1
|
Ozarslan N, Mong C, Ategeka J, Li L, Buarpung S, Robinson JF, Kizza J, Kakuru A, Kamya MR, Dorsey G, Rosenthal PJ, Gaw SL. Placental malaria induces a unique methylation profile associated with fetal growth restriction. Epigenetics 2025; 20:2475276. [PMID: 40051167 PMCID: PMC11901535 DOI: 10.1080/15592294.2025.2475276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/12/2025] Open
Abstract
Fetal growth restriction (FGR) is associated with perinatal death and adverse birth outcomes, as well as long-term complications, including increased childhood morbidity, abnormal neurodevelopment, and cardio-metabolic diseases in adulthood. Placental epigenetic reprogramming associated with FGR may mediate these long-term outcomes. Placental malaria (PM), characterized by sequestration of Plasmodium falciparum-infected erythrocytes in placental intervillous space, is the leading global cause of FGR, but its impact on placental epigenetics is unknown. We hypothesized that placental methylomic profiling would reveal common and distinct mechanistic pathways of non-malarial and PM-associated FGR. We analyzed placentas from a US cohort with no malaria exposure (n = 12) and a cohort from eastern Uganda, a region with a high prevalence of malaria (n = 12). From each site, 8 cases of FGR and 4 healthy controls were analyzed. PM was diagnosed by placental histopathology. We compared the methylation levels of over 850K CpGs of the placentas using Infinium MethylationEPIC v1 microarray. Non-malarial FGR was associated with 65 differentially methylated CpGs (DMCs), whereas PM-FGR was associated with 133 DMCs, compared to their corresponding controls without FGR. One DMC (cg16389901, located in the promoter region of BMP4) was commonly hypomethylated in both groups. We identified 522 DMCs between non-malarial FGR vs. PM-FGR placentas, independent of differing geographic location or cellular composition. Placentas with PM-associated FGR have distinct methylation profiles compared to placentas with non-malarial FGR, suggesting novel epigenetic reprogramming in response to malaria. Larger cohort studies are needed to determine the distinct long-term health outcomes in PM-associated FGR pregnancies.
Collapse
Affiliation(s)
- Nida Ozarslan
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Corina Mong
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - John Ategeka
- Infectious Diseases Research Collaboration, Uganda
| | - Lin Li
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Sirirak Buarpung
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Joshua F. Robinson
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Jimmy Kizza
- Infectious Diseases Research Collaboration, Uganda
| | - Abel Kakuru
- Infectious Diseases Research Collaboration, Uganda
| | - Moses R. Kamya
- Infectious Diseases Research Collaboration, Uganda
- Department of Medicine, Makerere University, Kampala, Uganda
| | - Grant Dorsey
- Division of HIV, Global Medicine, and Infectious Diseases, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Philip J. Rosenthal
- Division of HIV, Global Medicine, and Infectious Diseases, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Stephanie L. Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
| |
Collapse
|
2
|
Fioretto MN, Barata LA, de Andrade Felipe VA, Dos Santos SAA, Maciel FA, Ribeiro IT, Mattos R, Baptista HS, Bueno G, Fagundes FL, Portela LMF, Scarano WR, Seiva FRF, Lima CAH, Justulin LA. Long-term effects of maternal protein restriction on adrenal proteomic profile and steroidogenesis in male offspring rats. Cell Signal 2025; 130:111707. [PMID: 40032160 DOI: 10.1016/j.cellsig.2025.111707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/10/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Maternal protein restriction (MPR) can significantly affect offspring's early development and aging, impacting several organs, including the adrenal glands. This study evaluated the adrenal proteomic profile in male rat offspring exposed to MPR during pregnancy and lactation. Male offspring were divided into two groups: Control (CTR), born to dams fed a normoprotein diet (17 % protein), and Gestational and Lactational Low-Protein (GLLP), born to dams fed a low-protein diet (6 % protein) throughout gestation and lactation, and after received control diet. Offspring were euthanized at postnatal day (PND) 21 or PND 540. Blood samples and adrenal glands were processed for histological, metabolic, molecular, and proteomic assessments. At PND21, the GLLP group exhibited reduced adrenal gland mass and cortical thickness. At PND21, the proteomic landscape showed that the most impacted biological pathways were associated with decreased steroid hormone synthesis, increased glucose metabolism, and stress response. At PND540, the main impacts were increased apoptotic pathway, stress response, and steroid hormone synthesis, with decreased glucose metabolism. At PND 540, the GLLP group showed higher adrenal collagen content and elevated apoptosis. Age-related changes included decreased peroxiredoxin 3 and increased expression of aldosterone synthase (Cyp11b2). Furthermore, steroid 11-Beta-Hydroxylase (Cyp11b1) expression decreased at PND540, alongside reduced serum aldosterone and elevated serum corticosterone levels. These results suggest that MPR modulates the adrenal glands' proteomic profile, serving as a pivotal mechanism underpinning diverse systemic diseases. It influences adrenal morphophysiology early in life, with long-lasting consequences for cellular stress, immune response, and catabolic pathways in male offspring with aging.
