, Specker BL, Thacher TD, Ozono recommendations/vitamin_d.htm; 2008, Accessed date: 22
K, et al. Global consensus recommendations on prevention April 2016.
and management of nutritional rickets. J Clin Endocrinol [80] Perrine CG, Sharma AJ, Jefferds ME, Serdula MK, Scanlon KS.
Metab 2016;101:394–415. Adherence to vitamin D recommendations among US
[63] ACOG Committee On obstetric practice. ACOG Committee infants. Pediatrics 2010;125:627–32.
opinion no. 495: vitamin D: screening and supplementation [81] Canada S. Table 13-10-0793-01 types of nutrients from
during pregnancy. Obstet Gynecol 2011;118:197–8. nutritional supplements—consumed in the past month, by
[64] Calvo MS, Whiting SJ, Barton CN. Vitamin D fortification in age group and sex, household population aged 1 and over,
the United States and Canada: current status and data Canadian Community Health Survey (CCHS)–Nutrition,
needs. Am J Clin Nutr 2004;80:1710S-6S. Canada. https://www150.statcan.gc.ca/t1/tbl1/en/cv.action;
[65] Morisset AS, Weiler HA, Dubois L, Ashley-Martin J, Shapiro 2015, Accessed date: 29 June 2018.
GD, Dodds L, et al. Rankings of iron, vitamin D, and calcium [82] Jasti S, Siega-Riz AM, Cogswell ME, Hartzema AG, Bentley
intakes in relation to maternal characteristics of pregnant ME. Pill count adherence to prenatal multivitamin/mineral
Canadian women. Appl Physiol Nutr Metab 2016;41:749–57. supplement use among low-income women. J Nutr 2005;
[66] Aghajafari F, Field CJ, Kaplan BJ, Rabi DM, Maggiore JA, 135:1093–101.
O'Beirne M, et al. The current recommended vitamin D [83] Health Canada. Vitamin D supplementation of breastfed
intake guideline for diet and supplements during pregnancy infants in Canada: key statistics and graphics (2009-2010).
is not adequate to achieve vitamin D sufficiency for most http://www.hc-sc.gc.ca/fn-an/surveill/nutrition/commun/
pregnant women. PLoS One 2016;11:e0157262. prenatal/vit_d_sup-eng.php; 2012, Accessed date: 3 May
[67] Ginde AA, Sullivan AF, Mansbach JM, Camargo Jr CA. 2016.
Vitamin D insufficiency in pregnant and nonpregnant [84] Nechitilo M, Nguyen P, Webb-Girard A, Gonzalez-Casanova
women of childbearing age in the United States. Am J Obstet I, Martorell R, DiGirolamo A, et al. A qualitative study of
Gynecol 2010;202:436.e1-8. factors influencing initiation and adherence to micronutri-
[68] Palacios C, De-Regil LM, Lombardo LK, Pena-Rosas JP. ent supplementation among women of reproductive age in
Vitamin D supplementation during pregnancy: updated Vietnam. Food Nutr Bull 2016;37:461–74.
meta-analysis on maternal outcomes. J Steroid Biochem [85] Toher C, Lindsay K, McKenna M, Kilbane M, Curran S,
Mol Biol 2016;164:148–55. Harrington L, et al. Relationship between vitamin D knowl-
[69] Hollis BW, Johnson D, Hulsey TC, Ebeling M, Wagner CL. edge and 25-hydroxyvitamin D levels amongst pregnant
Vitamin D supplementation during pregnancy: double- women. J Hum Nutr Diet 2014;27:261–9.
blind, randomized clinical trial of safety and effectiveness. J [86] Awker AL, Herbranson AT, Rhee TG, Westberg SM. Impact of
Bone Miner Res 2011;26:2341–57. a vitamin D protocol in pregnancy at an urban women's
[70] Grant CC, Stewart AW, Scragg R, Milne T, Rowden J, Ekeroma health clinic. Ann Pharmacother 2016;50:935–41.
A, et al. Vitamin D during pregnancy and infancy and infant [87] Kramer CK, Ye C, Swaminathan B, Hanley AJ, Connelly PW,
serum 25-hydroxyvitamin D concentration. Pediatrics 2014; Sermer M, et al. The persistence of maternal vitamin D
133:e143–53. deficiency and insufficiency during pregnancy and lactation
[71] Perumal N, Al Mahmud A, Baqui AH, Roth DE. Prenatal irrespective of season and supplementation. Clin
vitamin D supplementation and infant vitamin D status in Endocrinol 2016;84:680–6.
