1
|
Palacios C, Kostiuk LL, Cuthbert A, Weeks J. Vitamin D supplementation for women during pregnancy. Cochrane Database Syst Rev 2024; 7:CD008873. [PMID: 39077939 PMCID: PMC11287789 DOI: 10.1002/14651858.cd008873.pub5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
BACKGROUND Vitamin D supplementation during pregnancy may help improve maternal and neonatal health outcomes (such as fewer preterm birth and low birthweight babies) and reduce the risk of adverse pregnancy outcomes (such as severe postpartum haemorrhage). OBJECTIVES To examine whether vitamin D supplementation alone or in combination with calcium or other vitamins and minerals given to women during pregnancy can safely improve certain maternal and neonatal outcomes. SEARCH METHODS We searched the Cochrane Pregnancy and Childbirth Trials Register (which includes results of comprehensive searches of CENTRAL, MEDLINE, Embase, CINAHL, ClinicalTrials.gov, the WHO International Clinical Trials Registry Platform, and relevant conference proceedings) (3 December 2022). We also searched the reference lists of retrieved studies. SELECTION CRITERIA Randomised and quasi-randomised trials evaluating the effect of supplementation with vitamin D alone or in combination with other micronutrients for women during pregnancy in comparison to placebo or no intervention. DATA COLLECTION AND ANALYSIS Two review authors independently i) assessed the eligibility of studies against the inclusion criteria, ii) assessed trustworthiness based on pre-defined criteria of scientific integrity, iii) extracted data from included studies, and iv) assessed the risk of bias of the included studies. We assessed the certainty of the evidence using the GRADE approach. MAIN RESULTS The previous version of this review included 30 studies; in this update, we have removed 20 of these studies to 'awaiting classification' following assessments of trustworthiness, one study has been excluded, and one new study included. This current review has a total of 10 included studies, 117 excluded studies, 34 studies in awaiting assessment, and seven ongoing studies. We used the GRADE approach to assess the certainty of the evidence. This removal of the studies resulted in evidence that was downgraded to low-certainty or very low-certainty due to study design limitations, inconsistency between studies, and imprecision. Supplementation with vitamin D compared to no intervention or a placebo A total of eight studies involving 2313 pregnant women were included in this comparison. We assessed four studies as having a low risk of bias for most domains and four studies as having high risk or unclear risk of bias for most domains. The evidence is very uncertain about the effect of supplementation with vitamin D during pregnancy compared to placebo or no intervention on pre-eclampsia (risk ratio (RR) 0.53, 95% confidence interval (CI) 0.21 to 1.33; 1 study, 165 women), gestational diabetes (RR 0.53, 95% CI 0.03 to 8.28; 1 study, 165 women), preterm birth (< 37 weeks) (RR 0.76, 95% CI 0.25 to 2.33; 3 studies, 1368 women), nephritic syndrome (RR 0.17, 95% CI 0.01 to 4.06; 1 study, 135 women), or hypercalcaemia (1 study; no cases reported). Supplementation with vitamin D during pregnancy may reduce the risk of severe postpartum haemorrhage; however, only one study reported this outcome (RR 0.68, 95% CI 0.51 to 0.91; 1 study, 1134 women; low-certainty evidence) and may reduce the risk of low birthweight; however, the upper CI suggests that an increase in risk cannot be ruled out (RR 0.69, 95% CI 0.44 to 1.08; 3 studies, 371 infants; low-certainty evidence). Supplementation with vitamin D + calcium compared to no intervention or a placebo One study involving 84 pregnant women was included in this comparison. Overall, this study was at moderate to high risk of bias. Pre-eclampsia, gestational diabetes, and maternal adverse events were not reported. The evidence is very uncertain about the effect of supplementation with vitamin D and calcium on preterm birth (RR not estimable; very low-certainty evidence) or for low birthweight (RR 1.45, 95% CI 0.14 to 14.94; very low-certainty evidence) compared to women who received placebo or no intervention. Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D) One study involving 1298 pregnant women was included in this comparison. We assessed this study as having a low risk of bias in all domains. Pre-eclampsia was not reported. The evidence is very uncertain about the effect of supplementation with vitamin D, calcium, and other vitamins and minerals during pregnancy compared to no vitamin D on gestational diabetes (RR 0.42, 95% CI 0.10 to 1.73; very low-certainty evidence), maternal adverse events (hypercalcaemia no events and hypercalciuria RR 0.25, 95% CI 0.02 to 3.97; very low-certainty evidence), preterm birth (RR 1.04, 95% CI 0.68 to 1.59; low-certainty evidence), or low birthweight (RR 1.12, 95% CI 0.82 to 1.51; low-certainty evidence). AUTHORS' CONCLUSIONS This updated review using the trustworthy assessment tool removed 21 studies from the previous update and added one new study for a total of 10 included studies. In this setting, supplementation with vitamin D alone compared to no intervention or a placebo resulted in very uncertain evidence on pre-eclampsia, gestational diabetes, preterm birth, or nephritic syndrome. It may reduce the risk of severe postpartum haemorrhage; however, only one study reported this outcome. It may also reduce the risk of low birthweight; however, the upper CI suggests that an increase in risk cannot be ruled out. Supplementation with vitamin D and calcium versus placebo or no intervention resulted in very uncertain evidence on preterm birth and low birthweight. Pre-eclampsia, gestational diabetes, and maternal adverse events were not reported in the only study included in this comparison. Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D) resulted in very uncertain evidence on gestational diabetes and maternal adverse events (hypercalciuria) and uncertain evidence on preterm birth and low birthweight. Pre-eclampsia was not reported in the only study included in this comparison. All findings warrant further research. Additional rigorous, high-quality, and larger randomised trials are required to evaluate the effects of vitamin D supplementation in pregnancy, particularly in relation to the risk of maternal adverse events.
Collapse
Affiliation(s)
- Cristina Palacios
- Department of Dietetics and Nutrition, Florida International University, Miami, Florida, USA
| | - Lia L Kostiuk
- Clinical Safety, Daiichi Sankyo, Basking Ridge, New Jersey, USA
| | - Anna Cuthbert
- Cochrane Pregnancy and Childbirth Group, Department of Women's and Children's Health, University of Liverpool, Liverpool, UK
| | - Jo Weeks
- Cochrane Pregnancy and Childbirth Group, Department of Women's and Children's Health, University of Liverpool, Liverpool, UK
| |
Collapse
|
2
|
Wagner CL, Hollis BW. Vitamin D as a modifier of genomic function and phenotypic expression during pregnancy. FELDMAN AND PIKE' S VITAMIN D 2024:669-692. [DOI: 10.1016/b978-0-323-91386-7.00045-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
3
|
Wright ML, Goin DE, Smed MK, Jewell NP, Nelson JL, Olsen J, Hetland ML, Zoffmann V, Jawaheer D. Pregnancy-associated systemic gene expression compared to a pre-pregnancy baseline, among healthy women with term pregnancies. Front Immunol 2023; 14:1161084. [PMID: 37342349 PMCID: PMC10277629 DOI: 10.3389/fimmu.2023.1161084] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/09/2023] [Indexed: 06/22/2023] Open
Abstract
Background Pregnancy is known to induce extensive biological changes in the healthy mother. Little is known, however, about what these changes are at the molecular level. We have examined systemic expression changes in protein-coding genes and long non-coding (lnc) RNAs during and after pregnancy, compared to before pregnancy, among healthy women with term pregnancies. Methods Blood samples were collected from 14 healthy women enrolled in our prospective pregnancy cohort at 7 time-points (before, during and after pregnancy). Total RNA from frozen whole blood was used for RNA sequencing. Following raw read alignment and assembly, gene-level counts were obtained for protein-coding genes and long non-coding RNAs. At each time-point, cell type proportions were estimated using deconvolution. To examine associations between pregnancy status and gene expression over time, Generalized Estimating Equation (GEE) models were fitted, adjusting for age at conception, and with and without adjusting for changes in cell type proportions. Fold-changes in expression at each trimester were examined relative to the pre-pregnancy baseline. Results Numerous immune-related genes demonstrated pregnancy-associated expression, in a time-dependent manner. The genes that demonstrated the largest changes in expression included several that were neutrophil-related (over-expressed) and numerous immunoglobulin genes (under-expressed). Estimated cell proportions revealed a marked increase in neutrophils, and less so of activated CD4 memory T cells, during pregnancy, while most other cell type proportions decreased or remained unchanged. Adjusting for cell type proportions in our model revealed that although most of the expression changes were due to changes in cell type proportions in the bloodstream, transcriptional regulation was also involved, especially in down-regulating expression of type I interferon inducible genes. Conclusion Compared to a pre-pregnancy baseline, there were extensive systemic changes in cell type proportions, gene expression and biological pathways associated with different stages of pregnancy and postpartum among healthy women. Some were due to changes in cell type proportions and some due to gene regulation. In addition to providing insight into term pregnancy among healthy women, these findings also provide a "normal" reference for abnormal pregnancies and for autoimmune diseases that improve or worsen during pregnancy, to assess deviations from normal.
Collapse
Affiliation(s)
- Matthew L. Wright
- Children’s Hospital Oakland Research Institute, Oakland, CA, United States
- Northwestern University, Feinberg School of Medicine, Chicago, IL, United States
| | - Dana E. Goin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA, United States
| | | | - Nicholas P. Jewell
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - J. Lee Nelson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Jørn Olsen
- Department of Epidemiology, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Merete Lund Hetland
- DANBIO Registry and Copenhagen Centre for Arthritis Research, Centre for Rheumatology and Spine Diseases, Rigshospitalet, Glostrup, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vibeke Zoffmann
- Juliane Marie Centeret, Rigshospitalet, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Damini Jawaheer
- Children’s Hospital Oakland Research Institute, Oakland, CA, United States
- Northwestern University, Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
4
|
Genomic or Non-Genomic? A Question about the Pleiotropic Roles of Vitamin D in Inflammatory-Based Diseases. Nutrients 2023; 15:nu15030767. [PMID: 36771473 PMCID: PMC9920355 DOI: 10.3390/nu15030767] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/10/2023] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Vitamin D (vit D) is widely known for its role in calcium metabolism and its importance for the bone system. However, various studies have revealed a myriad of extra-skeletal functions, including cell differentiation and proliferation, antibacterial, antioxidant, immunomodulatory, and anti-inflammatory properties in various cells and tissues. Vit D mediates its function via regulation of gene expression by binding to its receptor (VDR) which is expressed in almost all cells within the body. This review summarizes the pleiotropic effects of vit D, emphasizing its anti-inflammatory effect on different organ systems. It also provides a comprehensive overview of the genetic and epigenetic effects of vit D and VDR on the expression of genes pertaining to immunity and anti-inflammation. We speculate that in the context of inflammation, vit D and its receptor VDR might fulfill their roles as gene regulators through not only direct gene regulation but also through epigenetic mechanisms.
