Influence of Vitamin D Supplementation on Growth, Body Composition, and Pubertal Development Among School-aged Children in an Area With a High Prevalence of Vitamin D Deficiency: A Randomized Clinical Trial (2024)

  • Journal List
  • JAMA Network
  • PMC9706398

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

Influence of Vitamin D Supplementation on Growth, Body Composition, and Pubertal Development Among School-aged Children in an Area With a High Prevalence of Vitamin D Deficiency: A Randomized Clinical Trial (1)

JAMA NetworkView Article

JAMA Pediatrics

JAMA Pediatr. 2023 Jan; 177(1): 32–41.

Published online 2022 Nov 28. doi:10.1001/jamapediatrics.2022.4581

PMCID: PMC9706398

PMID: 36441522

A Randomized Clinical Trial

Davaasambuu Ganmaa, PhD,Influence of Vitamin D Supplementation on Growth, Body Composition, and Pubertal Development Among School-aged Children in an Area With a High Prevalence of Vitamin D Deficiency: A Randomized Clinical Trial (2)1,2 Sabri Bromage, ScD,2 Polyna Khudyakov, PhD,3 Sumiya Erdenenbaatar, BSc,4 Baigal Delgererekh, MD,5 and Adrian R. Martineau, PhDInfluence of Vitamin D Supplementation on Growth, Body Composition, and Pubertal Development Among School-aged Children in an Area With a High Prevalence of Vitamin D Deficiency: A Randomized Clinical Trial (3)6

Author information Article notes Copyright and License information PMC Disclaimer

Associated Data

Supplementary Materials

Key Points

Question

Does oral vitamin D replacement influence linear growth, body composition, or pubertal development in school-aged children living in areas with a high prevalence of vitamin D deficiency?

Findings

This secondary analysis of a randomized clinical trial including 8851 children with a high prevalence of vitamin D deficiency at baseline found that oral vitamin D supplementation was effective in elevating 25(OH)D levels into the physiological range. However, this intervention had no impact on height for age, body mass index, body composition, or pubertal development, either in the study population as a whole or within subgroups defined by baseline 25(OH)D concentration, estimated calcium intake, and sex.

Meaning

In this study, weekly oral administration of vitamin D for 3 years did not influence linear growth, body composition, or pubertal development among school-aged children with a high prevalence of vitamin D deficiency at baseline.

This secondary analysis of a randomized clinical trial evaluates the effect of vitamin D supplementation on linear growth, body composition, and pubertal development in school-aged children with a high prevalence of vitamin D deficiency.

Abstract

Importance

Vitamin D deficiency (defined as 25-hydroxyvitamin D [25(OH)D] <20 ng/mL) is prevalent among children living in temperate climates and has been reported to associate independently with stunting, obesity, and early activation of the hypothalamic-pituitary-gonadal axis. Phase 3 randomized clinical trials to investigate the influence of long-term vitamin D replacement on growth, body composition, and pubertal development of school-aged children with vitamin D deficiency are lacking.

Objective

To determine whether weekly oral vitamin D supplementation influences linear growth, body composition, or pubertal development in school-aged children living in a setting where vitamin D deficiency is highly prevalent.

Design, Setting, and Participants

This secondary analysis of a double-blind, placebo-controlled randomized clinical trial was conducted from June 2016 to June 2019 at 18 grade schools in Ulaanbaatar, Mongolia. School-aged children (6 to 13 years at baseline) attending participating schools were included. Exclusion criteria included a positive QuantiFERON-TB Gold in-tube assay result, conditions or medications associated with altered vitamin D metabolism, use of vitamin D supplements, signs of rickets, or intention to move from Ulaanbaatar within 4 years. Of 11 475 children invited to participate in the study, 9814 underwent QFT testing, and 8851 with negative results were included in the study. All but 1 participant in the placebo group completed follow-up and were included in the present analysis. Data were analyzed from November 2021 to February 2022.

Interventions

Weekly oral doses of vitamin D3, 14 000 IU, (n = 4418), or placebo (n = 4433) for 3 years.

Main Outcomes and Measures

Mean z scores for height for age, body mass index for age, and waist-to-height ratio; mean percentage body fat, fat mass, and fat-free mass; and mean Tanner scores for pubertal development.

Results

Of 8851 participants, 4366 (49.3%) were female, and 8165 (92.2%) were of Khalkh ethnicity; the mean (SD) age was 9.4 (1.6) years. A total of 8453 participants (95.5%) were vitamin D deficient at baseline, and mean end-of-study 25(OH)D concentrations among participants randomized to vitamin D vs placebo were 31.0 vs 10.7 ng/mL (mean difference, 20.3; 95% CI; 19.9-20.6). However, vitamin D supplementation did not influence mean height for age, body mass index for age, waist-to-height ratio, percentage body fat, fat mass, fat-free mass, or Tanner scores, either overall or within subgroups defined by baseline 25(OH)D concentration less than 10 ng/mL vs 10 ng/mL or greater, estimated calcium intake less than 500 mg/d vs 500 mg/d or greater, or male vs female sex.

Conclusions and Relevance

In school-aged children in this study with low baseline vitamin D status, oral vitamin D3 supplementation at a dose of 14 000 IU per week for 3 years was effective in elevating 25(OH)D concentrations but did not influence growth, body composition, or pubertal development.

Trial Registration

ClinicalTrials.gov Identifier: NCT02276755

Introduction

The US Institute of Medicine recommends maintenance of 25(OH)D concentrations of 20 ng/mL or higher (to convert to nanomoles per liter, multiply by 2.496) to support optimal skeletal health in children.1 Vitamin D affects growth by promoting adequate bone mineralization and mass and influences somatic growth and macronutrient metabolism through regulation of the cell cycle and cell proliferation.2 It may also exert effects on body composition via effects on myocyte function and development3 and by regulating insulin signaling, adipogenesis, and adipocyte apoptosis.4 Through its actions on macrophage, dendritic, and T cell function, vitamin D is a potent mediator of innate and adaptive immunity,5 and vitamin D deficiency may contribute to systemic inflammation.6 Thus, by modulating diverse physiological pathways, vitamin D status is a potentially important determinant of growth, body composition, and metabolic health—a concept supported by the observation that these diverse axes are simultaneously perturbed in animal models of vitamin D deficiency.7 Given the high global prevalence of vitamin D deficiency8 and the rising prevalence of cardiometabolic disease,9 there is interest in evaluating the potential relationship between vitamin D status—an actionable target for supplementation and fortification—and measures of metabolic health and fitness.