Bangladesh. Public Health Nutr 2017;20:1865–73. [88] Bassir M, Laborie S, Lapillonne A, Claris O, Chappuis MC,
[72] Sablok A, Batra A, Thariani K, Batra A, Bharti R, Aggarwal Salle BL. Vitamin D deficiency in Iranian mothers and their
AR, et al. Supplementation of vitamin D in pregnancy and neonates: a pilot study. Acta Paediatr 2001;90:577–9.
its correlation with feto-maternal outcome. Clin Endocrinol [89] Sachan A, Gupta R, Das V, Agarwal A, Awasthi PK, Bhatia V.
2015;83:536–41. High prevalence of vitamin D deficiency among pregnant
[73] Abrams SA, Hawthorne KM, Rogers SP, Hicks PD, Carpenter women and their newborns in northern India. Am J Clin
TO. Effects of ethnicity and vitamin D supplementation on Nutr 2005;81:1060–4.
vitamin D status and changes in bone mineral content in [90] Nicolaidou P, Hatzistamatiou Z, Papadopoulou A, Kaleyias J,
infants. BMC Pediatr 2012;12:6. Floropoulou E, Lagona E, et al. Low vitamin D status in
[74] Wagner CL, McNeil R, Hamilton SA, Winkler J, Rodriguez mother-newborn pairs in Greece. Calcif Tissue Int 2006;78:
Cook C, Warner G, et al. A randomized trial of vitamin D 337–42.
supplementation in 2 community health center networks in [91] Hollis BW, Wagner CL. Nutritional vitamin D status during
South Carolina. Am J Obstet Gynecol 2013;208:137.e1-13. pregnancy: reasons for concern. CMAJ 2006;174:1287–90.
[75] Hossain N, Kanani FH, Ramzan S, Kausar R, Ayaz S, Khanani R, [92] Matsuoka LY, Wortsman J, Haddad JG, Kolm P, Hollis BW.
et al. Obstetric and neonatal outcomes of maternal vitamin D Racial pigmentation and the cutaneous synthesis of vitamin
supplementation: results of an open-label, randomized D. Arch Dermatol 1991;127:536–8.
controlled trial of antenatal vitamin D supplementation in [93] Marcus R, Feldman D, Nelson D, Rosen CJ. Fundamentals of
Pakistani women. J Clin Endocrinol Metab 2014;99:2448–55. osteoporosis. Elsevier Science; 2009.
[76] Bi WG, Nuyt AM, Weiler H, Leduc L, Santamaria C, Wei SQ. [94] Powe CE, Evans MK, Wenger J, Zonderman AB, Berg AH,
Association between vitamin D supplementation during preg- Nalls M, et al. Vitamin D-binding protein and vitamin D
nancy and offspring growth, morbidity, and mortality: a sys- status of black Americans and white Americans. N Engl J
tematic review and meta-analysis. JAMA Pediatr 2018;172:635–45. Med 2013;369:1991–2000.
[77] Picciano MF, Dwyer JT, Radimer KL, Wilson DH, Fisher KD, [95] Martin CA, Gowda U, Renzaho AM. The prevalence of
Thomas PR, et al. Dietary supplement use among infants, vitamin D deficiency among dark-skinned populations
children, and adolescents in the United States, 1999-2002. according to their stage of migration and region of birth: a
Arch Pediatr Adolesc Med 2007;161:978–85. meta-analysis. Nutrition 2016;32:21–32.
[78] Ahrens KA, Rossen LM, Simon AE. Adherence to vitamin D [96] Gangat M, Ponnapakkam T, Bradford E, Katikaneni R, Gensure
recommendations among US infants aged 0 to 11 months, R. Reversed seasonal variation in maternal vitamin D levels in
NHANES, 2009 to 2012. Clin Pediatr 2016;55:555–6. southern Louisiana. Clin Pediatr (Phila) 2012;51:718–22.
N U TR IT ION RE S EA R CH 6 3 ( 20 1 9 ) 1– 2 0 19

[97] Hoath SB, Maibach HI. Neonatal skin: structure and func- [118] Bodnar LM, Catov JM, Simhan HN, Holick MF, Powers RW,
tion. Taylor & Francis; 2003. Roberts JM. Maternal vitamin D deficiency increases the risk
[98] Walsh R. Variation in the melanin content of the skin of of preeclampsia. J Clin Endocrinol Metab 2007;92:3517–22.