Collapse
|
5
|
Vitamin D supplementation and immune-related markers: an update from nutrigenetic and nutrigenomic studies. Br J Nutr 2022; 128. [PMCID: PMC9557210 DOI: 10.1017/s0007114522002392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Vitamin D is both a nutrient and a neurologic hormone that plays a critical role in modulating immune responses. While low levels of vitamin D are associated with increased susceptibility to infections and immune-related disorders, vitamin D supplementation has demonstrated immunomodulatory effects that can be protective against various diseases and infections. Vitamin D receptor is expressed in immune cells that have the ability to synthesise the active vitamin D metabolite. Thus, vitamin D acts in an autocrine manner in a local immunologic milieu in fighting against infections. Nutrigenetics and nutrigenomics are the new disciplines of nutritional science that explore the interaction between nutrients and genes using distinct approaches to decipher the mechanisms by which nutrients can influence disease development. Though molecular and observational studies have proved the immunomodulatory effects of vitamin D, only very few studies have documented the molecular insights of vitamin D supplementation. Until recently, researchers have investigated only a few selected genes involved in the vitamin D metabolic pathway that may influence the response to vitamin D supplementation and possibly disease risk. This review summarises the impact of vitamin D supplementation on immune markers from nutrigenetics and nutrigenomics perspective based on evidence collected through a structured search using PubMed, EMBASE, Science Direct and Web of Science. The research gaps and shortcomings from the existing data and future research direction of vitamin D supplementation on various immune-related disorders are discussed.
Collapse
|
6
|
Sufriyana H, Salim HM, Muhammad AR, Wu YW, Su ECY. Blood biomarkers representing maternal-fetal interface tissues used to predict early-and late-onset preeclampsia but not COVID-19 infection. Comput Struct Biotechnol J 2022; 20:4206-4224. [PMID: 35966044 PMCID: PMC9359600 DOI: 10.1016/j.csbj.2022.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/04/2022] [Accepted: 08/04/2022] [Indexed: 11/06/2022] Open
Abstract
Endothelial dysfunction misleads blood marker discovery by differential expression. Blood-derived surrogate transcriptome of target-tissue avoids the false discovery. ITGA5 implies polymicrobial infection of maternal-fetal interface in preeclampsia. ITGA5 and IRF6 implies viral co-infection in early-onset preeclampsia. ITGA5, IRF6, and P2RX7 differ imminent preeclampsia from COVID-19 infection.
Background A well-known blood biomarker (soluble fms-like tyrosinase-1 [sFLT-1]) for preeclampsia, i.e., a pregnancy disorder, was found to predict severe COVID-19, including in males. True biomarker may be masked by more-abrupt changes related to endothelial instead of placental dysfunction. This study aimed to identify blood biomarkers that represent maternal-fetal interface tissues for predicting preeclampsia but not COVID-19 infection. Methods The surrogate transcriptome of tissues was determined by that in maternal blood, utilizing four datasets (n = 1354) which were collected before the COVID-19 pandemic. Applying machine learning, a preeclampsia prediction model was chosen between those using blood transcriptome (differentially expressed genes [DEGs]) and the blood-derived surrogate for tissues. We selected the best predictive model by the area under the receiver operating characteristic (AUROC) using a dataset for developing the model, and well-replicated in datasets both with and without an intervention. To identify eligible blood biomarkers that predicted any-onset preeclampsia from the datasets but that were not positive in the COVID-19 dataset (n = 47), we compared several methods of predictor discovery: (1) the best prediction model; (2) gene sets of standard pipelines; and (3) a validated gene set for predicting any-onset preeclampsia during the pandemic (n = 404). We chose the most predictive biomarkers from the best method with the significantly largest number of discoveries by a permutation test. The biological relevance was justified by exploring and reanalyzing low- and high-level, multiomics information. Results A prediction model using the surrogates developed for predicting any-onset preeclampsia (AUROC of 0.85, 95 % confidence interval [CI] 0.77 to 0.93) was the only that was well-replicated in an independent dataset with no intervention. No model was well-replicated in datasets with a vitamin D intervention. None of the blood biomarkers with high weights in the best model overlapped with blood DEGs. Blood biomarkers were transcripts of integrin-α5 (ITGA5), interferon regulatory factor-6 (IRF6), and P2X purinoreceptor-7 (P2RX7) from the prediction model, which was the only method that significantly discovered eligible blood biomarkers (n = 3/100 combinations, 3.0 %; P =.036). Most of the predicted events (73.70 %) among any-onset preeclampsia were cluster A as defined by ITGA5 (Z-score ≥ 1.1), but were only a minority (6.34 %) among positives in the COVID-19 dataset. The remaining were predicted events (26.30 %) among any-onset preeclampsia or those among COVID-19 infection (93.66 %) if IRF6 Z-score was ≥-0.73 (clusters B and C), in which none was the predicted events among either late-onset preeclampsia (LOPE) or COVID-19 infection if P2RX7 Z-score was <0.13 (cluster C). Greater proportions of predicted events among LOPE were cluster A (82.85 % vs 70.53 %) compared to early-onset preeclampsia (EOPE). The biological relevance by multiomics information explained the biomarker mechanism, polymicrobial infection in any-onset preeclampsia by ITGA5, viral co-infection in EOPE by ITGA5-IRF6, a shared prediction with COVID-19 infection by ITGA5-IRF6-P2RX7, and non-replicability in datasets with a vitamin D intervention by ITGA5. Conclusions In a model that predicts preeclampsia but not COVID-19 infection, the important predictors were genes in maternal blood that were not extremely expressed, including the proposed blood biomarkers. The predictive performance and biological relevance should be validated in future experiments.
Collapse
Affiliation(s)
- Herdiantri Sufriyana
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Xing Street, Taipei 11031, Taiwan.,Department of Medical Physiology, Faculty of Medicine, Universitas Nahdlatul Ulama Surabaya, 57 Raya Jemursari Road, Surabaya 60237, Indonesia
| | - Hotimah Masdan Salim
- Department of Molecular Biology, Faculty of Medicine, Universitas Nahdlatul Ulama Surabaya, 57 Raya Jemursari Road, Surabaya 60237, Indonesia
| | - Akbar Reza Muhammad
- Faculty of Medicine, Universitas Nahdlatul Ulama Surabaya, 57 Raya Jemursari Road, Surabaya 60237, Indonesia
| | - Yu-Wei Wu
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Xing Street, Taipei 11031, Taiwan.,Clinical Big Data Research Center, Taipei Medical University Hospital, 250 Wu-Xing Street, Taipei 11031, Taiwan
| | - Emily Chia-Yu Su
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Xing Street, Taipei 11031, Taiwan.,Clinical Big Data Research Center, Taipei Medical University Hospital, 250 Wu-Xing Street, Taipei 11031, Taiwan.,Research Center for Artificial Intelligence in Medicine, Taipei Medical University, 250 Wu-Xing Street, Taipei 11031, Taiwan
| |
Collapse
|
7
|
Rees G, Brough L, Orsatti GM, Lodge A, Walker S. Do Micronutrient and Omega-3 Fatty Acid Supplements Affect Human Maternal Immunity during Pregnancy? A Scoping Review. Nutrients 2022; 14:367. [PMID: 35057548 PMCID: PMC8781537 DOI: 10.3390/nu14020367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/04/2022] [Accepted: 01/10/2022] [Indexed: 02/04/2023] Open
Abstract
Maternal dietary micronutrients and omega-3 fatty acids support development of the fetal and neonatal immune system. Whether supplementation is similarly beneficial for the mother during gestation has received limited attention. A scoping review of human trials was conducted looking for evidence of biochemical, genomic, and clinical effects of supplementation on the maternal immune system. The authors explored the literature on PubMed, Cochrane Library, and Web of Science databases from 2010 to the present day using PRISMA-ScR methodology. Full-length human trials in English were searched for using general terms and vitamin A, B12, C, D, and E; choline; iodine; iron; selenium; zinc; and docosahexaenoic/eicosapentaenoic acid. Of 1391 unique articles, 36 were eligible for inclusion. Diverse biochemical and epigenomic effects of supplementation were identified that may influence innate and adaptive immunity. Possible clinical benefits were encountered in malaria, HIV infections, anemia, Type 1 diabetes mellitus, and preventing preterm delivery. Only limited publications were identified that directly explored maternal immunity in pregnancy and the effects of micronutrients. None provided a holistic perspective. It is concluded that supplementation may influence biochemical aspects of the maternal immune response and some clinical outcomes, but the evidence from this review is not sufficient to justify changes to current guidelines.
Collapse
Affiliation(s)
- Gail Rees
- School of Biomedical Sciences, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - Louise Brough
- School of Food and Advanced Technology, Massey University, Palmerston North 4442, New Zealand
| | | | - Anna Lodge
- St Gilesmedical, London WC2H 8LG, UK; (A.L.); (S.W.)
| | - Steven Walker
- St Gilesmedical, London WC2H 8LG, UK; (A.L.); (S.W.)
| |
Collapse
|
8
|
Raia-Barjat T, Sarkis C, Rancon F, Thibaudin L, Gris JC, Alfaidy N, Chauleur C. Vitamin D deficiency during late pregnancy mediates placenta-associated complications. Sci Rep 2021; 11:20708. [PMID: 34671075 PMCID: PMC8528848 DOI: 10.1038/s41598-021-00250-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
During pregnancy, maternal vitamin D insufficiency could increase the risk of preeclampsia. Aim of the study was to evaluate the relationship between vitamin D status and the occurrence of placenta-mediated complications (PMCs) in a population at high risk. A prospective multicenter cohort study of 200 pregnant patients was conducted. The vitamin D level of patients with placenta-mediated complications was lower at 32 weeks compared to uncomplicated pregnancies (P = 0.001). At 32 weeks, the risk of occurrence of PMCs was five times higher in patients with vitamin D deficiency (RR: 5.14 95% CI (1.50-17.55)) compared to patients with normal vitamin D levels. There was a strong, inverse relationship between serum 25(OH)D levels at 32 weeks and the subsequent risk of PMCs (P = 0.001). At 32 weeks, the vitamin D level of patients with late-onset PMCs was lower than the one of patients with early-onset PMCs and of patients without PMCs (P < 0.0001). These results suggest a role of vitamin D in the maintenance of placental performance and therefore in the prevention of the onset of late PMC.