Collapse
Affiliation(s)
- Matheus Naia Fioretto
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luísa Annibal Barata
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | | | - Sérgio Alexandre Alcantara Dos Santos
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil.; Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Flávia Alessandra Maciel
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Isabelle Tenori Ribeiro
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Renato Mattos
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Hecttor Sebástian Baptista
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Gabriela Bueno
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Felipe Leonardo Fagundes
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luiz Marcos Frediane Portela
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Wellerson Rodrigo Scarano
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Fábio Rodrigues Ferreira Seiva
- Department of Chemical and Biological Sciences, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Clélia Akiko Hiruma Lima
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luis Antonio Justulin
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil..
| |
Collapse
|
3
|
Haiden N, Luque V, Domellöf M, Hill S, Kivelä L, de Koning B, Kӧglmeier J, Moltu SJ, Norsa L, De Pipaon MS, Savino F, Verduci E, Bronsky J. Assessment of growth status and nutritional management of prematurely born infants after hospital discharge: A position paper of the ESPGHAN Nutrition Committee. J Pediatr Gastroenterol Nutr 2025. [PMID: 40341618 DOI: 10.1002/jpn3.70054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 02/28/2025] [Accepted: 03/26/2025] [Indexed: 05/10/2025]
Abstract
OBJECTIVES The aim of the manuscript is to provide evidence-based or expert consensus-based recommendations for growth assessment and nutritional management of preterm-born infants during the post-discharge period. METHODS The search was conducted in Pubmed, MEDLINE, EMBASE, and Cochrane Database of Systematic Reviews using the MESH terms: infant, preterm infant, low birth weight, infant food, nutritional status, nutrients, breast feeding, infant formula, human milk, dietary supplements, vitamins, iron, vitamin D, minerals, energy intake, weaning, and baby led. Overall, 402 papers were identified and screened, from which 101 publications were included in the present position paper. In the absence of evidence, recommendations reflect the authors' combined expert opinion. Final consensus was obtained through multiple e-mail exchanges and meetings with the Committee of Nutrition of the European Society for Paediatric Gastroenterology, Hepatology, and Nutrition. RESULTS Continuous growth monitoring through measurements of weight, length, and HC post-discharge is recommended to identify growth faltering (GF) or undernutrition. To prevent disproportionate growth, weight-for-length z-scores should be included in the assessment when term equivalent age is reached. Infants discharged with a significant drop in weight and length, exceeding a -2 standard deviation loss, require tailored nutritional support to address long-term growth challenges and to support recovery to normal growth rates. Breastfeeding is highly recommended for all infants when feasible. Infants needing to catch up in growth should be given supplements, such as HMF For those fed with formula, an adequate protein: energy ratio, minerals, and trace elements should be supplied to facilitate catch-up growth. The start of solid foods should coincide with the infant's neurological developmental milestones, rather than adhering strictly to a set age. Vitamin D and iron supplementation (with regular ferritin monitoring) is recommended through at least 12 months CA. CONCLUSION For preterm infants, close monitoring of growth after discharge and nutritional assessment is essential to identify those at high risk for GF or undernutrition and to provide individualized nutritional support when needed. These patients should either be referred to a specialized center for pediatric nutritional care or, alternatively, their general pediatrician should receive appropriate training on the subject.