New Guinea natives at different ages. J Invest Dermatol [119] Achkar M, Dodds L, Giguere Y, Forest JC, Armson BA,
1964;42:261–5. Woolcott C, et al. Vitamin D status in early pregnancy and
[99] Cuhaci-Cakir B, Demirel F. Effects of seasonal variation and risk of preeclampsia. Am J Obstet Gynecol 2015;212:511.e1-7.
maternal clothing style on vitamin D levels of mothers and [120] Baker AM, Haeri S, Camargo Jr CA, Espinola JA, Stuebe AM. A
their infants. Turk J Pediatr 2014;56:475–81. nested case-control study of midgestation vitamin D
[100] Dijkstra SH, van Beek A, Janssen JW, de Vleeschouwer LH, deficiency and risk of severe preeclampsia. J Clin Endocrinol
Huysman WA, van den Akker EL. High prevalence of Metab 2010;95:5105–9.
vitamin D deficiency in newborn infants of high-risk [121] Rostami M, Ramezani Tehrani F, Simbar M, Bidhendi
mothers. Arch Dis Child 2007;92:750–3. Yarandi R, Minooee S, Hollis BW, et al. Effectiveness of
[101] Davenport ML, Uckun A, Calikoglu AS. Pediatrician patterns prenatal vitamin D deficiency screening and treatment
of prescribing vitamin supplementation for infants: do they program: a stratified randomized field trial. J Clin Endocrinol
contribute to rickets? Pediatrics 2004;113:179–80. Metab 2018;103:2936–48.
[102] Shaikh U, Alpert PT. Practices of vitamin D recommendation [122] Mirzakhani H, Litonjua AA, McElrath TF, O'Connor G, Lee-
in Las Vegas, Nevada. J Hum Lact 2004;20:56–61. Parritz A, Iverson R, et al. Early pregnancy vitamin D status
[103] Taylor JA, Geyer LJ, Feldman KW. Use of supplemental and risk of preeclampsia. J Clin Invest 2016;126:4702–15.
vitamin D among infants breastfed for prolonged periods. [123] Ali AM, Alobaid A, Malhis TN, Khattab AF. Effect of vitamin
Pediatrics 2010;125:105–11. D3 supplementation in pregnancy on risk of pre-eclampsia
[104] World Health Organization. Infant and young child feeding : —randomized controlled trial. Clin Nutr 2018. https://doi.
model chapter for textbooks for medical students and allied org/10.1016/j.clnu.2018.02.023 [in Press].
health professionals. WHO Library Cataloguing-in- [124] Behjat Sasan S, Zandvakili F, Soufizadeh N, Baybordi E. The
Publication Data; 2009. effects of vitamin D supplement on prevention of recur-
[105] Sherman EM, Svec RV. Barriers to vitamin D supplementa- rence of preeclampsia in pregnant women with a history of
tion among military physicians. Mil Med 2009;174:302–7. preeclampsia. Obstet Gynecol Int 2017;2017. https://doi.org/
[106] Li W, Green TJ, Innis SM, Barr SI, Whiting SJ, Shand A, et al. 10.1155/2017/8249264.
Suboptimal vitamin D levels in pregnant women despite [125] Wei SQ, Qi HP, Luo ZC, Fraser WD. Maternal vitamin D status
supplement use. Can J Public Health 2011;102:308–12. and adverse pregnancy outcomes: a systematic review and
[107] Statistics Canada. Median total income, by family type, by meta-analysis. J Matern Fetal Neonatal Med 2013;26:889–99.
province and territory (all census families). http://www. [126] Organization WH. WHO recommendations for prevention
statcan.gc.ca/tables-tableaux/sum-som/l01/cst01/ and treatment of pre-eclampsia and eclampsia. http://apps.
famil108a-eng.htm; 2015, Accessed date: 12 March 2018. who.int/iris/bitstream/handle/10665/44703/9789241548335_
[108] Grant WB, Schwalfenberg GK, Genuis SJ, Whiting SJ. An eng.pdf;jsessionid=AE4940D67F2746CCE33DE3EE12713076?
estimate of the economic burden and premature deaths due sequence=1; 2011, Accessed date: 13 April 2016.
to vitamin D deficiency in Canada. Mol Nutr Food Res 2010; [127] Bellamy L, Casas JP, Hingorani AD, Williams D. Type 2
54:1172–81. diabetes mellitus after gestational diabetes: a systematic
[109] Wagner CL, McNeil RB, Johnson DD, Hulsey TC, Ebeling M, review and meta-analysis. Lancet 2009;373:1773–9.