Collapse
Affiliation(s)
- Tiphaine Raia-Barjat
- Department of Gynaecology and Obstetrics, Hôpital Nord, University Hospital, Avenue Albert Raimond, 42270, Saint Etienne, France. .,Jean Monet Saint-Etienne University, INSERM, SAINBIOSE (SAnte, INgénierie, BIOlogie, Saint- Etienne), U1059, Saint-Etienne, France. .,Institut National de La Santé Et de La Recherche Médicale, U1292, Biosanté, Grenoble, France. .,University Grenoble-Alpes, 38000, Grenoble, France.
| | - Camille Sarkis
- Department of Gynaecology and Obstetrics, Hôpital Nord, University Hospital, Avenue Albert Raimond, 42270, Saint Etienne, France
| | - Florence Rancon
- Jean Monet Saint-Etienne University, INSERM, SAINBIOSE (SAnte, INgénierie, BIOlogie, Saint- Etienne), U1059, Saint-Etienne, France.,INSERM, Centre d'Investigation Clinique 1408, Saint-Etienne, France
| | - Lise Thibaudin
- Biochemistry Laboratory, University Hospital, Saint-Etienne, France
| | - Jean-Christophe Gris
- Laboratory of Hematology, University Hospital, Nimes, France.,University of Montpellier, Montpellier, France.,I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation.,Institut Desbrest d'Epidémiologie et de Santé Publique UMR INSERM - Université de Montpellier, Montpellier, France
| | - Nadia Alfaidy
- Institut National de La Santé Et de La Recherche Médicale, U1292, Biosanté, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, Grenoble, France.,Centre Hospitalo-Universitaire Grenoble Alpes, Service Obstétrique, CS 10217, 38043, Grenoble Cedex 9, France.,Université Grenoble Alpes, Grenoble, France
| | - Céline Chauleur
- Department of Gynaecology and Obstetrics, Hôpital Nord, University Hospital, Avenue Albert Raimond, 42270, Saint Etienne, France.,Jean Monet Saint-Etienne University, INSERM, SAINBIOSE (SAnte, INgénierie, BIOlogie, Saint- Etienne), U1059, Saint-Etienne, France
| |
Collapse
|
9
|
Alabduljabbar S, Zaidan SA, Lakshmanan AP, Terranegra A. Personalized Nutrition Approach in Pregnancy and Early Life to Tackle Childhood and Adult Non-Communicable Diseases. Life (Basel) 2021; 11:life11060467. [PMID: 34073649 PMCID: PMC8224671 DOI: 10.3390/life11060467] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/05/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023] Open
Abstract
The development of childhood and adult non-communicable diseases (NCD) is associated with environmental factors, starting from intrauterine life. A new theory finds the roots of epigenetic programming in parental gametogenesis, continuing during embryo development, fetal life, and finally in post-natal life. Maternal health status and poor nutrition are widely recognized as implications in the onset of childhood and adult diseases. Early nutrition, particularly breastfeeding, also plays a primary role in affecting the health status of an individual later in life. A poor maternal diet during pregnancy and lack of breastfeeding can cause a nutrient deficiency that affects the gut microbiota, and acts as a cofactor for many pathways, impacting the epigenetic controls and transcription of genes involved in the metabolism, angiogenesis, and other pathways, leading to NCDs in adult life. Both maternal and fetal genetic backgrounds also affect nutrient adsorption and functioning at the cellular level. This review discusses the most recent evidence on maternal nutrition and breastfeeding in the development of NCD, the potentiality of the omics technologies in uncovering the molecular mechanisms underlying it, with the future prospective of applying a personalized nutrition approach to prevent and treat NCD from the beginning of fetal life.
Collapse
|
10
|
Wagner CL, Hollis BW. Early-Life Effects of Vitamin D: A Focus on Pregnancy and Lactation. ANNALS OF NUTRITION AND METABOLISM 2020; 76 Suppl 2:16-28. [PMID: 33232956 DOI: 10.1159/000508422] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 03/15/2020] [Indexed: 11/19/2022]
Abstract
Vitamin D is an endocrine regulator of calcium and bone metabolism. Yet, its effects include other systems, such as innate and adaptive immunity. Unique to pregnancy, circulating 1,25-dihydroxyvitamin D (1,25[OH]2D) increases early on to concentrations that are 2-3 times prepregnant values. At no other time during the lifecycle is the conversion of 25-hydroxyvitamin D (25[OH]D) to 1,25(OH)2D directly related and optimized at ≥100 nmol/L. Vitamin D deficiency appears to affect pregnancy outcomes, yet randomized controlled trials of vitamin D supplementation achieve mixed results depending on when supplementation is initiated during pregnancy, the dose and dosing interval, and the degree of deficiency at the onset of pregnancy. Analysis of trials on an intention-to-treat basis as opposed to the use of 25(OH)D as the intermediary biomarker of vitamin D metabolism yields differing results, with treatment effects often noted only in the most deficient women. Immediately after delivery, maternal circulating 1,25(OH)2D concentrations return to prepregnancy baseline, at a time when a breastfeeding woman has increased demands of calcium, beyond what was needed during the last trimester of pregnancy, making one question why 1,25(OH)2D increases so significantly during pregnancy. Is it to serve as an immune modulator? The vitamin D content of mother's milk is directly related to maternal vitamin D status, and if a woman was deficient during pregnancy, her milk will be deficient unless she is taking higher doses of vitamin D. Because of this relative "deficiency," there is a recommendation that all breastfed infants receive 400 IU vitamin D3/day starting a few days after birth. The alternative - maternal supplementation with 6,400 IU vitamin D3/day, effective in safely raising maternal circulating vitamin D, that of her breast milk, and effective in achieving sufficiency in her recipient breastfeeding infant - remains a viable option. Additional research is needed to understand vitamin D's influence on pregnancy health and the effect of maternal supplementation on breast milk's immune signaling.
Collapse
Affiliation(s)
- Carol L Wagner
- Division of Neonatology, Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA,
| | - Bruce W Hollis
- Division of Neonatology, Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
11
|
Kiely ME, Wagner CL, Roth DE. Vitamin D in pregnancy: Where we are and where we should go. J Steroid Biochem Mol Biol 2020; 201:105669. [PMID: 32302652 DOI: 10.1016/j.jsbmb.2020.105669] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/18/2020] [Accepted: 04/02/2020] [Indexed: 01/06/2023]
Abstract
Vitamin D deficiency has been widely reported among pregnant women and infants around the world. Women with low sun exposure, high BMI, low vitamin D intakes and socioeconomic disadvantage with poor quality diets are at greatest risk of vitamin D deficiency, leading to very low serum concentrations of 25-hydroxyvitamin D (25(OH)D) in their offspring and an increased risk of nutritional rickets. Many observational studies, supported by compelling in vitro and in vivo data, have generated evidence suggesting that low vitamin D status in pregnancy may also contribute to the risk of adverse perinatal outcomes including hypertensive disorders (e.g., preeclampsia), fetal growth restriction, and preterm birth. However, the few large randomized controlled trials (RCTs) conducted to date have generated conflicting evidence for a role of vitamin D supplementation in improving perinatal outcomes. Vitamin D supplementation policies during pregnancy and implementation of policies vary within and between jurisdictions. Regulatory authorities have cited insufficient evidence to establish pregnancy-specific targets for serum 25(OH)D concentrations or prenatal vitamin D intake that effectively reduce the risks of adverse perinatal and infant outcomes. This paper arises from a Debate on Vitamin D Requirements during Pregnancy, held at the 22nd Vitamin D Workshop, 2019. From varied perspectives, our objectives were to evaluate the evidence for: vitamin D metabolism in pregnancy and the prevalence of gestational vitamin D deficiency worldwide; the translation of laboratory research findings to clinical studies on the role of vitamin D in perinatal health; the challenges of designing and conducting clinical trials to establish prenatal vitamin D requirements; and results to date of major large RCTs of prenatal vitamin D supplementation. Lastly, we explored potential next steps towards generating robust clinical data in this field to address both public health protection and patient care.
Collapse
Affiliation(s)
- M E Kiely
- Cork Centre for Vitamin D and Nutrition Research, School of Food and Nutritional Sciences, University College Cork, Ireland; INFANT Research Centre, University College Cork, Ireland.