Collapse
Affiliation(s)
- Nadja Haiden
- Department of Neonatology, Kepler University Hospital Linz, Linz, Austria
| | - Veronica Luque
- Paediatric Nutrition and Development Research Unit, Universitat Rovira i Virgili-IISPV, Tarragona, Spain
| | - Magnus Domellöf
- Department of Clinical Sciences, Umeå University, Umeå, Sweden
| | - Susan Hill
- Nutrition and Intestinal Failure Division, Gastroenterology Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Laura Kivelä
- Celiac Disease Research Center, Tampere University, Tampere, Finland
- Department of Pediatrics, Tampere University Hospital, Tampere, Finland
- Children's Hospital, Helsinki University Hospital, Helsinki, Finland
- Department of Pediatric Research, University of Oslo, Oslo, Norway
| | - Barbara de Koning
- Department of Pediatric Gastroenterology, Erasmus MC Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Jutta Kӧglmeier
- Unit of Nutrition and Intestinal Failure Rehabilitation, Department of Paediatric Gastroenterology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Sissel J Moltu
- Department of Neonatal Intensive Care, Oslo University Hospital, Oslo, Norway
| | - Lorenzo Norsa
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| | - Miguel Saenz De Pipaon
- Neonatology Hospital La Paz Institute for Health Research - IdiPAZ (Universidad Autónoma de Madrid), Madrid, Spain
| | - Francesco Savino
- Department of Patologia e cura del bambino "Regina Margherita" Regina Margherita Children Hospital, Azienda Ospedaliera Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Elvira Verduci
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| | - Jiri Bronsky
- Department of Paediatrics, University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
4
|
Blank M, Donald S, Parkin L. Small for Gestational Age Coded Diagnoses in Aotearoa New Zealand's Administrative Health Datasets: A Validation Study. Health Sci Rep 2025; 8:e70610. [PMID: 40309634 PMCID: PMC12042771 DOI: 10.1002/hsr2.70610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 02/19/2025] [Accepted: 03/11/2025] [Indexed: 05/02/2025] Open
Abstract
Background and Aims Inaccurate coding of small for gestational age (SGA) infants in routinely collected health data has implications for research based on those data. We aimed to estimate the sensitivity and specificity of coded SGA diagnoses in New Zealand's routinely collected hospitalisation and mortality data, and determine whether sensitivity and specificity varied by infant, pregnancy, and maternal characteristics. Methods We estimated birthweight centiles of live and stillborn infants delivered in New Zealand between 2005 and 2020 using the Fenton Population Reference Calculator and the GROW Customised Bulk Centile Calculator (New Zealand version); values of the relevant variables (including gestational age, birthweight, infant sex, and others) were sourced from routinely collected national health data. We compared the SGA status derived from the calculators with coded SGA diagnoses (ICD-10-AM P051) in hospitalisation and mortality data. We estimated sensitivity and specificity ratios comparing coded diagnoses with each of the birthweight calculators using a generalised linear model, adjusting for infant, pregnancy, and maternal characteristics. Results This analysis included 887,871 infants, with 15,850 (1.8%) having a coded SGA diagnosis. By contrast, the number and proportion of babies classified as SGA using the Fenton and GROW calculators were 80,541 (9.1%) and 138,866 (15.6%), respectively. Overall, compared with the Fenton calculator, the sensitivity of coded SGA diagnoses was 13.1% (specificity 99.3%). Compared with the GROW calculator, the sensitivity was 9.8% (specificity 99.7%). Conclusion In New Zealand, population-level research involving SGA diagnoses should derive birthweight centiles using an appropriate calculator instead of using ICD-10-AM coded diagnoses.
Collapse
Affiliation(s)
- Mei‐Ling Blank
- Department of Preventive and Social Medicine (Te Tari Hauora Tūmatanui)University of Otago (Ōtākou Whakaihu Waka)DunedinNew Zealand
| | - Sarah Donald
- Department of Preventive and Social Medicine (Te Tari Hauora Tūmatanui)University of Otago (Ōtākou Whakaihu Waka)DunedinNew Zealand
- Pharmacoepidemiology Research NetworkUniversity of Otago (Ōtākou Whakaihu Waka)DunedinNew Zealand
| | - Lianne Parkin
- Department of Preventive and Social Medicine (Te Tari Hauora Tūmatanui)University of Otago (Ōtākou Whakaihu Waka)DunedinNew Zealand
- Pharmacoepidemiology Research NetworkUniversity of Otago (Ōtākou Whakaihu Waka)DunedinNew Zealand
| |
Collapse
|
5
|
Ushida T, Nosaka R, Nakatochi M, Kobayashi Y, Tano S, Fuma K, Matsuo S, Imai K, Sato Y, Hayakawa M, Kajiyama H, Kotani T. Effect of chorioamnionitis on postnatal growth in very preterm infants: a population-based study in Japan. Arch Gynecol Obstet 2025; 311:1321-1330. [PMID: 39354115 PMCID: PMC12033191 DOI: 10.1007/s00404-024-07757-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/21/2024] [Indexed: 10/03/2024]
Abstract
PURPOSE There is growing evidence that preterm infants born to mothers with chorioamnionitis (CAM) have increased risk of various neonatal morbidities and long-term neurological disorders; however, the effect of CAM on postnatal growth remains insufficiently investigated. This study evaluated the effect of histological CAM on postnatal growth trajectories in very preterm infants using a nationwide neonatal database in Japan. METHOD A multicenter retrospective study was conducted using clinical data of 4220 preterm neonates who weighed ≤ 1500 g and were born at < 32 weeks of gestation between 2003-2017 (CAM group: n = 2110; non-CAM group: n = 2110). Z-scores for height and weight were evaluated at birth and 3 years of age. Univariable and multivariable analyses were conducted to evaluate the effect of histological CAM on ΔZ-scores of height and weight during the first three years with a stratification by infant sex and the stage of histological CAM. RESULTS Multivariable analyses showed that histological CAM was associated with accelerated postnatal increase (ΔZ-score) in weight (β coefficient [95% confidence interval]; 0.10 [0.00 to 0.20]), but not in height among females (0.06 [- 0.04 to 0.15]) and not in height and weight among males (0.04 [- 0.04 to 0.12] and 0.02 [- 0.07 to 0.11], respectively). An interaction analysis demonstrated no significant difference in the effect of histological CAM on the ΔZ-scores of height and weight during the first three years between male and female infants (height, p = 0.81; weight p = 0.25). CONCLUSIONS Intrauterine exposure to maternal CAM contributes to accelerated postnatal weight gain in female preterm infants during the first three years.