Robinson C, et al. Health characteristics and outcomes of [128] Chiu KC, Chu A, Go VLW, Saad MF. Hypovitaminosis D is
two randomized vitamin D supplementation trials during associated with insulin resistance and beta cell dysfunction.
pregnancy: a combined analysis. J Steroid Biochem Mol Biol Am J Clin Nutr 2004;79:820–5.
2013;136:313–20. [129] Clifton-Bligh RJ, McElduff P, McElduff A. Maternal vitamin D
[110] Wortsman J, Matsuoka LY, Chen TC, Lu Z, Holick MF. deficiency, ethnicity and gestational diabetes. Diabet Med 2008;
Decreased bioavailability of vitamin D in obesity. Am J Clin 25:678–84.
Nutr 2000;72:690–3. [130] Farrant HJW, Krishnaveni GV, Hill JC, Boucher BJ, Fisher DJ,
[111] Cheng S, Massaro JM, Fox CS, Larson MG, Keyes MJ, McCabe EL, Noonan K, et al. Vitamin D insufficiency is common in Indian
et al. Adiposity, cardiometabolic risk, and vitamin D status: the mothers but is not associated with gestational diabetes or
Framingham heart study. Diabetes 2010;59:242–8. variation in newborn size. Eur J Clin Nutr 2009;63:646–52.
[112] Josefson JL, Reisetter A, Scholtens DM, Price HE, Metzger BE, [131] Makgoba M, Nelson SM, Savvidou M, Messow CM, Nicolaides
Langman CB, et al. Maternal BMI associations with maternal K, Sattar N. First-trimester circulating 25-hydroxyvitamin D
and cord blood vitamin D levels in a North American subset levels and development of gestational diabetes mellitus.
of Hyperglycemia and Adverse Pregnancy Outcome (HAPO) Diabetes Care 2011;34:1091–3.
study participants. PLoS One 2016;11:e0150221. [132] Parlea L, Bromberg I, Feig D, Vieth R, Merman E, Lipscombe L.
[113] Dietitians of Canada. BMI for adults. http://www.dietitians. Association between serum 25-hydroxyvitamin D in early
ca/your-health/assess-yourself/assess-your-bmi/bmi-adult. pregnancy and risk of gestational diabetes mellitus. Diabet Med
aspx; 2016, Accessed date: 14 June 2016. 2012;29:e25–32.
[114] Evans KN, Bulmer JN, Kilby MD, Hewison M. Vitamin D and [133] Lacroix M, Battista MC, Doyon M, Houde G, Menard J,
placental-decidual function. J Soc Gynecol Investig 2004;11: Ardilouze JL, et al. Lower vitamin D levels at first trimester
263–71. are associated with higher risk of developing gestational
[115] Cardus A, Parisi E, Gallego C, Aldea M, Fernandez E, diabetes mellitus. Acta Diabetol 2014;51:609–16.
Valdivielso JM. 1,25-dihydroxyvitamin D-3 stimulates vas- [134] Dodds L, Woolcott CG, Weiler H, Spencer A, Forest JC,
cular smooth muscle cell proliferation through a VEGF- Armson BA, et al. Vitamin D status and gestational diabetes:
mediated pathway. Kidney Int 2006;69:1377–84. effect of smoking status during pregnancy. Paediatr Perinat
[116] Aranow C. Vitamin D and the immune system. J Investig Epidemiol 2016;30:229–37.
Med 2011;59:881–6. [135] Soheilykhah S, Mojibian M, Moghadam MJ, Shojaoddiny-
[117] Li YC, Kong J, Wei MJ, Chen ZF, Liu SQ, Cao LP. 1,25- Ardekani A. The effect of different doses of vitamin D
Dihydroxyvitamin D-3 is a negative endocrine regulator of supplementation on insulin resistance during pregnancy.
the renin-angiotensin system. J Clin Invest 2002;110:229–38. Gynecol Endocrinol 2013;29:396–9.