A potential role for vitamin D supplementation in improving anthropometric and metabolic outcomes is supported by findings of increased lean mass, decreased fat mass, and decreased body mass index (BMI; calculated as weight in kilograms divided by height in meters squared) in long-term follow-up with infants who received vitamin D supplementation10,11 and observational studies reporting independent associations between vitamin D deficiency and underweight and stunting in infants and toddlers12; lower lean mass,13 slowed growth,14 childhood obesity15,16,17; and precocious puberty.18 Findings of observational studies may be explained by confounding or reverse causation, and randomized clinical trials of vitamin D supplementation are required to evaluate causality. We have previously reported results of a phase 2 randomized clinical trial showing that a 6-month course of vitamin D supplementation increased height gain in 113 Mongolian children aged 12 to 15 years with vitamin D deficiency.19 We therefore wished to verify this finding by conducting a definitive phase 3 randomized clinical trial of sustained vitamin D replacement in a similar population and extend the investigation to test for potential effects of vitamin D on BMI, body composition, and pubertal development.

Methods

Study Design, Setting, Participants, and Randomization

We conducted a parallel 2-arm double-blind individually randomized placebo-controlled trial from June 2016 to June 2019 in 18 public schools in Ulaanbaatar, Mongolia, as previously described.20 The primary outcome of the trial was acquisition of latent tuberculosis infection. This study assessed the effects of the intervention on prespecified secondary outcomes relating to growth, body composition, and pubertal development. Principal inclusion criteria were ages 6 to 13 years at screening and attendance at a participating school; principal exclusion criteria were a positive QuantiFERON-TB Gold in-tube assay (QFT) result, presence of conditions associated with vitamin D hypersensitivity (primary hyperparathyroidism or sarcoidosis) or immunocompromise (taking immunosuppressant medication or cytotoxic therapy), use of vitamin D supplements, signs of rickets (all participants were screened for rickets via physical examination by a pediatrician), or intention to move from Ulaanbaatar within 4 years of enrollment. Of 11 475 children invited to participate in the study, 9814 underwent QFT testing, and 8851 with negative results were included in the study. All but 1 participant in the placebo group completed follow-up and were included in the present analysis. Eligible participants were individually randomized to receive a weekly capsule containing 14 000 IU vitamin D3 or placebo for 3 years with a 1-to-1 allocation ratio and stratification by school of attendance. Treatment allocation was concealed from participants, clinicians, and all trial staff (including senior investigators and those assessing outcomes) so that the double-blinding was maintained. Each child and their parent or guardian provided written informed assent and consent, respectively, prior to participation. Because Khalkha is the largest ethnic group in Mongolia (constituting more than 80% of the population), Khalkh vs non-Khalkh ethnicity was gathered by interview. The study was approved by institutional review boards of the Mongolian Ministry of Health, Mongolian National University, and Harvard T. H. Chan School of Public Health. The study report followed the Consolidated Standards of Reporting Trials (CONSORT) reporting guideline. Data were analyzed from November 2021 to February 2022. The trial protocol can be found in Supplement 1.

Baseline Procedures

At baseline, participants’ parents were asked to complete questionnaires detailing their socioeconomic circ*mstances, lifestyle, and dietary factors influencing vitamin D status, including intake of foods previously shown to be major contributors to dietary calcium intake in urban Mongolia.21 Participating children’s height was then measured using a portable stadiometer (seca GmbH & Co), weight was measured using a digital floor scale (seca GmbH & Co), waist circumference was measured using a tape measure, and percentage body fat, fat mass, and fat-free mass were determined using the SC-331S body composition analyzer (Tanita). Finally, a blood sample was drawn for separation and storage of serum for determination of baseline 25(OH)D concentrations.

Follow-up Procedures

During school terms, study participants had weekly in-person visits at which time study medication was administered and adverse events were recorded. During school holidays, either children were given a single bolus dose of up to 36 000 IU (shorter holidays), study staff traveled to participants’ homes to administer medication, or parents were supplied with sufficient trial medication to cover the holiday period, along with instructions on its storage and administration. Weight, height, waist circumference, and body composition were reassessed at 12, 24, and 36 (end of study) months’ follow-up using the same methods as at baseline. Pubertal status was evaluated at 3-year follow-up in a randomly selected subset of 1356 participants who completed Tanner self-assessment questionnaires.22 A second blood sample was drawn from all participants at 3-year follow-up for separation and storage of serum for determination of end-of-study 25(OH)D concentrations.

Measurement of Vitamin D Status

Concentrations of 25(OH)D were determined in serum samples from baseline and 3-year follow-up using the VIDAS 25OH Vitamin D total enzyme-linked fluorescent assay (bioMérieux). These measurements were validated and standardized using standards provided by the Vitamin D External Quality Assessment Scheme.23 Total coefficient of variation was 7.9%, mean bias was 7.7%, and the limit of quantitation was 8.1 ng/mL. Nonzero 25(OH)D values were standardized using a published method,24 using a set of 40 serum samples provided by the Vitamin D External Quality Assessment Scheme (DEQAS). Values below the limit of quantitation were classified as less than 8.1 ng/mL.

Outcomes

The primary anthropometric outcome of interest was mean height-for-age z score at 1, 2, and 3 years’ follow-up. Secondary outcomes were mean BMI-for-age z score; mean waist-to-height ratio z score; mean percentage body fat; mean fat mass; mean fat-free mass; mean Tanner scores for pubic hair (male and female), external genitalia (male only), and breast development (female only); the proportion of participants reaching menarche by the end of the trial (female only); and mean age at onset of menarche (female individuals who had reached menarche by the end of the trial only).