Collapse
Affiliation(s)
- Takafumi Ushida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
- Division of Reproduction and Perinatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan.
| | - Rena Nosaka
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
- Anne Women's Clinic, Nagoya, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yumiko Kobayashi
- Data Science Division, Data Coordinating Center, Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Sho Tano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Kazuya Fuma
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Seiko Matsuo
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Kenji Imai
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Masahiro Hayakawa
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tomomi Kotani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
- Division of Reproduction and Perinatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
6
|
Yan L, Zhou Z, Chen S, Feng X, Mao J, Luo F, Zhu J, Chen X, Hu Y, Wang Y, Wu B, Du L, Wang C, Gong L, Zhu Y. Reshaping the chromatin landscape in HUVECs from small-for-gestational-age newborns. JCI Insight 2025; 10:e186812. [PMID: 40260916 PMCID: PMC12038915 DOI: 10.1172/jci.insight.186812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 03/11/2025] [Indexed: 04/24/2025] Open
Abstract
Small for gestational age (SGA), with increased risk of adult-onset cardiovascular diseases and metabolic syndromes, is known to associate with endothelial dysfunction, but the pathogenic mechanisms remain unclear. In this study, the pathological state of human umbilical vein endothelial cells (HUVECs) from SGA individuals was characterized by presenting increased angiogenesis, migration, proliferation, and wound healing ability relative to their normal counterparts. Genome-wide mapping of transcriptomes and open chromatins unveiled global gene expression alterations and chromatin remodeling in SGA-HUVECs. Specifically, we revealed increased chromatin accessibility at active enhancers, along with dysregulation of genes associated with angiogenesis, and further identified CD44 as the key gene driving HUVECs' dysfunction by regulating pro-angiogenic genes' expression and activating phosphorylated ERK1/2 and phosphorylated endothelial NOS expression in SGA. In SGA-HUVECs, CD44 was abnormally upregulated by 3 active enhancers that displayed increased chromatin accessibility and interacted with CD44 promoter. Subsequent motif analysis uncovered activating protein-1 (AP-1) as a crucial transcription factor regulating CD44 expression by binding to CD44 promoter and associated enhancers. Enhancers CRISPR interference and AP-1 inhibition restored CD44 expression and alleviated the hyperangiogenesis of SGA-HUVECs. Together, our study provides a foundational understanding of the epigenetic alterations driving pathological angiogenesis and offers potential therapeutic insights into addressing endothelial dysfunction in SGA.
Collapse
Affiliation(s)
- Lingling Yan
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | | | - Xin Feng
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junwen Mao
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fang Luo
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianfang Zhu
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiuying Chen
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Yingying Hu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Yuan Wang
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bingbing Wu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Lizhong Du
- Department of Pediatrics, Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Chunlin Wang
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liang Gong
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | | |
Collapse
|
7
|
Price SAL, Koye DN, Lewin A, Nankervis A, Kane SC. Maternal Metabolic Health and Mother and Baby Health Outcomes (MAMBO): Protocol of a Prospective Observational Study. JMIR Res Protoc 2025; 14:e72542. [PMID: 40215105 PMCID: PMC12032496 DOI: 10.2196/72542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 02/24/2025] [Accepted: 03/02/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Metabolic disease is increasingly impacting women of reproductive age. In pregnancy, uncontrolled metabolic disease can result in offspring with major congenital anomalies, preterm birth, and abnormal fetal growth. Pregnancy also accelerates the complications of metabolic diseases in mothers resulting in an increased risk of premature cardiovascular events. Despite the convincing evidence that preconception care can largely mitigate the risks of metabolic disease in pregnancy, there are few data about how to identify the highest-risk women so that they can be connected with appropriate preconception care services. OBJECTIVE The aim of the study is to determine the maternal phenotype that represents the highest risk of having adverse neonatal and maternal pregnancy outcomes. METHODS This will be a prospective cohort study of 500 women recruited in early pregnancy. The primary outcome is a composite of offspring born small for gestational age (SGA) or large for gestational age (LGA) (customized birthweight ≤10th and ≥90th centile for gestational age). Secondary outcomes are (1) composite of adverse neonatal birth outcomes (SGA, LGA, major congenital abnormalities, preterm birth [<37 weeks' gestation]) and (2) composite of new maternal metabolic outcomes (gestational diabetes, diabetes in pregnancy, type 2 diabetes [T2D] or prediabetes; gestational hypertension, preeclampsia, eclampsia or new essential hypertension after pregnancy; and gestational weight gain ≥20kg or new overweight/obesity at the 12-18 months postpartum visit). A multivariable logistic regression analysis will be conducted to identify candidate predictors of poor pregnancy outcomes due to metabolic disease. From this model, model coefficients and the associated 95% CIs will be extracted to derive the risk score for predicting the delivery of LGA/SGA offspring (primary outcome) and composites of adverse neonatal outcomes and maternal outcomes (secondary outcomes). RESULTS Seed funding for the project was acquired in November 2022 and subsequent funding was acquired in May 2024. The first participant was recruited on March 23, 2023. At the time of manuscript submission, 402 participants have been recruited. Data analysis has not yet been performed. Results are expected to be published in the first half of 2027. CONCLUSIONS This is a prospective observational cohort study that intends to identify the metabolic disease risk factors, or combination of factors, that are most likely to cause adverse maternal and fetal health outcomes. These characteristics will be used to develop a risk calculator which will assist in identifying the highest risk women and in triaging them to appropriate services. The study has been approved by the institutional Human Research Ethics Committee (HREC/90080/MH-2022). TRIAL REGISTRATION Australian New Zealand Clinical Trials Registry ACTRN12623000037606; https://tinyurl.com/yeytsxtp. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/72542.