| | - C L Wagner
- Division of Neonatology, Medical University of South Carolina, Charleston, SC 29425, United States
| | - D E Roth
- Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
12
|
Gomez-Lopez N, Romero R, Hassan SS, Bhatti G, Berry SM, Kusanovic JP, Pacora P, Tarca AL. The Cellular Transcriptome in the Maternal Circulation During Normal Pregnancy: A Longitudinal Study. Front Immunol 2019; 10:2863. [PMID: 31921132 PMCID: PMC6928201 DOI: 10.3389/fimmu.2019.02863] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/21/2019] [Indexed: 12/16/2022] Open
Abstract
Pregnancy represents a unique immunological state in which the mother adapts to tolerate the semi-allogenic conceptus; yet, the cellular dynamics in the maternal circulation are poorly understood. Using exon-level expression profiling of up to six longitudinal whole blood samples from 49 pregnant women, we undertook a systems biology analysis of the cellular transcriptome dynamics and its correlation with the plasma proteome. We found that: (1) chromosome 14 was the most enriched in transcripts differentially expressed throughout normal pregnancy; (2) the strongest expression changes followed three distinct longitudinal patterns, with genes related to host immune response (e.g., MMP8, DEFA1B, DEFA4, and LTF) showing a steady increase in expression from 10 to 40 weeks of gestation; (3) multiple biological processes and pathways related to immunity and inflammation were modulated during gestation; (4) genes changing with gestation were among those specific to T cells, B cells, CD71+ erythroid cells, natural killer cells, and endothelial cells, as defined based on the GNF Gene Expression Atlas; (5) the average expression of mRNA signatures of T cells, B cells, and erythroid cells followed unique patterns during gestation; (6) the correlation between mRNA and protein abundance was higher for mRNAs that were differentially expressed throughout gestation than for those that were not, and significant mRNA-protein correlations were observed for genes part of the T-cell signature. In summary, unique changes in immune-related genes were discovered by longitudinally assessing the cellular transcriptome in the maternal circulation throughout normal pregnancy, and positive correlations were noted between the cellular transcriptome and plasma proteome for specific genes/proteins. These findings provide insights into the immunobiology of normal pregnancy.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD and Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD and Detroit, MI, United States
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, United States
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, United States
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
- Detroit Medical Center, Detroit, MI, United States
- Department of Obstetrics & Gynecology, Florida International University, Miami, FL, United States
| | - Sonia S. Hassan
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD and Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Stanley M. Berry
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD and Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Juan Pedro Kusanovic
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD and Detroit, MI, United States
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Center for Research and Innovation in Maternal-Fetal Medicine (CIMAF), Department of Obstetrics and Gynecology, Sótero del Río Hospital, Santiago, Chile
| | - Percy Pacora
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD and Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Adi L. Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD and Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, United States
| |
Collapse
|
13
|
Pique-Regi R, Romero R, Tarca AL, Sendler ED, Xu Y, Garcia-Flores V, Leng Y, Luca F, Hassan SS, Gomez-Lopez N. Single cell transcriptional signatures of the human placenta in term and preterm parturition. eLife 2019; 8:52004. [PMID: 31829938 PMCID: PMC6949028 DOI: 10.7554/elife.52004] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/12/2019] [Indexed: 01/02/2023] Open
Abstract
More than 135 million births occur each year; yet, the molecular underpinnings of human parturition in gestational tissues, and in particular the placenta, are still poorly understood. The placenta is a complex heterogeneous organ including cells of both maternal and fetal origin, and insults that disrupt the maternal-fetal dialogue could result in adverse pregnancy outcomes such as preterm birth. There is limited knowledge of the cell type composition and transcriptional activity of the placenta and its compartments during physiologic and pathologic parturition. To fill this knowledge gap, we used scRNA-seq to profile the placental villous tree, basal plate, and chorioamniotic membranes of women with or without labor at term and those with preterm labor. Significant differences in cell type composition and transcriptional profiles were found among placental compartments and across study groups. For the first time, two cell types were identified: 1) lymphatic endothelial decidual cells in the chorioamniotic membranes, and 2) non-proliferative interstitial cytotrophoblasts in the placental villi. Maternal macrophages from the chorioamniotic membranes displayed the largest differences in gene expression (e.g. NFKB1) in both processes of labor; yet, specific gene expression changes were also detected in preterm labor. Importantly, several placental scRNA-seq transcriptional signatures were modulated with advancing gestation in the maternal circulation, and specific immune cell type signatures were increased with labor at term (NK-cell and activated T-cell signatures) and with preterm labor (macrophage, monocyte, and activated T-cell signatures). Herein, we provide a catalogue of cell types and transcriptional profiles in the human placenta, shedding light on the molecular underpinnings and non-invasive prediction of the physiologic and pathologic parturition.
Collapse
Affiliation(s)
- Roger Pique-Regi
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, United States.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, United States
| | - Roberto Romero
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, United States.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, United States.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, United States.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, United States.,Detroit Medical Center, Detroit, United States
| | - Adi L Tarca
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, United States.,Department of Computer Science, College of Engineering, Wayne State University, Detroit, United States
| | - Edward D Sendler
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, United States
| | - Yi Xu
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, United States
| | - Valeria Garcia-Flores
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, United States
| | - Yaozhu Leng
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, United States
| | - Francesca Luca
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, United States.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States
| | - Sonia S Hassan
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States.,Department of Physiology, Wayne State University School of Medicine, Detroit, United States
| | - Nardhy Gomez-Lopez
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, United States.,Department of Immunology, Microbiology, and Biochemistry, Wayne State University School of Medicine, Detroit, United States
| |
Collapse
|
14
|
Palacios C, Trak‐Fellermeier MA, Martinez RX, Lopez‐Perez L, Lips P, Salisi JA, John JC, Peña‐Rosas JP. Regimens of vitamin D supplementation for women during pregnancy. Cochrane Database Syst Rev 2019; 10:CD013446. [PMID: 31581312 PMCID: PMC6776191 DOI: 10.1002/14651858.cd013446] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Vitamin D deficiency during pregnancy increases the risk of pre-eclampsia, gestational diabetes, preterm birth, and low birthweight. In a previous Cochrane Review we found that supplementing pregnant women with vitamin D alone compared to no vitamin D supplementation may reduce the risk of pre-eclampsia, gestational diabetes, and low birthweight and may increase the risk of preterm births if it is combined with calcium. However the effects of different vitamin D regimens are not yet clear. OBJECTIVES To assess the effects and safety of different regimens of vitamin D supplementation alone or in combination with calcium or other vitamins, minerals or nutrients during pregnancy, specifically doses of 601 international units per day (IU/d) or more versus 600 IU/d or less; and 4000 IU/d or more versus 3999 IU/d or less. SEARCH METHODS We searched the Cochrane Pregnancy and Childbirth's Trials Register, ClinicalTrials.gov, the WHO International Clinical Trials Registry Platform (ICTRP) (12 July 2018), and the reference lists of retrieved studies. SELECTION CRITERIA Randomised trials evaluating the effect of different vitamin D regimens (dose, frequency, duration, and time of commencement of supplementation during pregnancy), alone or in combination with other nutrients on pregnancy and neonatal health outcomes. We only included trials that compared 601 IU/d or more versus 600 IU/d or less and 4000 IU/d or more versus 3999 IU/d or less. We did not include in the analysis groups that received no vitamin D, as that comparison is assessed in another Cochrane Review. DATA COLLECTION AND ANALYSIS Two review authors independently: i) assessed the eligibility of studies against the inclusion criteria; ii) extracted data from included studies, and iii) assessed the risk of bias of the included studies. Our primary maternal outcomes were: pre-eclampsia, gestational diabetes, and any adverse effects; our primary infant outcomes were preterm birth and low birthweight. Data were checked for accuracy. The certainty of the evidence was assessed using the GRADE approach. MAIN RESULTS In this review, we included data from 30 trials involving 7289 women. We excluded 11 trials, identified 16 ongoing/unpublished trials and two trials are awaiting classification. Overall risk of bias for the trials was mixed.Comparison 1. 601 IU/d or more versus 600 IU/d or less of vitamin D alone or with any other nutrient (19 trials; 5214 participants)Supplementation with 601 IU/d or more of vitamin D during pregnancy may make little or no difference to the risk of pre-eclampsia (risk ratio (RR) 0.96, 95% confidence interval (CI) 0.65 to 1.42); 5 trials; 1553 participants,low-certainty evidence), may reduce the risk of gestational diabetes (RR 0.54, 95% CI 0.34 to 0.86; 5 trials; 1846 participants; moderate-certainty evidence), may make little or no difference to the risk of preterm birth (RR 1.25, 95% CI 0.92 to 1.69; 4 trials; 2294 participants; low-certainty evidence); and may make little or no difference to the risk of low birthweight (RR 0.90, 95% CI 0.66 to 1.24; 4 trials; 1550 participants; very low-certainty evidence) compared to women receiving 600 IU/d or less.Comparison 2. 4000 IU or more versus 3999 IU or less of vitamin D alone (15 trials; 4763 participants)Supplementation with 4000 IU/d or more of vitamin D during pregnancy may make little or no difference to the risk of: pre-eclampsia (RR 0.87, 95% CI 0.62 to 1.22; 4 trials, 1903 participants, low-certainty evidence); gestational diabetes (RR 0.89, 95% CI 0.56 to 1.42; 5 trials, 2276 participants; low-certainty evidence); preterm birth (RR 0.85, 95% CI 0.64 to 1.12; 6 trials, 2948 participants, low-certainty evidence); and low birthweight (RR 0.92, 95% CI 0.49 to 1.70; 2 trials; 1099 participants; low-certainty evidence) compared to women receiving 3999 IU/d or less.Adverse events (such as hypercalcaemia, hypocalcaemia, hypercalciuria, and hypovitaminosis D) were reported differently in most trials; however, in general, there was little to no side effects reported or similar cases between groups. AUTHORS' CONCLUSIONS Supplementing pregnant women with more than the current vitamin D recommendation may reduce the risk of gestational diabetes; however, it may make little or no difference to the risk of pre-eclampsia, preterm birth and low birthweight. Supplementing pregnant women with more than the current upper limit for vitamin D seems not to increase the risk of the outcomes evaluated. In general, the GRADE was considered low certainty for most of the primary outcomes due to serious risk of bias and imprecision of results. With respect to safety, it appears that vitamin D supplementation is a safe intervention during pregnancy, although the parameters used to determine this were either not reported or not consistent between trials. Future trials should be consistent in their reports of adverse events. There are 16 ongoing trials that when published, will increase the body of knowledge.
Collapse
Affiliation(s)
- Cristina Palacios
- Robert Stempel College of Public Health and Social Work, Florida International UniversityDepartment of Dietetics and Nutrition11200 SW 8th Street, AHC 5 – 323MiamiFloridaUSA33199
| | - Maria Angelica Trak‐Fellermeier
- University of Puerto RicoCenter for Clinical Research and Health PromotionMedical Science Campus Suite A107, PO Box 365067San JuanPuerto RicoPR00935
| | | | - Lucero Lopez‐Perez
- WHO ConsultantIsla del Socorro 7Col. Prado VallejoTlalnepantlaMexico54170
| | - Paul Lips
- VU University Medical CenterInternal Medicine, Endocrine SectionP.O. Box 7057AmsterdamNetherlands1071 MC
| | - James A Salisi
- WHO Regional Office of the Western PacificNutrition Unit, Division of Non‐communicable Diseases and Health through the Life‐courseUN AvenueManilaPhilippines1000
| | - Jessica C John
- Eat, Drink and Be Healthy173 Eastern Main RoadTunapunaTrinidad and Tobago
| | - Juan Pablo Peña‐Rosas
- World Health OrganizationEvidence and Programme Guidance, Department of Nutrition for Health and Development20 Avenue AppiaGenevaGESwitzerland1211
| | | |
Collapse
|
15
|
Hajizadeh S, Rankin Shary J, Gayle Reed S, Lynn Wagner C. The prevalence of hypovitaminosis D and its risk factors in pregnant women and their newborns in the Middle East: A systematic review. Int J Reprod Biomed 2019; 17:685-708. [PMID: 31807718 PMCID: PMC6844283 DOI: 10.18502/ijrm.v17i10.5284] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 10/31/2018] [Accepted: 12/15/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Pregnant women and newborns are at risk for vitamin D deficiency (VDD). Also, poor health outcomes for pregnant women with VDD are reported in the published literature. OBJECTIVE The aim of this systematic review was to estimate the prevalence of hypovitaminosis D and the associated risk factors for hypovitaminosis D in Middle Eastern pregnant women and their newborns. RESULTS The prevalence of circulating 25-hydroxyvitamin D (25(OH)D) < 50 nmol/L as a marker of vitamin D status in pregnant women and their newborns was between 24.5-98% and 22-100%, respectively. The prevalence of 25(OH) D < 25 nmol/L in pregnant women and their newborns was over a wide range between 16.7-80% and 22-82%, respectively. Predictors for low maternal and neonatal 25(OH)D concentrations included decreased vitamin D synthesis due to reduced exposure to sunlight and decreased nutritional intake of vitamin D. A predictor of low neonatal 25(OH)D concentrations included maternal vitamin D status and the correlation between vitamin D concentrations in maternal and cord blood. CONCLUSION The high prevalence of VDD in the pregnant women of the Middle East underscores the necessity of implementing national prevention and intervention strategies. A clear policy for clinicians and healthcare workers is needed for screening and maintaining sufficient vitamin D status during pregnancy.