Sample Size and Statistical Methods

The sample size calculation was based on the power needed to detect a clinically significant effect of the intervention on the primary end point (incident latent tuberculosis infection) as described previously.20 Estimated calcium intake values were calculated on the basis of parental responses to a 1-time food frequency questionnaire administered in March 2018 (eTable 1 in Supplement 2), which captured participants’ frequency of intake of certain calcium-containing foods and the calcium content of those foods based on food composition data compiled from analysis of their calcium content.21 Anthropometric measurements and data on participants’ age and sex were then used to compute z scores for height for age, BMI for age, and waist-to-height ratio for age using the who2007 Shiny App (Canadian Pediatric Endocrine Group) based on 2007 World Health Organization growth reference data for children aged 5 to 19 years.25

Serum 25(OH)D values were adjusted for seasonal variation prior to analysis using a sinusoidal model.26 Anthropometric and body composition outcomes were analyzed overall and in each subgroup using mixed models for repeated measures with fixed effects for treatment and time and treatment-by-time interaction adjusted for school of attendance and random effects for individuals. Adjusted treatment mean differences at different time points were presented with 95% CIs, and significance tests were conducted for the treatment effect at each time point and the overall treatment-by-time interaction. Where overall P values were less than the significance threshold of .05, we applied a Benjamini Hochberg procedure with a 5% false discovery rate27 to the relevant family of P values to adjust for multiple comparisons. End-of-study pubertal development analyses were done using general linear models with gaussian distribution and identity link function for all outcomes except for the proportion of female participants menstruating at the end of the trial, which was analyzed using binomial distribution and log link function. Prespecified subgroup analyses were conducted according to participants’ sex (male vs female), estimated calcium intake (<500 mg/d vs ≥500 mg/d), baseline deseasonalized 25(OH)D concentration (<10 ng/mL vs ≥10 ng/mL), and baseline z scores (−2.00 vs ≥−2.00 for height for age and <−2.00 vs −2.00 to 1.00 vs 1.01 to 2.00 vs >2.00 for BMI for age and waist-to-height ratio, percentage body fat, fat mass, and fat-free mass). An exploratory subgroup analysis was conducted according to participants’ age at baseline (≤8 vs >8 years) in response to a reviewer request. The primary P values for outcome modeling were the overall P values, that is, those associated with the interaction between follow-up time point and treatment allocation.

Results

Participants

Of 8851 children with negative QFT results who were included in the trial (mean [SD] age, 9.4 [1.6] years; 4366 [49.3%] female; 8165 [92.2%] of Khalkh ethnicity), 4418 were randomly assigned to receive vitamin D3 and 4433 to receive placebo, all but 1 of whom (allocated to placebo) had anthropometry data available at baseline and contributed to the current analysis (Figure). Table 1 shows baseline characteristics of participants contributing data to analyses of anthropometric outcomes. Mean (SD) baseline serum 25(OH)D concentration was 11.9 (4.2) ng/mL; 8453 participants (95.5%) had 25(OH)D levels less than 20 ng/mL and 2813 (31.8%) less than 10 ng/mL. Baseline prevalence of stunting was 3.7%; underweight, 1.2%; overweight, 4.5%; and obesity, 1.1%. Baseline characteristics were balanced between participants randomized to vitamin D vs placebo, both for those contributing data to anthropometric analyses and for the subset of 1356 participants whose pubertal status was assessed at the end of the trial (eTable 2 in Supplement 2). End-of-trial anthropometric outcomes were available for 8128 participants (91.8% of those randomized). Mean (SD) end-of-trial serum 25(OH)D was 31.0 (9.1) ng/mL in the treatment group and 10.7 (5.3) in the placebo group (mean difference, 20.3; 95% CI, 19.9-20.6). End-of-study 25(OH)D levels were 20 ng/mL or higher in 3528 participants randomized to vitamin D (89.8%) vs 187 randomized to placebo (5.6%).

Open in a separate window

Figure.

Participant Flow Diagram

Table 1.

Baseline Characteristics of Participants by Allocation

CharacteristicNo. (%)
Overall (n = 8851)Placebo (n = 4432)Vitamin D (n = 4418)
Age, mean (SD), y9.4 (1.6)9.4 (1.6)9.4 (1.6)
Female4366 (49.3)2224 (50.2)2142 (48.5)
Male4485 (50.7)2209 (49.8)2276 (51.5)
Ethnic origina
Khalkh8165 (92.2)4103 (92.6)4062 (91.9)
Non-Khalkh686 (7.8)330 (7.4)356 (8.1)
Parental educationb
≤Secondary school4858 (54.9)2457 (55.4)2401 (54.3)
≥University/polytechnic school3993 (45.1)1976 (44.6)2017 (45.7)
Type of residence
Ger (yurt)3271 (37.0)1628 (36.7)1643 (37.2)
House without central heating3387 (38.3)1722 (38.9)1665 (37.7)
House with central heating2193 (24.8)1083 (24.4)1110 (25.1)
Home ownership, yes6963 (78.7)3470 (78.3)3493 (79.1)
Monthly household income, mean (SD), $c848 (579)846 (604)851 (554)
Household environmental tobacco smoked3143 (35.5)1573 (35.5)1570 (35.5)
Child actively smoking47 (0.5)21 (0.5)26 (0.6)
BMI-for-age z score, mean (SD)c0.2 (1.1)0.2 (1.1)0.2 (1.0)
Height-for-age z score, mean (SD)c−0.3 (1.0)−0.3 (1.0)−0.3 (1.0)
Serum 25(OH)D, mean (SD), ng/mLc,e11.9 (4.2)11.9 (4.2)11.9 (4.2)
Serum 25(OH)D, ng/mLc,e
<102813 (31.8)1420 (32.1)1393 (31.5)
10-19.95640 (63.7)2812 (63.5)2828 (64.0)
20-29.9381 (4.3)193 (4.4)188 (4.3)
≥3012 (0.1)4 (0.1)8 (0.2)

Open in a separate window

Abbreviations: 25(OH)D, 25-hydroxyvitamin D; BMI, body mass index.

SI conversion factor: To convert 25(OH)D to nmol/L, multiply by 2.496.

aBecause Khalkha is the largest ethnic group in Mongolia (constituting more than 80% of the population), Khalkh vs non-Khalkh ethnicity was gathered by interview.

bHighest educational level attained by either parent.

cHousehold income missing for 1 participant in each arm; baseline BMI-for-age z score missing for 1 participant in placebo arm; baseline height-for-age z score missing for 1 participant in placebo arm; baseline serum 25(OH)D missing for 4 participants in placebo arm and 1 participant in vitamin D arm.

dDefined as 1 or more people in the household smoking indoors.

eBaseline 25(OH)D values deseasonalized.