Collapse
Affiliation(s)
- Sarah A L Price
- Department of Medicine, University of Melbourne, Melbourne, Australia
- Department of Obstetric Medicine, Royal Women's Hospital, Melbourne, Australia
- Department of Diabetes and Endocrinology, The Royal Melbourne Hospital, Melbourne, Australia
| | - Digsu N Koye
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Alice Lewin
- Department of Obstetric Medicine, Royal Women's Hospital, Melbourne, Australia
| | - Alison Nankervis
- Department of Medicine, University of Melbourne, Melbourne, Australia
- Department of Obstetric Medicine, Royal Women's Hospital, Melbourne, Australia
- Department of Diabetes and Endocrinology, The Royal Melbourne Hospital, Melbourne, Australia
| | - Stefan C Kane
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Melbourne, Australia
- Maternity Services, Royal Women's Hospital, Melbourne, Australia
| |
Collapse
|
8
|
Ribeiro IT, Fioretto MN, Dos Santos SAA, Alvarez MVN, Portela LMF, Mattos R, Sebastian HB, Vitali PM, Seiva FRF, Barbisan LF, Lima CAH, Damasceno DC, Zambrano E, Justulin LA. Maternal protein restriction and postnatal sugar consumption increases inflammatory response and deregulates metabolic pathways in the liver of male offspring rats with aging. Mol Cell Endocrinol 2025; 599:112484. [PMID: 39900277 DOI: 10.1016/j.mce.2025.112484] [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] [Received: 11/18/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/05/2025]
Abstract
This study investigated the late effects of maternal protein restriction (MPR) and early postnatal sugar consumption on liver health in male Sprague-Dawley rat offspring, focusing on changes observed throughout the aging process. The animals were divided into the following groups: Control (CTR): Male offspring whose dams consumed a normal protein diet (NPD, 17% protein) and water ad libitum during gestation and lactation, and then fed a NPD and water until PND 540; Control + Sugar (CTR + SUG): The same treatment as CTR, but consuming a sugar solution (10% diluted in water) from postnatal day (PND) 21-90, and then fed a NPD and water until PND 540; Gestational and Lactational Low Protein (GLLP): Male offspring whose dams consumed a low-protein diet (LPD, 6% protein) during gestation and lactation and, then fed a NPD and water ad libitum until PND 540; Gestational and Lactational Low Protein + Sugar (GLLP + SUG): male offspring whose dams consumed a LPD during gestation and lactation, and then fed a NPD and a sugar solution (10% diluted in water) ad libitum from PND 21 to 90. On PND 540, the animals were anesthetized, weighed, and euthanized, and their livers were collected for morphological and molecular analyses. The GLLP and GLLP + SUG groups showed lower body weight and lower retroperitoneal fat weight compared to the CTR and CTR + SUG groups. Morphological analysis revealed inflammatory foci in the liver from the CTR + SUG, GLLP, and GLLP + SUG groups, compared to the CTR group. Hepatic activities of CAT, SOD, and GSH-Px were increased in the GLLP + SUG group and decreased in the GLLP group, compared to the CTR group. Immunohistochemistry showed a significant increase in occupied area per foci de hepatocytes positive for GSTpi (placental form) in the CTR + SUG, GLLP, and GLLP + SUG groups, compared to the CTR group. Proteomic analysis of the groups revealed significant changes in hepatic metabolic and inflammatory pathways. In the CTR + SUG group, upregulated pathways associated with non-alcoholic fatty liver disease (NAFLD) and downregulated pathways related to autophagy were observed. In the GLLP and GLLP + SUG groups, there was a significant impact on metabolic pathways, including glucose metabolism, gluconeogenesis, glycogenesis, and cellular stress responses. An upregulation of pathways associated with chemokine- and cytokine-mediated inflammatory processes was also identified, indicating activation of the immune system in the liver during aging. Therefore, MPR, with or without postnatal sugar consumption, resulted in hepatic changes in metabolism and the antioxidant defense in old male offspring.