Collapse
Affiliation(s)
- Shayesteh Hajizadeh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Judy Rankin Shary
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Susan Gayle Reed
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Carol Lynn Wagner
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
16
|
Abstract
BACKGROUND Vitamin D supplementation during pregnancy may be needed to protect against adverse pregnancy outcomes. This is an update of a review that was first published in 2012 and then in 2016. OBJECTIVES To examine whether vitamin D supplementation alone or in combination with calcium or other vitamins and minerals given to women during pregnancy can safely improve maternal and neonatal outcomes. SEARCH METHODS For this update, we searched Cochrane Pregnancy and Childbirth's Trials Register (12 July 2018), contacted relevant organisations (15 May 2018), reference lists of retrieved trials and registries at clinicaltrials.gov and WHO International Clinical Trials Registry Platform (12 July 2018). Abstracts were included if they had enough information to extract the data. SELECTION CRITERIA Randomised and quasi-randomised trials evaluating the effect of supplementation with vitamin D alone or in combination with other micronutrients for women during pregnancy in comparison to placebo or no intervention. DATA COLLECTION AND ANALYSIS Two review authors independently i) assessed the eligibility of trials against the inclusion criteria, ii) extracted data from included trials, and iii) assessed the risk of bias of the included trials. The certainty of the evidence was assessed using the GRADE approach. MAIN RESULTS We included 30 trials (7033 women), excluded 60 trials, identified six as ongoing/unpublished trials and two trials are awaiting assessments.Supplementation with vitamin D alone versus placebo/no interventionA total of 22 trials involving 3725 pregnant women were included in this comparison; 19 trials were assessed as having low-to-moderate risk of bias for most domains and three trials were assessed as having high risk of bias for most domains. Supplementation with vitamin D alone during pregnancy probably reduces the risk of pre-eclampsia (risk ratio (RR) 0.48, 95% confidence interval (CI) 0.30 to 0.79; 4 trials, 499 women, moderate-certainty evidence) and gestational diabetes (RR 0.51, 95% CI 0.27 to 0.97; 4 trials, 446 women, moderate-certainty evidence); and probably reduces the risk of having a baby with low birthweight (less than 2500 g) (RR 0.55, 95% CI 0.35 to 0.87; 5 trials, 697 women, moderate-certainty evidence) compared to women who received placebo or no intervention. Vitamin D supplementation may make little or no difference in the risk of having a preterm birth < 37 weeks compared to no intervention or placebo (RR 0.66, 95% CI 0.34 to 1.30; 7 trials, 1640 women, low-certainty evidence). In terms of maternal adverse events, vitamin D supplementation may reduce the risk of severe postpartum haemorrhage (RR 0.68, 95% CI 0.51 to 0.91; 1 trial, 1134 women, low-certainty evidence). There were no cases of hypercalcaemia (1 trial, 1134 women, low-certainty evidence), and we are very uncertain as to whether vitamin D increases or decreases the risk of nephritic syndrome (RR 0.17, 95% CI 0.01 to 4.06; 1 trial, 135 women, very low-certainty evidence). However, given the scarcity of data in general for maternal adverse events, no firm conclusions can be drawn.Supplementation with vitamin D and calcium versus placebo/no interventionNine trials involving 1916 pregnant women were included in this comparison; three trials were assessed as having low risk of bias for allocation and blinding, four trials were assessed as having high risk of bias and two had some components having a low risk, high risk, or unclear risk. Supplementation with vitamin D and calcium during pregnancy probably reduces the risk of pre-eclampsia (RR 0.50, 95% CI 0.32 to 0.78; 4 trials, 1174 women, moderate-certainty evidence). The effect of the intervention is uncertain on gestational diabetes (RR 0.33,% CI 0.01 to 7.84; 1 trial, 54 women, very low-certainty evidence); and low birthweight (less than 2500 g) (RR 0.68, 95% CI 0.10 to 4.55; 2 trials, 110 women, very low-certainty evidence) compared to women who received placebo or no intervention. Supplementation with vitamin D and calcium during pregnancy may increase the risk of preterm birth < 37 weeks in comparison to women who received placebo or no intervention (RR 1.52, 95% CI 1.01 to 2.28; 5 trials, 942 women, low-certainty evidence). No trial in this comparison reported on maternal adverse events.Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D)One trial in 1300 participants was included in this comparison; it was assessed as having low risk of bias. Pre-eclampsia was not assessed. Supplementation with vitamin D + other nutrients may make little or no difference in the risk of preterm birth < 37 weeks (RR 1.04, 95% CI 0.68 to 1.59; 1 trial, 1298 women, low-certainty evidence); or low birthweight (less than 2500 g) (RR 1.12, 95% CI 0.82 to 1.51; 1 trial, 1298 women, low-certainty evidence). It is unclear whether it makes any difference to the risk of gestational diabetes (RR 0.42, 95% CI 0.10 to 1.73) or maternal adverse events (hypercalcaemia no events; hypercalciuria RR 0.25, 95% CI 0.02 to 3.97; 1 trial, 1298 women,) because the certainty of the evidence for both outcomes was found to be very low. AUTHORS' CONCLUSIONS We included 30 trials (7033 women) across three separate comparisons. Our GRADE assessments ranged from moderate to very low, with downgrading decisions based on limitations in study design, imprecision and indirectness.Supplementing pregnant women with vitamin D alone probably reduces the risk of pre-eclampsia, gestational diabetes, low birthweight and may reduce the risk of severe postpartum haemorrhage. It may make little or no difference in the risk of having a preterm birth < 37 weeks' gestation. Supplementing pregnant women with vitamin D and calcium probably reduces the risk of pre-eclampsia but may increase the risk of preterm births < 37 weeks (these findings warrant further research). Supplementing pregnant women with vitamin D and other nutrients may make little or no difference in the risk of preterm birth < 37 weeks' gestation or low birthweight (less than 2500 g). Additional rigorous high quality and larger randomised trials are required to evaluate the effects of vitamin D supplementation in pregnancy, particularly in relation to the risk of maternal adverse events.
Collapse
Affiliation(s)
- Cristina Palacios
- Robert Stempel College of Public Health and Social Work, Florida International UniversityDepartment of Dietetics and Nutrition11200 SW 8th Street, AHC 5 – 323MiamiFloridaUSA33199
| | - Lia K Kostiuk
- University of Wisconsin ‐ MadisonPreventive MedicineMadisonWisconsinUSA53718
| | - Juan Pablo Peña‐Rosas
- World Health OrganizationEvidence and Programme Guidance, Department of Nutrition for Health and Development20 Avenue AppiaGenevaGESwitzerland1211
| | | |
Collapse
|
17
|
Targeted expression profiling by RNA-Seq improves detection of cellular dynamics during pregnancy and identifies a role for T cells in term parturition. Sci Rep 2019; 9:848. [PMID: 30696862 PMCID: PMC6351599 DOI: 10.1038/s41598-018-36649-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/26/2018] [Indexed: 12/29/2022] Open
Abstract
Development of maternal blood transcriptomic markers to monitor placental function and risk of obstetrical complications throughout pregnancy requires accurate quantification of gene expression. Herein, we benchmark three state-of-the-art expression profiling techniques to assess in maternal circulation the expression of cell type-specific gene sets previously discovered by single-cell genomics studies of the placenta. We compared Affymetrix Human Transcriptome Arrays, Illumina RNA-Seq, and sequencing-based targeted expression profiling (DriverMapTM) to assess transcriptomic changes with gestational age and labor status at term, and tested 86 candidate genes by qRT-PCR. DriverMap identified twice as many significant genes (q < 0.1) than RNA-Seq and five times more than microarrays. The gap in the number of significant genes remained when testing only protein-coding genes detected by all platforms. qRT-PCR validation statistics (PPV and AUC) were high and similar among platforms, yet dynamic ranges were higher for sequencing based platforms than microarrays. DriverMap provided the strongest evidence for the association of B-cell and T-cell gene signatures with gestational age, while the T-cell expression was increased with spontaneous labor at term according to all three platforms. We concluded that sequencing-based techniques are more suitable to quantify whole-blood gene expression compared to microarrays, as they have an expanded dynamic range and identify more true positives. Targeted expression profiling achieved higher coverage of protein-coding genes with fewer total sequenced reads, and it is especially suited to track cell type-specific signatures discovered in the placenta. The T-cell gene expression signature was increased in women who underwent spontaneous labor at term, mimicking immunological processes at the maternal-fetal interface and placenta.