Outcomes

Allocation to vitamin D vs placebo did not influence mean height-for-age z score, either overall or within subgroups defined by male vs female sex, age at baseline 8 years or younger vs older than 8 years, estimated calcium intake less than 500 vs 500 or more mg/d, baseline 25(OH)D concentration less than 10 vs 10 or more ng/mL, or presence vs absence of stunting at baseline (Table 2). Similarly, no effect of the intervention was seen on mean BMI-for-age z score (Table 3), mean waist-to-height ratio z score (eTable 3 in Supplement 2), mean percentage body fat (eTable 4 in Supplement 2), mean fat mass (eTable 5 in Supplement 2), or mean fat-free mass (eTable 6 in Supplement 2), either overall or by subgroups defined by sex, age, estimated calcium intake, baseline 25(OH)D concentration, or baseline anthropometry. Among the subset of 1356 participants in whom end-of-trial pubertal development was assessed, no interarm differences were seen in mean Tanner scores for pubic hair (male and female), external genitalia (male only), or breast development (female only), either overall or within subgroups defined by calcium intake and baseline 25(OH)D concentration (Table 4). When Tanner scores were dichotomized to distinguish prepubertal vs pubertal status (Tanner score 1 vs 2-5, respectively), the proportions of participants reporting Tanner score 2 to 5 did not differ between study arms, either overall or by subgroup (eTable 7 in Supplement 2). The proportion of female participants reaching menarche by the end of the trial did not differ between arms, either overall or by subgroup. Among female participants who reached menarche by the end of the trial, no difference in mean age at onset of menarche was seen between arms, either overall or by subgroup (Table 4).

Table 2.

Mean Height-for-Age z Scores by Allocation, Overall and by Subgroup

GroupFollow-up time point, yMean (SD); No.Adjusted mean difference (95% CI)aP value for time pointOverall P valueb
PlaceboVitamin D
Overall1−.0.29 (1.00); 4230−0.30 (1.00); 4219−0.01 (−0.02 to 0.01).24.45
2−0.27 (1.00); 3830−0.30 (1.00); 3803−0.01 (−0.03 to 0.00).17
3−0.17 (1.00); 4052−0.17 (1.02); 40760.00 (−0.02 to 0.01).80
By sex
Female1−0.35 (1.02); 2126−0.34 (0.99); 2036−0.01 (−0.04 to 0.01).20.48
2−0.34 (1.00); 1927−0.32 (0.99); 1851−0.01 (−0.03 to 0.02).66
3−0.22 (0.99); 2043−0.19 (0.98); 19750.00 (−0.02 to 0.02).84
Male1−0.23 (0.98); 2104−0.26 (1.00); 21830.00 (−0.02 to 0.02).77.49
2−0.21 (1.01); 1903–0.29 (1); 1952−0.01 (−0.03 to 0.00).14
3−0.12 (1.03); 2009−0.16 (1.05); 21010.00 (−0.02 to 0.01).64
By age at baseline, y
≤81−0.24 (1.00); 1076−0.24 (0.92); 1079−0.02 (−0.05 to 0.01).12.42
2−0.21 (0.99); 1040−0.21 (0.94); 1049−0.01 (−0.04 to 0.01).37
3−0.13 (1.03); 1027−0.11 (0.98); 1056−0.02 (−0.04 to 0.01).21
>81−0.31 (1.00); 3239−0.32 (1.02); 3216−0.01 (−0.02 to 0.01).52.52
2−0.30 (1.01); 2868−0.34 (1.01); 2822−0.01 (−0.03 to 0.01).22
3−0.19 (1.00); 3106−0.20 (1.03); 30940.00 (−0.02, 0.02).90
By calcium intake
<500 mg/d1−0.30 (1.00); 2171−0.30 (1.02); 2161−0.01 (−0.03 to 0.01).50.42
2−0.29 (1.01); 1973−0.33 (1.01); 1927−0.01 (−0.03 to 0.01).23
3−0.19 (1.02); 2141−0.18 (1.02); 21430.00 (−0.02 to 0.02).71
≥500 mg/d1−0.25 (0.99); 1210−0.27 (0.98); 1261−0.01 (−0.03 to 0.01).50.42
2−0.24 (1.01); 1095−0.27 (0.98); 1151−0.01 (−0.03 to 0.01).23
3−0.10 (1.01); 1194−0.14 (1.01); 12410.00 (−0.02 to 0.02).71
By baseline 25(OH)D concentration, ng/mLc
<101−0.33 (1.02); 1348−0.31 (0.98); 13400.01 (−0.02 to 0.03).56.90
2−0.33 (1.02); 1223−0.32 (0.98); 12120.00 (−0.02 to 0.03).75
3−0.24 (1.00); 1305−0.21 (1.00); 12900.01 (−0.02 to 0.04).46
≥101−0.28 (0.99); 2878−0.29 (1.00); 2878−0.02 (−0.03 to 0.00.08.20
2−0.25 (1.00); 2604−0.29 (1.01); 2590−0.02 (−0.03 to 0.00).07
3−0.14 (1.01); 2744−0.16 (1.02); 2785−0.01 (−0.02 to 0.01).43
By presence/absence of stuntingd at baseline
Present1−2.37 (0.60); 164−2.38 (0.41); 149−0.04 (−0.13 to 0.05).43.78
2−2.30 (0.62); 155−2.28 (0.51); 144−0.01 (−0.10 to 0.08).78
3−2.10 (0.72); 153−2.13 (0.62); 140−0.04 (−0.13 to 0.05).37
Absent1−0.21 (0.92); 4065−0.22 (0.93); 4070−0.02 (−0.06 to 0.02).38.56
2−0.19 (0.93); 3674−0.23 (0.93); 3659−0.02 (−0.06 to 0.02).31
3−0.10 (0.94); 3898−0.10 (0.96); 3936−0.01 (−0.05 to 0.03).62

Open in a separate window

Abbreviation: 25(OH)D, 25-hydroxyvitamin D.

SI conversion factor: To convert 25(OH)D to nmol/L, multiply by 2.496.

aAdjusted for random effects of school and individual.

bP value for treatment-by-time interaction.

c25(OH)D values deseasonalized.

dStunting was defined as weight-for-height z score <−2.00.

Table 3.