Collapse
Affiliation(s)
- Isabelle Tenori Ribeiro
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil.
| | - Matheus Naia Fioretto
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Sérgio Alexandre Alcantara Dos Santos
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil; Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | - Luiz Marcos Frediani Portela
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Renato Mattos
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Hecttor Baptista Sebastian
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Pedro Menchini Vitali
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Fábio Rodrigues Ferreira Seiva
- Department of Chemical and Biological Sciences, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luís Fernando Barbisan
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Clélia Akiko Hiruma Lima
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Débora Cristina Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics (UNIPEX), Course of Postgraduate on Tocogynecology, Botucatu Medical School, Sao Paulo State University, Botucatu, SP, Brazil
| | - Elena Zambrano
- Department Reproductive Biology, Salvador Zubirán National Institute of Medical Sciences and Nutrition, Mexico City, Mexico; Facultad de Química, Universidad Nacional Autónoma de, Mexico
| | - Luis Antonio Justulin
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil.
| |
Collapse
|
9
|
Borden CG, Shabanova V, Taylor SN, Buck CO. Alterations in infant adipokine concentrations in the first postnatal week with exposure to diabetes in pregnancy. J Perinatol 2025:10.1038/s41372-025-02269-2. [PMID: 40121331 DOI: 10.1038/s41372-025-02269-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/13/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
OBJECTIVE Characterize changes in neonatal energy metabolism hormones in first postnatal week. STUDY DESIGN Concentrations of leptin, adiponectin, insulin, resistin, and ghrelin were measured in cord and infant blood collected in the first postnatal week in a prospective cohort of term and preterm infants. Change over time in each hormone was modeled using linear mixed effects regression. RESULT Among 106 infants, 65 (61%) were preterm and 44 (42%) were exposed to diabetes (DM) in pregnancy. DM group had higher leptin [between group difference β 2.5 (95%CI: 1.72-3.70)] and resistin [β 1.5 (95%CI:1.14, 1.92)] and lower ghrelin [β 0.49] [95%CI: (0.32-0.76)] versus non-DM group. Preterm infants had lower adiponectin [β 0.80 (95%CI: 0.67-0.96)] versus term group. Insulin varied by DM and prematurity (interaction-term p value < 0.05). CONCLUSION In this cohort, hormone concentrations varied by DM and prematurity. Early alterations in energy metabolism hormones may reflect changes in developmental programming which persist in the early postnatal period.
Collapse
Affiliation(s)
- Caroline G Borden
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | | | - Sarah N Taylor
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Catherine O Buck
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
10
|
Cheng J, Jin H, Zhang Y, Ren J, Huang K, Tong J, Gan H, Lv J, Wang Q, Tao F, Zhu Y. Small for gestational age children at risk: Identifying placenta-brain axis genes as biomarkers for early prediction of neurodevelopmental delay. Life Sci 2025; 365:123450. [PMID: 39922426 DOI: 10.1016/j.lfs.2025.123450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/26/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
AIMS Small for gestational age (SGA) is a prevalent issue in global public health. The relationship between SGA and neurodevelopmental delay remains a topic of debate and the exploration of potential biomarkers is crucial. The identification of placental-brain axis genes offers novel perspectives for anticipating neurodevelopmental delay. MAIN METHODS First, we utilized multiple logistic regression to assess Ages and Stages Questionnaire of China (ASQ-C) scores in children at 6 months, 18 months, and 48 months of age. Next, we analyzed the placental transcriptome data from SGA and appropriate for gestational age (AGA) children in the Ma'anshan Birth Cohort (MABC) and validated it through Real-time quantitative PCR (RT-qPCR). Finally, we combined the experimental data with clinical data to establish a predictive model. KEY FINDINGS SGA children were found to have a higher risk of neurodevelopmental delay at 6 months and 18 months of age. Further experimental validation found that decreased RPS27A gene expression was associated with developmental delay in solving-problem and personal-social domain at 6 months of age in SGA children. SIGNIFICANCE Our study focused on the neurodevelopmental results of children from three time points, analyzed the mechanism of neurodevelopmental delay in SGA from the perspective of placenta-brain axis, and conducted experimental verification of the selected biomarkers. Therefore, our study has certain novelty and persuasive, providing new insights for early detection of neurodevelopmental delay in children with SGA.