Collapse
|
18
|
Pashapour S, Golmohammadlou S, Behroozi-Lak T, Ghasemnejad-Berenji H, Sadeghpour S, Ghasemnejad-Berenji M. Relationship between low maternal vitamin D status and the risk of severe preeclampsia: A case control study. Pregnancy Hypertens 2019; 15:161-165. [DOI: 10.1016/j.preghy.2019.01.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/09/2018] [Accepted: 01/10/2019] [Indexed: 10/27/2022]
|
19
|
Vitamin D and associated perinatal-neonatal outcomes among extremely low-birth-weight infants. J Perinatol 2018; 38:1318-1323. [PMID: 30108343 DOI: 10.1038/s41372-018-0203-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 08/03/2018] [Accepted: 08/06/2018] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To evaluate vitamin D inadequacy among extremely low-birth-weight (ELBW, <1000 g) infants and the association between circulating vitamin D concentrations and perinatal-neonatal outcomes. STUDY DESIGN Prospective cohort study of ELBW infants in the neonatal ICU. Blood was collected within the first 3 days after birth after obtaining informed consent. Circulating 25-hydroxyvitamin D concentrations (25(OH)D) were quantified using liquid chromatography-tandem mass spectroscopy and classified as vitamin D deficient, insufficient, or adequate ( < 20, 20-30, or > 30 ng/mL, respectively). Associations between 25(OH)D and perinatal-neonatal outcomes were determined by multivariable regression, adjusted for covariates that differ in the bivariate analysis. RESULTS Of the 60 ELBW infants enrolled, 13 (22%) were vitamin D deficient, 15 (25%) were insufficient, and 32 (53%) were adequate. 25(OH)D levels were positively associated with fetal growth restriction and prolonged rupture of the membranes. CONCLUSIONS Vitamin D inadequacy was frequent among ELBW infants. Circulating vitamin D concentrations were significantly associated with perinatal outcomes in this contemporary cohort.
Collapse
|
20
|
Wagner CL, Hollis BW. The Implications of Vitamin D Status During Pregnancy on Mother and her Developing Child. Front Endocrinol (Lausanne) 2018; 9:500. [PMID: 30233496 PMCID: PMC6127214 DOI: 10.3389/fendo.2018.00500] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/08/2018] [Indexed: 12/19/2022] Open
Abstract
Pregnancy is a time of tremendous growth and physiological changes for mother and her developing fetus with lifelong implications for the child. The concert of actions that must occur so mother does not reject the foreign tissue of the fetus is substantial. There must be exquisite balance between maternal tolerance to these foreign proteins of paternal origin but also immune surveillance and function such that the mother is not immunocompromised. When this process goes awry, the mother may experience such pregnancy complications as preeclampsia and infections. Vitamin D deficiency affects these processes. Controversy continues with regard to the optimal daily intake of vitamin D, when sunlight exposure should be taken into account, and how to define sufficiency during such vulnerable and critical periods of development. The importance of vitamin D supplementation during pregnancy in preventing some of the health risks to the mother and fetus appears linked to achieving 25(OH)D concentrations >40 ng/mL, the beginning point of the plateau where conversion of the vitamin D metabolite 25(OH)D, the pre-hormone, to 1,25(OH)2D, the active hormone, is optimized. Throughout pregnancy, the delivery of adequate vitamin D substrate-through sunlight or supplement-is required to protect both mother and fetus, and when in sufficient supply, favorably impacts the epigenome of the fetus, and in turn, long term health. There is a growing need for future research endeavors to focus not only on critical period(s) from pre-conception through pregnancy, but throughout life to prevent certain epigenetic changes that adversely affect health. There is urgency based on emerging research to correct deficiency and maintain optimal vitamin D status. The impact of vitamin D and its metabolites on genetic signaling during pregnancy in both mother and fetus is an area of great activity and still in its early stages. While vitamin D repletion during pregnancy minimizes the risk of certain adverse outcomes (e.g., preterm birth, asthma, preeclampsia, and gestational diabetes), the mechanisms of how these processes occur are not fully understood. As we intensify our research efforts in these areas. it is only a matter of time that such mechanisms will be defined.
Collapse
Affiliation(s)
- Carol L. Wagner
- Neonatology, Shawn Jenkins Children's Hospital, Medical University of South Carolina, Charleston, SC, United States
| | | |
Collapse
|
21
|
Rostami M, Tehrani FR, Simbar M, Bidhendi Yarandi R, Minooee S, Hollis BW, Hosseinpanah F. Effectiveness of Prenatal Vitamin D Deficiency Screening and Treatment Program: A Stratified Randomized Field Trial. J Clin Endocrinol Metab 2018; 103:2936-2948. [PMID: 29788364 DOI: 10.1210/jc.2018-00109] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 04/27/2018] [Indexed: 01/02/2023]
Abstract
CONTEXT Despite evidence on the association between hypovitaminosis D and adverse pregnancy outcomes and the positive impact of vitamin D supplementation, no evidence exists supporting a universal screening program in pregnancy as part of routine prenatal care. OBJECTIVE We sought to determine the effectiveness of a prenatal screening program on optimizing 25-hydroxyvitamin D [25(OH)D] levels and preventing pregnancy complications. Also, to identify a safe regimen, we compared several regimens in a subgroup of vitamin D-deficient pregnant women. DESIGN Two cities of Masjed-Soleyman and Shushtar from Khuzestan province, Iran, were selected as the screening and nonscreening arms, respectively. Within the screening arm, a randomized controlled trial was conducted on 800 pregnant women. SETTING Health centers of Masjed-Soleyman and Shushtar cities. PATIENTS OR PARTICIPANTS Pregnant women aged 18 to 40 years. INTERVENTION Women with moderate [25(OH)D, 10 to 20 ng/mL] and severe [25(OH)D, <10 ng/mL] deficiency were randomly divided into four subgroups and received vitamin D3 (D3) until delivery. MAIN OUTCOME MEASURE Maternal concentration of 25(OH)D at delivery and rate of pregnancy complications. RESULTS After supplementation, only 2% of the women in the nonscreening site met the sufficiency level (>20 ng/mL) vs 53% of the women in the screening site. Adverse pregnancy outcomes, including preeclampsia, gestational diabetes mellitus, and preterm delivery, were decreased by 60%, 50%, and 40%, respectively, in the screening site. A D3 injection in addition to monthly 50,000 IU maintenance therapy contributed the most to achievement of sufficient levels at delivery. CONCLUSIONS A prenatal vitamin D screening and treatment program is an effective approach in detecting deficient women, improving 25(OH)D levels, and decreasing pregnancy adverse outcomes.
Collapse
Affiliation(s)
- Maryam Rostami
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medical Sciences, Islamic Azad University, Masjed-Soleyman Branch, Masjed-Soleyman, Khuzestan, Iran
| | - Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Simbar
- Department of Midwifery and Reproductive Health, Midwifery and Reproductive Health Research Center, School of Nursing and Midwifery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Razieh Bidhendi Yarandi
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sonia Minooee
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bruce W Hollis
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Farhad Hosseinpanah
- Obesity Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Vitamin D supplementation for women during pregnancy. Women Birth 2018; 31:e286. [DOI: 10.1016/j.wombi.2017.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 11/06/2017] [Indexed: 11/21/2022]
|
23
|
Curtis EM, Moon RJ, Harvey NC, Cooper C. Maternal vitamin D supplementation during pregnancy. Br Med Bull 2018; 126:57-77. [PMID: 29684104 PMCID: PMC6003599 DOI: 10.1093/bmb/ldy010] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 03/26/2018] [Indexed: 12/19/2022]
Abstract
Introduction Maternal vitamin D status in pregnancy has been linked to many health outcomes in mother and offspring. A wealth of observational studies have reported on both obstetric outcomes and complications, including pre-eclampsia, gestational diabetes, mode and timing of delivery. Many foetal and childhood outcomes are also linked to vitamin D status, including measures of foetal size, body composition and skeletal mineralization, in addition to later childhood outcomes, such as asthma. Sources of data Synthesis of systematic and narrative reviews. Areas of agreement and controversy The findings are generally inconsistent in most areas, and, at present, there is a lack of data from high-quality intervention studies to confirm a causal role for vitamin D in these outcomes. In most areas, the evidence tends towards maternal vitamin D being of overall benefit, but often does not reach statistical significance in meta-analyses. Growing points and areas timely for developing research The most conclusive evidence is in the role of maternal vitamin D supplementation in the prevention of neonatal hypocalcaemia; as a consequence the UK department of health recommends that pregnant women take 400 IU vitamin D daily. High-quality randomized placebo-controlled trials, such as the UK-based MAVIDOS trial, will inform the potential efficacy and safety of vitamin D supplementation in pregnancy across a variety of outcomes.
Collapse
Affiliation(s)
- Elizabeth M Curtis
- MRC Lifecourse Epidemiology Unit, University of Southampton,
Southampton, SO16 6YD, UK
| | - Rebecca J Moon
- MRC Lifecourse Epidemiology Unit, University of Southampton,
Southampton, SO16 6YD, UK
- Paediatric Endocrinology, Southampton University Hospitals NHS
Foundation Trust, Southampton, SO16 6YD, UK
| | - Nicholas C Harvey
- MRC Lifecourse Epidemiology Unit, University of Southampton,
Southampton, SO16 6YD, UK
- NIHR Southampton Biomedical Research Centre, University of
Southampton and University Hospital Southampton NHS Foundation Trust, Tremona Road,
Southampton, SO16 6YD, UK
| | - Cyrus Cooper
- MRC Lifecourse Epidemiology Unit, University of Southampton,
Southampton, SO16 6YD, UK
- NIHR Southampton Biomedical Research Centre, University of
Southampton and University Hospital Southampton NHS Foundation Trust, Tremona Road,
Southampton, SO16 6YD, UK
- NIHR Oxford Biomedical Research Centre, University of Oxford,
Oxford, OX3 7LD, UK
| |
Collapse
|
24
|
Hong J, Hatchell KE, Bradfield JP, Bjonnes A, Chesi A, Lai CQ, Langefeld CD, Lu L, Lu Y, Lutsey PL, Musani SK, Nalls MA, Robinson-Cohen C, Roizen JD, Saxena R, Tucker KL, Ziegler JT, Arking DE, Bis JC, Boerwinkle E, Bottinger EP, Bowden DW, Gilsanz V, Houston DK, Kalkwarf HJ, Kelly A, Lappe JM, Liu Y, Michos ED, Oberfield SE, Palmer ND, Rotter JI, Sapkota B, Shepherd JA, Wilson JG, Basu S, de Boer IH, Divers J, Freedman BI, Grant SFA, Hakanarson H, Harris TB, Kestenbaum BR, Kritchevsky SB, Loos RJF, Norris JM, Norwood AF, Ordovas JM, Pankow JS, Psaty BM, Sanghera DK, Wagenknecht LE, Zemel BS, Meigs J, Dupuis J, Florez JC, Wang T, Liu CT, Engelman CD, Billings LK. Transethnic Evaluation Identifies Low-Frequency Loci Associated With 25-Hydroxyvitamin D Concentrations. J Clin Endocrinol Metab 2018; 103:1380-1392. [PMID: 29325163 PMCID: PMC6276579 DOI: 10.1210/jc.2017-01802] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 01/03/2018] [Indexed: 01/08/2023]
Abstract
Context Vitamin D inadequacy is common in the adult population of the United States. Although the genetic determinants underlying vitamin D inadequacy have been studied in people of European ancestry, less is known about populations with Hispanic or African ancestry. Objective The Trans-Ethnic Evaluation of Vitamin D (TRANSCEN-D) genomewide association study (GWAS) consortium was assembled to replicate genetic associations with 25-hydroxyvitamin D [25(OH)D] concentrations from the Study of Underlying Genetic Determinants of Vitamin D and Highly Related Traits (SUNLIGHT) meta-analyses of European ancestry and to identify genetic variants related to vitamin D concentrations in African and Hispanic ancestries. Design Ancestry-specific (Hispanic and African) and transethnic (Hispanic, African, and European) meta-analyses were performed with Meta-Analysis Helper software (METAL). Patients or Other Participants In total, 8541 African American and 3485 Hispanic American (from North America) participants from 12 cohorts and 16,124 European participants from SUNLIGHT were included in the study. Main Outcome Measures Blood concentrations of 25(OH)D were measured for all participants. Results Ancestry-specific analyses in African and Hispanic Americans replicated single nucleotide polymorphisms (SNPs) in GC (2 and 4 SNPs, respectively). An SNP (rs79666294) near the KIF4B gene was identified in the African American cohort. Transethnic evaluation replicated GC and DHCR7 region SNPs. Additionally, the transethnic analyses revealed SNPs rs719700 and rs1410656 near the ANO6/ARID2 and HTR2A genes, respectively. Conclusions Ancestry-specific and transethnic GWASs of 25(OH)D confirmed findings in GC and DHCR7 for African and Hispanic American samples and revealed findings near KIF4B, ANO6/ARID2, and HTR2A. The biological mechanisms that link these regions with 25(OH)D metabolism warrant further investigation.