Mean BMIa-for-Age z Scores by Allocation, Overall and by Subgroup

GroupFollow-up time point, yMean (SD), No.Adjusted mean difference (95% CI)bP value for time pointOverall P valuec
PlaceboVitamin D
Overall10.17 (1.08); 42290.16 (1.08); 42190.00 (−0.01 to 0.02).70.95
20.19 (1.08); 38290.16 (1.09); 38030.00 (–0.02 to 0.02).90
30.15 (1.11); 40510.14 (1.12); 40760.00 (–0.02 to 0.02).76
By sex
Male10.31 (1.11); 21040.26 (1.11); 21830.01 (–0.02 to 0.04).47.91
20.31 (1.13); 19030.23 (1.12); 19520.01 (–0.02 to 0.03).66
30.22 (1.17); 20090.16 (1.17); 21010.00 (–0.02 to 0.03).72
Female10.03 (1.03); 21250.06 (1.03); 20360.00 (–0.03 to 0.02).96.78
20.07 (1.02); 19260.10 (1.05); 1851–0.01 (–0.03 to 0.02).71
30.08 (1.04); 20420.13 (1.07); 19750.01 (–0.02 to 0.03).50
By age at baseline, y
≤810.35 (1.09); 10760.33 (1.02); 10790.03 (–0.01 to 0.06).17.17
20.38 (1.11); 10400.37 (1.08); 10490.04 (0.00 to 0.07).05
30.35 (1.16); 10270.36 (1.13); 10560.04 (0.00 to 0.07).06
>810.11 (1.07); 32390.11 (1.09); 32160.00 (–0.02 to 0.02).76.50
20.12 (1.07); 28680.09 (1.08); 2822–0.02 (–0.04 to 0.01).15
30.08 (1.08); 31060.07 (1.11); 3094–0.01 (–0.03 to 0.01).42
By calcium intake
<500 mg/d10.16 (1.08); 21710.16 (1.07); 21610.01 (–0.02 to 0.03).54.94
20.18 (1.08); 19730.17 (1.06); 19270.00 (–0.02 to 0.03).76
30.14 (1.11); 21410.15 (1.10); 21430.01 (–0.02 to 0.03).69
≥500 mg/d10.18 (1.06); 12100.20 (1.10); 12610.00 (–0.03 to 0.04).77.96
20.19 (1.07); 10950.22 (1.12); 11510.01 (–0.02 to 0.04).61
30.16 (1.09); 11940.21 (1.16); 12410.00 (–0.03 to 0.03).94
By baseline 25(OH)D concentration, mg/ML
<1010.08 (1.08); 13470.14 (1.05); 13400.00 (–0.02 to 0.02).83.86
20.07 (1.08); 12220.15 (1.06); 1212–0.01 (–0.03 to 0.02).61
30.04 (1.10); 13040.10 (1.11); 12900.00 (–0.02 to 0.03).76
≥1010.22 (1.08); 28780.17 (1.09); 2878–0.26 (–0.47 to –0.05).01.01d
20.24 (1.08); 26040.17 (1.10); 2590–0.26 (–0.49 to –0.04).02
30.20 (1.10); 27440.16 (1.13); 2785–0.25 (–0.46 to –0.05).02
By baseline BMIa category
Underweighte1−2.05 (0.61); 48−2.27 (0.53); 540.00 (–0.04 to 0.04).981.00
2−1.95 (0.72); 39−2.20 (0.59); 470.00 (–0.04 to 0.03).86
3−1.97 (0.84); 49−2.18 (0.76); 550.00 (–0.04 to 0.03).88
Normal rangef1−0.16 (0.76); 3381−0.16 (0.74); 33860.05 (–0.01 to 0.11).12.29
2−0.14 (0.79); 3072−0.14 (0.79); 30850.04 (–0.02 to 0.10).22
3−0.17 (0.83); 3240−0.16 (0.85); 32700.06 (0.00 to 0.12).05
Overweightg11.35 (0.50); 5621.40 (0.48); 5420.02 (–0.05 to 0.09).55.92
21.37 (0.56); 5081.41 (0.57); 4710.02 (–0.06 to 0.09).67
31.30 (0.63); 5361.36 (0.63); 5250.02 (–0.05 to 0.09).58
Obeseh12.55 (0.65); 2382.51 (0.59); 2370.04 (–0.13 to 0.20).68.93
22.50 (0.61); 2102.48 (0.60); 2000.04 (–0.14 to 0.22).65
32.37 (0.65); 2262.34 (0.61); 226–0.01 (–0.18 to 0.16).92

Open in a separate window

Abbreviations: BMI, body mass index; 25(OH)D, 25-hydroxyvitamin D.

SI conversion factor: To convert 25(OH)D to nmol/L, multiply by 2.496.

aCalculated as weight in kilograms divided by height in meters squared.

bAdjusted for random effects of school and individual.

cP value for treatment-by-time interaction.

dAfter adjustment for multiple comparisons using the Benjamini-Hochberg procedure, this P value was .14.

eDefined as BMI-for-age z score <–2.00.

fDefined as BMI-for-age z score −2.00 to 1.00.

gDefined as BMI-for-age z score 1.01 to 2.00.

hDefined as BMI-for-age z score >2.00.

Table 4.

End-of-Study Stage of Pubertal Development by Allocation and Sex, Overall and by Subgroup