Collapse
Affiliation(s)
- Jingjing Cheng
- Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Department of Maternal & Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Heyue Jin
- Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Department of Maternal & Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Yimin Zhang
- Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Department of Maternal & Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Jiawen Ren
- Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Department of Maternal & Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Kun Huang
- Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Department of Maternal & Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Juan Tong
- Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Department of Maternal & Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Hong Gan
- Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Department of Maternal & Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Jia Lv
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Qu'nan Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, China.
| | - Fangbiao Tao
- Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Department of Maternal & Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China.
| | - Yumin Zhu
- Medical School, Nanjing University, Nanjing, Jiangsu, China.
| |
Collapse
|
11
|
Lv L, Yang J, Li L, Huang C, Shi H, Fang Y, Zuo L, Liu T, Duan H, Wen J, Yang Q, Henry A, Han C, Yin A, Zhou X. The interdependence of mid-trimester blood pressure and glucose levels in shaping fetal growth and neonatal outcomes: implications for risk-benefit assessment and co-management. BMC Med 2025; 23:161. [PMID: 40087732 PMCID: PMC11909891 DOI: 10.1186/s12916-025-03990-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Maternal hypertension and hyperglycemia are closely related but have distinct impacts on fetal growth and are managed independently. How the interdependence of blood pressure (BP) and glucose levels quantitatively influences risk patterns for abnormal fetal growth and neonatal complications remains unexplored. METHODS Maternal BP and fasting plasma glucose (FPG) levels were measured between 20 and 28 weeks of gestation in a cohort including 56,881 singleton pregnancies. Linear and quantile regression analyses were used to evaluate the relationship between BP and FPG. We examined the dose-response relationships between BP and FPG with small-for-gestational age (SGA) and large-for-gestational age (LGA) by using restricted cubic spline (RCS) curves. Additionally, multivariable fractional polynomial interaction (MFPI) analysis was conducted to assess the effects of higher versus lower BP levels across the full range of FPG levels. Heatmaps were created to visualize the contributions of BP and FPG by categorizing them into ordered groups. RESULTS Quantile regression revealed consistent positive correlations between mean arterial pressure (MAP) and FPG, with a steeper increase in MAP coefficients above the 0.5 quantile of FPG. MAP had a non-linear positive association with SGA risk, while FPG showed a non-linear negative association. Heatmaps revealed the highest SGA risk with high BP (MAP ≥ 85 mmHg)/low glucose (< 85 mg/dL) combinations and the lowest risk with low BP (MAP < 85 mmHg)/high glucose (≥ 85 mg/dL), with equivalent risk at both high BP/high glucose and low BP/low glucose. In hypertensive patients, SGA risk worsened continuously as glucose levels decreased. LGA risk was not influenced by BP levels. Neonatal complications decreased by approximately 47% as MAP declined from the highest to lowest category, and by about 17% with decreasing glucose levels. CONCLUSIONS Based on a large pregnancy cohort in China, this study revealed an interdependent association between maternal BP and glucose levels and their combined impact on the risk of SGA. It provided quantitative evidence of how this interdependence shapes the transition of risk patterns for SGA, neonatal complications, and LGA. These findings underscore the need for an integrated approach to co-managing BP and glucose levels during pregnancy.
Collapse
Affiliation(s)
- Lijuan Lv
- Medical Genetic Center, Department of Obstetrics, Guangdong Women and Children Hospital, Xinnan Avenue, Panyu District, Guangzhou, 511442, China
| | - Jingbo Yang
- Department of Cardiology, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Linjie Li
- Department of Cardiology, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Chuanyi Huang
- Department of Cardiology, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Huihua Shi
- Department of Cardiology, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Yiwen Fang
- Department of Cardiology, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Lushu Zuo
- Department of Cardiology, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Ting Liu
- Department of Cardiology, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Hongli Duan
- Medical Genetic Center, Department of Obstetrics, Guangdong Women and Children Hospital, Xinnan Avenue, Panyu District, Guangzhou, 511442, China
| | - Jiying Wen
- Medical Genetic Center, Department of Obstetrics, Guangdong Women and Children Hospital, Xinnan Avenue, Panyu District, Guangzhou, 511442, China
| | - Qing Yang
- Department of Cardiology, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Amanda Henry
- Discipline of Women'S Health, School of Clinical Medicine, UNSW Medicine and Health, University of New South Wales, Sydney, Australia
- Department of Women'S and Children'S Health, St George Hospital, Sydney, Australia
- The George Institute for Global Health, UNSW Medicine and Health, Sydney, Australia
| | - Cha Han
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China.