20 N U TR IT ION RE S EA R CH 63 ( 20 1 9 ) 1 – 2 0

[136] Karamali M, Asemi Z, Ahmadi-Dastjerdi M, Esmaillzadeh A. [144] Aspray TJ, Bowring C, Fraser W, Gittoes N, Javaid MK,
Calcium plus vitamin D supplementation affects preg- Macdonald H, et al. National Osteoporosis Society vitamin D
nancy outcomes in gestational diabetes: randomized, guideline summary. Age Ageing 2014;43:592–5.
double-blind, placebo-controlled trial. Public Health Nutr [145] Pludowski P, Karczmarewicz E, Bayer M, Carter G, Chlebna-
2016;19:156–63. Sokol D, Czech-Kowalska J, et al. Practical guidelines for the
[137] Weinert LS, Reichelt AJ, Schmitt LR, Boff R, Oppermann MLR, supplementation of vitamin D and the treatment of deficits
Camargo JL, et al. Vitamin D deficiency increases the risk of in Central Europe—recommended vitamin D intakes in the
adverse neonatal outcomes in gestational diabetes. PLoS general population and groups at risk of vitamin D
One 2016;11:e0164999. deficiency. Endokrynol Pol 2013;64:319–27.
[138] Asemi Z, Karamali M, Esmaillzadeh A. Favorable effects of [146] German Nutrition S. New reference values for vitamin D.
vitamin D supplementation on pregnancy outcomes in Ann Nutr Metab 2012;60:241–6.
gestational diabetes: a double blind randomized controlled [147] Nordic Council of Ministers. Nordic nutrition recommenda-
clinical trial. Horm Metab Res 2015;47:565–70. tions 2012. Integrating nutrition and physical activity.
[139] Diaz-Gomez NM, Mendoza C, Gonzalez-Gonzalez NL, http://www.norden.org/en/theme/former-themes/themes-
Barroso F, Jimenez-Sosa A, Domenech E, et al. Maternal 2016/nordic-nutrition-recommendation/nordic-nutrition-
smoking and the vitamin D-parathyroid hormone recommendations-2012; 2014, Accessed date: 13 April 2016.
system during the perinatal period. J Pediatr 2007;151: [148] Health Council of the Netherlands. Evaluation of dietary
618–23. reference values for vitamin D. https://www.
[140] U.S. Department of Health and Human Services, Centres for gezondheidsraad.nl/en/task-and-procedure/areas-of-
Disease Control and Prevention, National Center for Chronic activity/healthy-nutrition/evaluation-of-the-dietary-
Disease Prevention and Health Promotion, Office on reference-values; 2012, Accessed date: 13 April 2016.
Smoking an Health. The health consequences of involun- [149] Nowson CA, McGrath JJ, Ebeling PR, Haikerwal A, Daly RM,
tary exposure to tobacco smoke. A report of the Surgeon Sanders KM, et al. Vitamin D and health in adults in
General. http://www.surgeongeneral.gov/library/ Australia and New Zealand: a position statement. Med J
secondhandsmoke/index.html; 2006, Accessed date: 13 Aust 2012;196:686–7.
April 2016. [150] Diamond TH, Eisman JA, Mason RS, Nowson CA, Pasco JA,
[141] U.S. Department of Health and Human Services, Centers for Sambrook PN, et al. Vitamin D and adult bone health in
Disease Control and Prevention, National Center for Chronic Australia and New Zealand: a position statement. Med J Aust
Disease Prevention and Health Promotion, Office on 2005;182:281–5.
Smoking and Health. A report of the Surgeon General: how [151] Dror DK, King JC, Fung EB, Loan MDV, Gertz ER, Allen LH.
tobacco smoke causes disease the biology and behavioral Evidence of associations between feto-maternal vitamin D
basis for smoking-attributable disease. http://www. status, cord parathyroid hormone and bone-specific alka-
surgeongeneral.gov/library/tobaccosmoke/report/chapter8. line phosphatase, and newborn whole body bone mineral
pdf; 2010, Accessed date: 13 April 2016. content. Nutrients 2012;4:68–77.
[142] Grober U, Kisters K. Influence of drugs on vitamin D and [152] Wang C, Gao JS, Yu SL, Qiu L, Zeng L, Wang DH. Correlation
calcium metabolism. Dermatoendocrinol 2012;4:158–66. between neonatal vitamin D level and maternal vitamin D
[143] Ross AC, Manson JE, Abrams SA, Aloia JF, Brannon PM, level. Zhongguo Dang Dai Er Ke Za Zhi 2016;18:20–3.
Clinton SK, et al. The 2011 report on dietary reference [153] Banihosseini SZ, Baheiraei A, Shirzad N, Heshmat R,
intakes for calcium and vitamin D from the Institute of Mohsenifar A. The effect of cigarette smoke exposure on
Medicine: what clinicians need to know. J Clin Endocrinol vitamin D level and biochemical parameters of mothers and
Metab 2011;96:53–8. neonates. J Diabetes Metab Disord 2013;12:19.

Das könnte Ihnen auch gefallen