Outcome measureSubgroup Mean (SD); No.Adjusted mean difference (95% CI)aP valueAdjusted risk ratio (95% CI)aP value
PlaceboVitamin D
Male
Pubic hair, Tanner score
OverallNA2.02 (1.06); 3582.06 (1.04); 3370.06 (−0.10 to 0.22).48NANA
Calcium intake<500 mg/d1.96 (1.05); 1922.09 (1.05); 1840.15 (−0.06 to 0.36).17NANA
≥500 mg/d2.04 (1.09); 972.01 (1.01); 1040.04 (−0.26 to 0.34).79NANA
Baseline 25(OH)D concentration, ng/mL<10 ng/mL2.31 (1.15); 1292.21 (1.13); 980.09 (−0.21 to 0.39).56NANA
≥10 ng/mL1.86 (0.98); 2292.00 (0.99); 2390.14 (−0.04 to 0.32).13NANA
External genitalia, Tanner score
OverallNA2.18 (1.02); 3582.14 (0.98); 337−0.04 (−0.19 to 0.11).59NANA
Calcium intake<500 mg/d2.11 (1.01); 1922.14 (0.97); 1840.04 (−0.16 to 0.24).70NANA
≥500 mg/d2.26 (1.01); 972.15 (1.02); 104−0.05 (−0.34 to 0.24).74NANA
Baseline 25(OH)D concentration, ng/mL<10 ng/mL2.36 (1.04); 1292.31 (1.08); 980.06 (−0.23 to 0.34).70NANA
≥10 ng/mL2.09 (1); 2292.07 (0.93); 239−0.01 (−0.18 to 0.17).94NANA
Female
Pubic hair, Tanner score
OverallNA2.08 (1.08); 3272.13 (0.99); 3340.05 (−0.11 to 0.20).57NANA
Calcium intake<500 mg/d2.07 (1.09); 1682.15 (0.99); 1960.06 (−0.15 to 0.28).57NANA
≥500 mg/d2.09 (1.07); 1042.17 (1.03); 880.05 (−0.25 to 0.34).75NANA
Baseline 25(OH)D concentration, ng/mL<10 ng/mL2.22 (1.12); 1252.16 (1.01); 135−0.10 (−0.35 to 0.15).43NANA
≥10 ng/mL2.00 (1.04); 2022.12 (0.97); 1990.12 (−0.08 to 0.31).25NANA
Breast development, Tanner score
OverallNA2.61 (0.93); 3272.69 (0.94); 3340.07 (−0.07 to 0.21).33NANA
Calcium intake<500 mg/d2.62 (0.95); 1682.72 (0.97); 1960.08 (−0.12 to 0.28).44NANA
≥500 mg/d2.63 (0.92); 1042.55 (0.84); 88−0.12 (−0.67 to 0.14).20NANA
Baseline 25(OH)D concentration, ng/mL<10 ng/mL2.67 (0.97); 1252.69 (0.97); 1350.01 (−0.22 to 0.24).92NANA
≥10 ng/mL2.57 (0.90); 2022.69 (0.93); 1990.11 (−0.07 to 0.29).24NANA
Proportion menstruating, No./total No.
OverallNA160/327161/334NANA1.00 (0.86 to 1.17).96
Calcium intake<500 mg/d79/168100/196NANA0.96 (0.78 to 1.19).73
≥500 mg/d53/10440/88NANA1.17 (0.80 to 1.72).41
Baseline 25(OH)D concentration, ng/mL<10 ng/mL72/12567/135NANA1.05 (0.86 to 1.29).64
≥10 ng/mL88/20284/199NANA0.90 (0.73 to 1.11).33
Age at menarche, y
OverallNA12.19 (1.14); 16012.20 (1.14); 161−0.04 (−0.29 to 0.21).75NANA
Calcium intake<500 mg/d12.05 (1.18); 7912.22 (1.19); 1000.07 (−0.27 to 0.42).67NANA
≥500 mg/d12.34 (1.02); 5312.15 (1.10); 40−0.38 (−0.82 to 0.06).09NANA
Baseline 25(OH)D concentration, ng/mL<10 ng/mL12.42 (1.16); 7212.31 (1.12); 67−0.10 (−0.49 to 0.29).62NANA
≥10 ng/mL12.01 (1.10); 8812.13 (1.15); 940.02 (−0.30 to 0.34).91NANA

Open in a separate window

Abbreviations: 25(OH)D, 25–hydroxyvitamin D; NA, not applicable.

SI conversion factor: To convert 25(OH)D to nmol/L, multiply by 2.496.

aAdjusted for random effects of school and individual.

Discussion

We present results of what is to our knowledge the first phase 3 randomized clinical trial to investigate whether vitamin D supplementation influences child growth, body composition, or pubertal development. Vitamin D deficiency was highly prevalent in the study population at baseline, and a substantial and sustained increased in 25(OH)D levels was achieved among participants randomized to the intervention arm of the study. However, vitamin D supplementation did not influence any anthropometric or developmental outcome studied, either in the trial population as a whole or in subgroups defined by baseline vitamin D status, sex, calcium intake, or baseline anthropometric status.

To our knowledge, there have been only 2 other randomized clinical trials conducted to evaluate effects of vitamin D on growth and development in school-aged children, both of which we conducted, also in Mongolia. In those studies, where the baseline prevalence of vitamin D deficiency was also very high, we observed a greater increase in stature among children randomized to vitamin D vs placebo.19 This contrasts with the null findings from the current trial. Differing results from the 2 trials may relate to differences in participant age (children aged 6 to 13 years at enrollment in the current trial vs 12 to 15 years in our previous studies) or outcome (height-for-age z scores in the current trial vs change in absolute height previously). Additionally, the duration of vitamin D supplementation and follow-up in our previous trials was only 2 to 6 months, and doses of vitamin D administered were lower at 300 to 800 IU per day; it is therefore possible that the longer period of vitamin D supplementation (3 years), the weekly dosing regimen, or the higher dose of vitamin D administered (14 000 IU per week) in the current trial may have been less effective in boosting growth than the shorter, lower-dose daily intervention previously investigated. Head-to-head studies directly comparing the effects of different vitamin D supplementation regimens on anthropometric outcomes are needed to resolve this discrepancy.

Our trial has several strengths. Its large size afforded ample power to detect small differences in anthropometric outcomes. In contrast with several recent phase 3 trials of vitamin D supplementation,28,29,30 our study population had a high baseline prevalence of vitamin D deficiency; null effects of the intervention cannot therefore be attributed to a paucity of participants with low vitamin D status at baseline. The supplementation regimen we used was highly effective in correcting this among participants randomized to the intervention arm of the trial. Weekly administration of vitamin D3 allowed us to directly observe and optimize adherence during term-time while avoiding large fluctuations in 25(OH)D that may result from larger and more widely spaced bolus doses used in some other studies and potential enzymatic dysregulation that could attenuate vitamin D’s pharmacological activity.31,32 Other important strengths of our study were the high retention rate (91.7% over 3 years) and standardization of serum 25(OH)D measurements.