Collapse
Affiliation(s)
- Jaeyoung Hong
- Department of Biostatistics, Boston University School of Public Health, Boston,
Massachusetts
| | - Kathryn E Hatchell
- Department of Population Health Sciences, University of Wisconsin–Madison
School of Medicine and Public Health, Madison, Wisconsin
| | - Jonathan P Bradfield
- Center for Applied Genomics, Division of Human Genetics, The Children’s
Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Andrew Bjonnes
- Center for Genomic Medicine, Massachusetts General Hospital, Boston,
Massachusetts
| | - Alessandra Chesi
- The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Chao-Qiang Lai
- USDA-ARS Human Nutrition Research Center on Aging at Tufts University, Boston,
Massachusetts
| | | | - Lingyi Lu
- Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of
Medicine at Mount Sinai, New York, New York
| | - Pamela L Lutsey
- Division of Epidemiology and Community Health, University of Minnesota,
Minneapolis, Minnesota
| | - Solomon K Musani
- Department of Medicine, University of Mississippi Medical Center, Jackson,
Mississippi
| | - Mike A Nalls
- Data Tecnica International, Glen Echo, Maryland
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes
of Health, Bethesda, Maryland
| | - Cassianne Robinson-Cohen
- Kidney Research Institute, Division of Nephrology, Department of Medicine,
University of Washington, Seattle, Washington
| | - Jeffery D Roizen
- Center for Applied Genomics, Division of Human Genetics, The Children’s
Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Endocrinology, Children’s Hospital of Philadelphia, Philadelphia,
Pennsylvania
| | - Richa Saxena
- Center for Genomic Medicine, Massachusetts General Hospital, Boston,
Massachusetts
| | - Katherine L Tucker
- Department of Biomedical and Nutritional Sciences, University of Massachusetts
Lowell, Lowell, Massachusetts
| | - Julie T Ziegler
- Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Dan E Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University
School of Medicine, Baltimore, Maryland
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of
Washington, Seattle, Washington
| | - Eric Boerwinkle
- University of Texas Health Science Center at Houston, Houston, Texas
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of
Medicine at Mount Sinai, New York, New York
| | - Donald W Bowden
- Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Vicente Gilsanz
- Department of Radiology, Children’s Hospital of Los Angeles, Keck School of
Medicine, University of Southern California, Los Angeles, California
| | - Denise K Houston
- Department of Internal Medicine, Section on Gerontology and Geriatric
Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Heidi J Kalkwarf
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s
Hospital Medical Center, Cincinnati, Ohio
| | - Andrea Kelly
- The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Perelman School of Medicine at University of Pennsylvania, Philadelphia,
Pennsylvania
| | | | - Yongmei Liu
- Department of Epidemiology and Prevention, Division of Public Health Sciences,
Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Erin D Michos
- Ciccarone Center for the Prevention of Heart Disease, Johns Hopkins University
School of Medicine, Baltimore, Maryland
| | - Sharon E Oberfield
- Division of Pediatric Endocrinology, Diabetes and Metabolism, Columbia
University Medical Center, New York, New York
| | | | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Department of
Pediatrics and Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,
Torrance, California
| | - Bishwa Sapkota
- Department of Pediatrics, College of Medicine, University of Oklahoma Health
Sciences Center, Oklahoma City, Oklahoma
| | - John A Shepherd
- University of California San Francisco School of Medicine, San Francisco,
California
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical
Center, Jackson, Mississippi
| | - Saonli Basu
- Division of Biostatistics, University of Minnesota, Minneapolis,
Minnesota
| | - Ian H de Boer
- Kidney Research Institute, Division of Nephrology, Department of Medicine,
University of Washington, Seattle, Washington
| | - Jasmin Divers
- Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | - Struan F A Grant
- Center for Applied Genomics, Division of Human Genetics, The Children’s
Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Endocrinology, Children’s Hospital of Philadelphia, Philadelphia,
Pennsylvania
- Department of Pediatrics, Perelman School of Medicine, University of
Pennsylvania, Philadelphia, Pennsylvania
| | - Hakon Hakanarson
- Center for Applied Genomics, Division of Human Genetics, The Children’s
Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine, University of
Pennsylvania, Philadelphia, Pennsylvania
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on
Aging, Bethesda, Maryland
| | - Bryan R Kestenbaum
- Kidney Research Institute, Division of Nephrology, Department of Medicine,
University of Washington, Seattle, Washington
| | - Stephen B Kritchevsky
- Department of Internal Medicine, Section on Gerontology and Geriatric
Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of
Medicine at Mount Sinai, New York, New York
- The Mindich Child Health and Development Institute, Icahn School of Medicine
at Mount Sinai, New York, New York
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, University of
Colorado Denver, Aurora, Colorado
| | - Arnita F Norwood
- Department of Medicine, University of Mississippi Medical Center, Jackson,
Mississippi
| | - Jose M Ordovas
- Nutrition and Genomics, JM-USDA Human Nutrition Research Center on Aging at
Tufts University, Boston, Massachusetts
| | - James S Pankow
- Division of Epidemiology and Community Health, University of Minnesota,
Minneapolis, Minnesota
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of
Washington, Seattle, Washington
- University of Washington and Department of Epidemiology and Health Sciences,
University of Washington, Seattle, Washington
- Kaiser Permanente Washington Health Research Institute, Seattle,
Washington
| | - Dharambir K Sanghera
- Department of Pediatrics, College of Medicine, University of Oklahoma Health
Sciences Center, Oklahoma City, Oklahoma
- Department of Pharmaceutical Sciences, College of Pharmacy, University of
Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Oklahoma Center for Neuroscience, Oklahoma City, Oklahoma
| | | | - Babette S Zemel
- The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Perelman School of Medicine at University of Pennsylvania, Philadelphia,
Pennsylvania
| | - James Meigs
- Division of General Internal Medicine, Massachusetts General Hospital, Harvard
Medical School, Boston, Massachusetts
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston,
Massachusetts
- National Heart, Lung, and Blood Institute’s Framingham Heart Study,
Framingham, Massachusetts
| | - Jose C Florez
- Center for Genomic Medicine, Massachusetts General Hospital, Boston,
Massachusetts
- Diabetes Unit, Massachusetts General Hospital, Boston, Massachusetts
- Programs in Metabolism and Medical & Population Genetics, Broad Institute,
Cambridge, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | | | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston,
Massachusetts
| | - Corinne D Engelman
- Department of Population Health Sciences, University of Wisconsin–Madison
School of Medicine and Public Health, Madison, Wisconsin
| | - Liana K Billings
- NorthShore University HealthSystem, Evanston, Illinois
- University of Chicago Pritzker School of Medicine, Chicago, Illinois
| |
Collapse
|
25
|
Iannuzzo G, Forte F, Lupoli R, Di Minno MND. Association of Vitamin D deficiency with peripheral arterial disease: a meta-analysis of literature studies. J Clin Endocrinol Metab 2018; 103:4951505. [PMID: 29590347 DOI: 10.1210/jc.2018-00136] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/19/2018] [Indexed: 02/04/2023]
Abstract
CONTEXT Vitamin D deficiency patients have an increased cardiovascular (CV) morbidity and mortality. Contrasting data are available about the association between peripheral arterial disease (PAD) and Vitamin D status. OBJECTIVE To perform a meta-analysis of studies evaluating the association between Vitamin D status and PAD. DATA SOURCES Studies were systematically searched in the PubMed, Web of Science, Scopus and EMBASE databases. RESULTS Ten studies with data on Vitamin D levels in 2,079 PAD patients and 18,233 non-PAD controls and 6 studies on the prevalence of PAD in 23,171 subjects with Vitamin D deficiency (<20 ng/ml), 48,311 subjects with Vitamin D insufficiency (20-30 ng/ml) and 27,910 with normal Vitamin D levels (>30 ng/ml) were included. Compared to controls, PAD patients showed significantly lower Vitamin D levels (MD: -2.24 ng/ml; 95%CI: -3.38, -1.10; p<0.001, I2=86.5%; p<0.001). Moreover, a higher prevalence of PAD was found both in subjects with Vitamin D insufficiency (OR: 1.098, 95%CI: 1.010-1.195, p=0.029, I2: 0%, p=0,600) and in subjects with Vitamin D deficiency (OR: 1.484, 95%CI: 1.348-1.635, p<0.001, I2: 7.65%, p=0,367) compared with controls with normal Vitamin D levels. Sensitivity analyses and the analysis of data on the cumulative risk of PAD according to Vitamin D levels derived from multivariate analysis consistently confirmed results. CONCLUSIONS PAD patients have lower vitamin D levels than controls and both Vitamin D deficiency and Vitamin D insufficiency are significantly associated with PAD. Reduced Vitamin D levels might represent an independent risk factor for PAD and, in turn, for CV events.