Limitations

Our trial also has limitations. One such relates to our need to recruit children across different age groups—spanning 6 to 13 years at baseline and 9 to 15 at end-of-trial—to ensure adequate power for studying the primary end point (latent tuberculosis infection). This is relevant because child growth and development trajectories differ by age: increases in height, weight, and accretion of lean mass accelerate considerably during early adolescence,33 as do levels of growth hormone and insulin-like growth factor 1, which have been postulated to interact with vitamin D to stimulate growth.34 A second limitation relates to our use of self-assessed pubertal status as opposed to Tanner staging performed by a health care professional, which involves examination of pubic hair (male and female), genitalia (male), and breasts (female). Major issues with acceptability of such examinations would have seriously compromised recruitment to the study since many children and their parents would likely have concerns about such assessments. Thus, use of self-assessment questionnaires maximized participation and generalizability of our findings. Due to the double-blind design of our study, any imprecision introduced by use of self-assessment is likely to be equally distributed between trial arms and should not therefore have introduced bias. A third limitation relates to the fact that 25(OH)D concentrations were only established at baseline and at 3 years; as a result, seasonal fluctuations in 25(OH)D concentrations during follow-up would not have been captured. However, we highlight that steady state 25(OH)D concentrations among participants randomized to the intervention arm of the trial are likely to have been attained within 2 to 3 months of initiating supplementation. Given the high degree of adherence achieved by directly observing administration of study supplements, we anticipate that the large interarm difference in end-of-study 25(OH)D levels noted at 3-year follow-up is likely to have been sustained throughout follow-up. A fourth limitation relates to exclusion of children who were found to have QFT-positive results at baseline: this design feature potentially reduces the generalizability of our findings, although baseline demographic and anthropometric characteristics were similar for children with QFT positive vs negative results at screening.

Conclusions

In conclusion, this prespecified secondary analysis of a randomized clinical trial found that oral vitamin D3 supplementation at a dose of 14 000 IU per week was effective in elevating 25(OH)D concentrations in a large population of Mongolian school-aged children with low baseline vitamin D status. However, this intervention did not influence growth, body composition, or pubertal development, either in the study population as a whole or within subgroups having the lowest baseline 25(OH)D concentrations or calcium intake values.

Notes

Supplement 1.

Trial protocol

Click here for additional data file.(1.3M, pdf)

Supplement 2.

eTable 1. Questionnaire capturing participants’ intake of calcium-containing foods

eTable 2. Baseline characteristics of participants in pubertal development sub-study, by allocation

eTable 3. Mean waist-to-height ratio z-scores by allocation, overall and by sub-groups

eTable 4. Mean % fat by allocation, overall and by sub-groups

eTable 5. Mean fat mass by allocation, overall and by sub-groups

eTable 6. Mean fat-free mass by allocation, overall and by sub-groups

eTable 7. Proportion of children entering puberty (i.e. reporting Tanner stage 2-5) by allocation and gender, overall and by sub-groups

Click here for additional data file.(336K, pdf)

Supplement 3.

Data sharing statement

Click here for additional data file.(16K, pdf)

References

1. Institute of Medicine, Food and Nutrition Board . Dietary Reference Intakes for Calcium and Vitamin D. National Academy Press; 2010. [Google Scholar]

2. Samuel S, Sitrin MD. Vitamin D’s role in cell proliferation and differentiation. Nutr Rev. 2008;66(10)(suppl 2):S116-S124. doi: 10.1111/j.1753-4887.2008.00094.x [PubMed] [CrossRef] [Google Scholar]

3. Polly P, Tan TC. The role of vitamin D in skeletal and cardiac muscle function. Front Physiol. 2014;5:145. doi: 10.3389/fphys.2014.00145 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

4. Szymczak-Pajor I, Śliwińska A. Analysis of association between vitamin D deficiency and insulin resistance. Nutrients. 2019;11(4):794. doi: 10.3390/nu11040794 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

5. Aranow C. Vitamin D and the immune system. J Investig Med. 2011;59(6):881-886. doi: 10.2310/JIM.0b013e31821b8755 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

6. Prasad P, Kochhar A. Interplay of vitamin D and metabolic syndrome: A review. Diabetes Metab Syndr. 2016;10(2):105-112. doi: 10.1016/j.dsx.2015.02.014 [PubMed] [CrossRef] [Google Scholar]

7. Knuth MM, Mahapatra D, Jima D, et al.. Vitamin D deficiency serves as a precursor to stunted growth and central adiposity in zebrafish. Sci Rep. 2020;10(1):16032. doi: 10.1038/s41598-020-72622-2 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

8. Roth DE, Abrams SA, Aloia J, et al.. Global prevalence and disease burden of vitamin D deficiency: a roadmap for action in low- and middle-income countries. Ann N Y Acad Sci. 2018;1430(1):44-79. doi: 10.1111/nyas.13968 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

9. Eckel RH, Grundy SM, Zimmet PZ. The metabolic syndrome. Lancet. 2005;365(9468):1415-1428. doi: 10.1016/S0140-6736(05)66378-7 [PubMed] [CrossRef] [Google Scholar]

10. Hazell TJ, Gallo S, Vanstone CA, Agellon S, Rodd C, Weiler HA. Vitamin D supplementation trial in infancy: body composition effects at 3 years of age in a prospective follow-up study from Montréal. Pediatr Obes. 2017;12(1):38-47. doi: 10.1111/ijpo.12105 [PubMed] [CrossRef] [Google Scholar]

11. Trilok-Kumar G, Kaur M, Rehman AM, et al.. Effects of vitamin D supplementation in infancy on growth, bone parameters, body composition and gross motor development at age 3-6 years: follow-up of a randomized controlled trial. Int J Epidemiol. 2015;44(3):894-905. doi: 10.1093/ije/dyv116 [PubMed] [CrossRef] [Google Scholar]

12. Mokhtar RR, Holick MF, Sempértegui F, et al.. Vitamin D status is associated with underweight and stunting in children aged 6-36 months residing in the Ecuadorian Andes. Public Health Nutr. 2018;21(11):1974-1985. doi: 10.1017/S1368980017002816 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

13. Foo LH, Zhang Q, Zhu K, et al.. Relationship between vitamin D status, body composition and physical exercise of adolescent girls in Beijing. Osteoporos Int. 2009;20(3):417-425. doi: 10.1007/s00198-008-0667-2 [PubMed] [CrossRef] [Google Scholar]

14. Gilbert-Diamond D, Baylin A, Mora-Plazas M, et al.. Vitamin D deficiency and anthropometric indicators of adiposity in school-age children: a prospective study. Am J Clin Nutr. 2010;92(6):1446-1451. doi: 10.3945/ajcn.2010.29746 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

15. Zakharova I, Klimov L, Kuryaninova V, et al.. Vitamin D insufficiency in overweight and obese children and adolescents. Front Endocrinol (Lausanne). 2019;10:103. doi: 10.3389/fendo.2019.00103 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