Collapse
Affiliation(s)
- Gabriella Iannuzzo
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Francesco Forte
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Roberta Lupoli
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, Naples, Italy
| | | |
Collapse
|
26
|
Schulz EV, Cruze L, Wei W, Gehris J, Wagner CL. Maternal vitamin D sufficiency and reduced placental gene expression in angiogenic biomarkers related to comorbidities of pregnancy. J Steroid Biochem Mol Biol 2017; 173:273-279. [PMID: 28216083 PMCID: PMC6349226 DOI: 10.1016/j.jsbmb.2017.02.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 01/30/2017] [Accepted: 02/05/2017] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Maternal circulating 25-hydroxyvitamin D [25(OH)D] has been shown to optimize production of 1,25-dihydroxyvitamin D [1,25(OH)2D] during pregnancy at approximately 100nmoles/L, which has pronounced effects on fetal health outcomes. Additionally, associations are noted between low maternal 25(OH)D concentrations and vascular pregnancy complications, such as preeclampsia. To further elucidate the effects of vitamin D activity in pregnancy, we investigated the role of maternal 25(OH)D, the nutritional indicator of vitamin D status, in relation to placental maintenance and, specifically, expression of placental gene targets related to angiogenesis and vitamin D metabolism. METHODS A focused analysis of placental mRNA expression related to angiogenesis, pregnancy maintenance, and vitamin D metabolism was conducted in placentas from 43 subjects enrolled in a randomized controlled trial supplementing 400IU or 4400IU of vitamin D3 per day during pregnancy. Placental mRNA was isolated from biopsies within one hour of delivery, followed by quantitative PCR. We classified pregnant women with circulating concentrations of <100nmoles/L as deficient and those with ≥100nmoles/L as sufficient. The value of each gene's change in the PCR cycle threshold (ΔCT), which is a relative measure of target concentration, was compared with maternal 25(OH)D concentrations <100nmoles/L and ≥100nmoles/L based on a two-sample Wilcoxon test. RESULTS Soluble FMS-like tyrosine kinase 1 (sFlt-1) and vascular endothelial growth factor (VEGF) gene expression was significantly downregulated in the maternal subgroup with circulating 25(OH)D ≥100ng/mL compared to the subgroup <100ng/mL. DISCUSSION Here, we report a significant association between maternal vitamin D status and the expression of sFlt-1 and VEGF at the mRNA level. Achieving maternal circulating 25(OH)D ≥100nmoles/L suggests the impact of maternal vitamin D3 supplementation on gene transcription in the placenta, thereby potentially decreasing antiangiogenic factors that may contribute to vascular pregnancy complications.
Collapse
Affiliation(s)
- Elizabeth V Schulz
- Departments of Pediatrics, 169 Ashley Avenue, Charleston, SC, 29425, USA.
| | - Lori Cruze
- OB/GYN, 169 Ashley Avenue, Charleston, SC, 29425, USA
| | - Wei Wei
- Public Health Sciences, 169 Ashley Avenue, Charleston, SC, 29425, USA
| | - John Gehris
- Comparative Medicine Medical University of South Carolina, 169 Ashley Avenue, Charleston, SC, 29425, USA
| | - Carol L Wagner
- Departments of Pediatrics, 169 Ashley Avenue, Charleston, SC, 29425, USA
| |
Collapse
|
27
|
Hollis BW, Wagner CL. Vitamin D supplementation during pregnancy: Improvements in birth outcomes and complications through direct genomic alteration. Mol Cell Endocrinol 2017; 453:113-130. [PMID: 28188842 DOI: 10.1016/j.mce.2017.01.039] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 12/16/2022]
Abstract
Pregnancy represents a time of rapid change, including dramatic shifts in vitamin D metabolism. Circulating concentrations of the active form of vitamin D-1,25(OH)2D skyrocket early in pregnancy to levels that would be toxic to a nonpregnant adult, signaling a decoupling of vitamin D from the classic endocrine calcium metabolic pathway, likely serving an immunomodulatory function in the mother and her developing fetus. In this review, we summarize the unique aspects of vitamin D metabolism and the data surrounding vitamin D requirements during this important period. Both observational and clinical trials are reviewed in the context of vitamin D's health effects during pregnancy that include preeclampsia, preterm birth, and later disease states such as asthma and multiple sclerosis. With enhanced knowledge about vitamin D's role as a preprohormone, it is clear that recommendations about supplementation must mirror what is clinically relevant and evidence-based. Future research that focuses on the critical period(s) leading up to conception and during pregnancy to correct deficiency or maintain optimal vitamin D status remains to be studied. In addition, what effects vitamin D has on genetic signatures that minimize the risk to the mother and her developing fetus have not been elucidated. Clearly, while there is much more research that needs to be performed, our understanding of vitamin D requirements during pregnancy has advanced significantly during the last few decades.
Collapse
Affiliation(s)
- Bruce W Hollis
- Medical University of South Carolina, Division of Neonatology, Department of Pediatrics, 173 Ashley Avenue, MSC 514, Charleston, SC 29425, United States.
| | - Carol L Wagner
- Medical University of South Carolina, Division of Neonatology, Department of Pediatrics, 173 Ashley Avenue, MSC 514, Charleston, SC 29425, United States
| |
Collapse
|
28
|
New insights into the vitamin D requirements during pregnancy. Bone Res 2017; 5:17030. [PMID: 28868163 PMCID: PMC5573964 DOI: 10.1038/boneres.2017.30] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/10/2017] [Accepted: 04/22/2017] [Indexed: 12/22/2022] Open
Abstract
Pregnancy represents a dynamic period with physical and physiological changes in both the mother and her developing fetus. The dramatic 2–3 fold increase in the active hormone 1,25(OH)2D concentrations during the early weeks of pregnancy despite minimal increased calcium demands during that time of gestation and which are sustained throughout pregnancy in both the mother and fetus suggests an immunomodulatory role in preventing fetal rejection by the mother. While there have been numerous observational studies that support the premise of vitamin D's role in maintaining maternal and fetal well-being, until recently, there have been few randomized clinical trials with vitamin D supplementation. One has to exhibit caution, however, even with RCTs, whose results can be problematic when analyzed on an intent-to-treat basis and when there is high non-adherence to protocol (as if often the case), thereby diluting the potential good or harm of a given treatment at higher doses. As such, a biomarker of a drug or in this case “vitamin” or pre-prohormone is better served. For these reasons, the effect of vitamin D therapies using the biomarker circulating 25(OH)D is a far better indicator of true “effect.” When pregnancy outcomes are analyzed using the biomarker 25(OH)D instead of treatment dose, there are notable differences in maternal and fetal outcomes across diverse racial/ethnic groups, with improved health in those women who attain a circulating 25(OH)D concentration of at least 100 nmol·L−1 (40 ng·mL−1). Because an important issue is the timing or initiation of vitamin D treatment/supplementation, and given the potential effect of vitamin D on placental gene expression and its effects on inflammation within the placenta, it appears crucial to start vitamin D treatment before placentation (and trophoblast invasion); however, this question remains unanswered. Additional work is needed to decipher the vitamin D requirements of pregnant women and the optimal timing of supplementation, taking into account a variety of lifestyles, body types, baseline vitamin D status, and maternal and fetal vitamin D receptor (VDR) and vitamin D binding protein (VDBP) genotypes. Determining the role of vitamin D in nonclassical, immune pathways continues to be a challenge that once answered will substantiate recommendations and public health policies.
Collapse
|
29
|
Lupoli R, Vaccaro A, Ambrosino P, Poggio P, Amato M, Di Minno MND. Impact of Vitamin D deficiency on subclinical carotid atherosclerosis: a pooled analysis of cohort studies. J Clin Endocrinol Metab 2017; 102:2146-2153. [PMID: 28609831 DOI: 10.1210/jc.2017-00342] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/02/2017] [Indexed: 01/24/2023]
Abstract
CONTEXT Vitamin D deficiency patients have an increased cardiovascular (CV) morbidity and mortality. Carotid intima-media thickness (IMT) and carotid plaques are markers of subclinical atherosclerosis and predictors of CV events. OBJECTIVE To perform a meta-analysis of studies evaluating the impact of Vitamin D deficiency on common carotid artery IMT (CCA-IMT) and on the prevalence of carotid plaques. DATA SOURCES Studies were systematically searched in the PubMed, Web of Science, Scopus and EMBASE databases. RESULTS Twenty-one studies (3,777 Vitamin D deficiency patients and 4,792 controls) with data on CCA-IMT and 6 studies (1,889 Vitamin D deficiency patients and 2,883 controls) on the prevalence of carotid plaques were included. Compared to controls, Vitamin D deficiency patients showed a significantly higher CCA-IMT (mean difference [MD]: 0.043 mm; 95%CI: 0.030, 0.056; P<0.001), and an increased prevalence of carotid plaques (Odds Ratio [OR]: 2.29, 95%CI: 1.03-5.11; P=0.043) with an attributable risk of 35.9%. When selecting studies specifically including patients with diabetes, the prevalence of carotid plaques in Vitamin D deficiency patients than in controls resulted higher (OR: 3.27; 95%CI: 1,62-6.62; P=0.001). A significant difference in CCA-IMT was confirmed when comparing patients with Vitamin D insufficiency to controls (MD: 0.011; 95%CI: 0.010-0.012, P<0.001). Sensitivity analyses substantially confirmed results and regression models showed that with the exception of LDL-cholesterol, HDL-cholesterol, triglycerides and the prevalence of hypercholesterolemia, all the other clinical and demographic co-variates significantly impacted on the difference in CCA-IMT between Vitamin D deficiency patients and controls. CONCLUSIONS Both Vitamin D deficiency and Vitamin D insufficiency are associated with subclinical atherosclerosis, potentially suggesting an increased CV risk in these clinical settings.
Collapse
Affiliation(s)
- Roberta Lupoli
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Andrea Vaccaro
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Pasquale Ambrosino
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Paolo Poggio
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | | | | |
Collapse
|