16. Plesner JL, Dahl M, Fonvig CE, et al.. Obesity is associated with vitamin D deficiency in Danish children and adolescents. J Pediatr Endocrinol Metab. 2018;31(1):53-61. doi: 10.1515/jpem-2017-0246 [PubMed] [CrossRef] [Google Scholar]

17. Theodoratou E, Tzoulaki I, Zgaga L, Ioannidis JP. Vitamin D and multiple health outcomes: umbrella review of systematic reviews and meta-analyses of observational studies and randomised trials. BMJ. 2014;348:g2035. doi: 10.1136/bmj.g2035 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

18. Lee HS, Kim YJ, Shim YS, Jeong HR, Kwon E, Hwang JS. Associations between serum vitamin D levels and precocious puberty in girls. Ann Pediatr Endocrinol Metab. 2014;19(2):91-95. doi: 10.6065/apem.2014.19.2.91 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

19. Ganmaa D, Stuart JJ, Sumberzul N, et al.. Vitamin D supplementation and growth in urban Mongol school children: Results from two randomized clinical trials. PLoS One. 2017;12(5):e0175237. doi: 10.1371/journal.pone.0175237 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

20. Ganmaa D, Uyanga B, Zhou X, et al.. Vitamin D supplements for prevention of tuberculosis infection and disease. N Engl J Med. 2020;383(4):359-368. doi: 10.1056/NEJMoa1915176 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

21. Bromage S, Daria T, Lander RL, et al.. Diet and nutrition status of Mongolian adults. Nutrients. 2020;12(5):1514. doi: 10.3390/nu12051514 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

22. Chavarro JE, Watkins DJ, Afeiche MC, et al.. Validity of self-assessed sexual maturation against physician assessments and hormone levels. J Pediatr. 2017;186:172-178.e3. doi: 10.1016/j.jpeds.2017.03.050 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

23. Ganmaa D, Khudyakov P, Buyanjargal U, et al.. Prevalence and determinants of QuantiFERON-diagnosed tuberculosis infection in 9810 Mongolian schoolchildren. Clin Infect Dis. 2019;69(5):813-819. doi: 10.1093/cid/ciy975 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

24. Sempos CT, Betz JM, Camara JE, et al.. General steps to standardize the laboratory measurement of serum total 25-hydroxyvitamin D. J AOAC Int. 2017;100(5):1230-1233. doi: 10.5740/jaoacint.17-0259 [PubMed] [CrossRef] [Google Scholar]

25. de Onis M, Onyango AW, Borghi E, Siyam A, Nishida C, Siekmann J. Development of a WHO growth reference for school-aged children and adolescents. Bull World Health Organ. 2007;85(9):660-667. doi: 10.2471/BLT.07.043497 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

26. Sachs MC, Shoben A, Levin GP, et al.. Estimating mean annual 25-hydroxyvitamin D concentrations from single measurements: the Multi-Ethnic Study of Atherosclerosis. Am J Clin Nutr. 2013;97(6):1243-1251. doi: 10.3945/ajcn.112.054502 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

27. Benjamini Y, Hochberg Y. Controlling the false discovery rate—a practical and powerful approach to multiple testing. J Roy Stat Soc B Met.1995;57(1):289-300. doi: 10.1111/j.2517-6161.1995.tb02031.x [CrossRef] [Google Scholar]

28. Scragg R, Waayer D, Stewart AW, et al.. The Vitamin D Assessment (ViDA) Study: design of a randomized controlled trial of vitamin D supplementation for the prevention of cardiovascular disease, acute respiratory infection, falls and non-vertebral fractures. J Steroid Biochem Mol Biol. 2016;164:318-325. doi: 10.1016/j.jsbmb.2015.09.010 [PubMed] [CrossRef] [Google Scholar]

29. Manson JE, Bassuk SS, Lee IM, et al.. The Vitamin D and Omega-3 Trial (VITAL): rationale and design of a large randomized controlled trial of vitamin D and marine omega-3 fatty acid supplements for the primary prevention of cancer and cardiovascular disease. Contemp Clin Trials. 2012;33(1):159-171. doi: 10.1016/j.cct.2011.09.009 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

30. Neale RE, Armstrong BK, Baxter C, et al.. The D-Health Trial: A randomized trial of vitamin D for prevention of mortality and cancer. Contemp Clin Trials. 2016;48:83-90. doi: 10.1016/j.cct.2016.04.005 [PubMed] [CrossRef] [Google Scholar]

31. Martineau AR, Jolliffe DA, Hooper RL, et al.. Vitamin D supplementation to prevent acute respiratory tract infections: systematic review and meta-analysis of individual participant data. BMJ. 2017;356:i6583. doi: 10.1136/bmj.i6583 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

32. Vieth R. How to optimize vitamin D supplementation to prevent cancer, based on cellular adaptation and hydroxylase enzymology. Anticancer Res. 2009;29(9):3675-3684. [PubMed] [Google Scholar]

33. Wells JC, Williams JE, Chomtho S, et al.. Body-composition reference data for simple and reference techniques and a 4-component model: a new UK reference child. Am J Clin Nutr. 2012;96(6):1316-1326. doi: 10.3945/ajcn.112.036970 [PubMed] [CrossRef] [Google Scholar]

34. Esposito S, Leonardi A, Lanciotti L, Cofini M, Muzi G, Penta L. Vitamin D and growth hormone in children: a review of the current scientific knowledge. J Transl Med. 2019;17(1):87. doi: 10.1186/s12967-019-1840-4 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

Articles from JAMA Pediatrics are provided here courtesy of American Medical Association

Influence of Vitamin D Supplementation on Growth, Body Composition, and Pubertal Development Among School-aged Children in an Area With a High Prevalence of Vitamin D Deficiency: A Randomized Clinical Trial (2024)

References

Top Articles
Latest Posts
Article information

Author: Laurine Ryan

Last Updated:

Views: 5478

Rating: 4.7 / 5 (77 voted)

Reviews: 84% of readers found this page helpful

Author information

Name: Laurine Ryan

Birthday: 1994-12-23

Address: Suite 751 871 Lissette Throughway, West Kittie, NH 41603

Phone: +2366831109631

Job: Sales Producer

Hobby: Creative writing, Motor sports, Do it yourself, Skateboarding, Coffee roasting, Calligraphy, Stand-up comedy

Introduction: My name is Laurine Ryan, I am a adorable, fair, graceful, spotless, gorgeous, homely, cooperative person who loves writing and wants to share my knowledge and understanding with you.