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China.
| | - Aihua Yin
- Medical Genetic Center, Department of Obstetrics, Guangdong Women and Children Hospital, Xinnan Avenue, Panyu District, Guangzhou, 511442, China.
| | - Xin Zhou
- Department of Cardiology, Tianjin Medical University General Hospital, 154, Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
12
|
Li X, Hegarty K, Lin F, Chang JL, Abdalla A, Dhanabalan K, Solomevich SO, Song W, Roder K, Yao C, Lu W, Carmeliet P, Choudhary G, Dennery PA, Yao H. Endothelial Cpt1a Inhibits Neonatal Hyperoxia-Induced Pulmonary Vascular Remodeling by Repressing Endothelial-Mesenchymal Transition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415824. [PMID: 39799584 PMCID: PMC11923872 DOI: 10.1002/advs.202415824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/15/2024] [Indexed: 01/15/2025]
Abstract
Pulmonary hypertension (PH) increases the mortality of preterm infants with bronchopulmonary dysplasia (BPD). There are no curative therapies for this disease. Lung endothelial carnitine palmitoyltransferase 1a (Cpt1a), the rate-limiting enzyme of the carnitine shuttle system, is reduced in a rodent model of BPD. It is unknown whether endothelial Cpt1a reduction causes pulmonary vascular (PV) remodeling. The latter can be the result of endothelial-mesenchymal transition (EndoMT). Here, endothelial cell (EC)-specific Cpt1a KO and WT mice (<12 h old) are exposed to hyperoxia (70% O2) for 14 days and allow them to recover in normoxia until postnatal day 28. Hyperoxia causes PH, which is aggravated in EC-specific Cpt1a KO mice. Upregulating endothelial Cpt1a expression inhibits hyperoxia-induced PV remodeling. Hyperoxia causes lung EndoMT, detected by immunofluorescence, scRNA-sequencing, and EC lineage tracing, which is further increased in EC-specific Cpt1a KO mice. Blocking EndoMT inhibits hyperoxia-induced PV remodeling. Male mice under the same high oxygen conditions develop a higher degree of PH than females, which is associated with reduced endothelial Cpt1a expression. Conclusively, neonatal hyperoxia causes PH by decreasing endothelial Cpt1a expression and upregulating EndoMT. This provides a valuable strategy for developing targeted therapies by upregulating endothelial Cpt1a levels or inhibiting EndoMT to treat BPD-associated PH.
Collapse
Affiliation(s)
- Xiaoyun Li
- Department of Molecular BiologyCellular Biology, and BiochemistryBrown UniversityProvidenceRI02912USA
- Providence VA Medical CenterProvidenceRI02908USA
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
- College of PharmacyJinan UniversityGuangzhouGuangdong510632China
| | - Katy Hegarty
- Department of Molecular BiologyCellular Biology, and BiochemistryBrown UniversityProvidenceRI02912USA
| | - Fanjie Lin
- State Key Laboratory of Respiratory DiseaseGuangdong Key Laboratory of Vascular DiseaseNational Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdong510120China
| | - Jason L. Chang
- Department of Molecular BiologyCellular Biology, and BiochemistryBrown UniversityProvidenceRI02912USA
| | - Amro Abdalla
- Providence VA Medical CenterProvidenceRI02908USA
| | - Karthik Dhanabalan
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Sergey O. Solomevich
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Wenliang Song
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Karim Roder
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Chenrui Yao
- College of Arts & SciencesBoston UniversityBostonMA02215USA
| | - Wenju Lu
- State Key Laboratory of Respiratory DiseaseGuangdong Key Laboratory of Vascular DiseaseNational Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdong510120China
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular MetabolismDepartment of Oncology and Leuven Cancer InstituteKU LeuvenVIB Center for Cancer Biology, VIBLeuvenBrussels3000Belgium
- Center for BiotechnologyKhalifa UniversityAbu Dhabi127788UAE
| | - Gaurav Choudhary
- Providence VA Medical CenterProvidenceRI02908USA
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Phyllis A. Dennery
- Department of Molecular BiologyCellular Biology, and BiochemistryBrown UniversityProvidenceRI02912USA
- Department of PediatricsWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| | - Hongwei Yao
- Department of Molecular BiologyCellular Biology, and BiochemistryBrown UniversityProvidenceRI02912USA
- Providence VA Medical CenterProvidenceRI02908USA
- Division of CardiologyDepartment of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRI02903USA
| |
Collapse
|
13
|
Dos Santos IBL, Fioretto MN, Jorge MS, Barata LA, Ribeiro IT, Franzolin AML, Stoppa EG, Mattos R, Portela LMF, Emílio Silva MT, Dos Santos SAA, de Arruda Miranda JR, Hiruma Lima CA, Justulin LA. Maternal protein restriction impairs intestinal morphophysiology and antioxidant system in young male offspring rats. Exp Cell Res 2025; 446:114464. [PMID: 39986598 DOI: 10.1016/j.yexcr.2025.114464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/10/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025]
Abstract
The developmental origins of health and disease (DOHaD) concept suggests that adverse conditions during gestation can influence the development and function of multiple organs, including the gastrointestinal tract. Maternal protein restriction (MPR) exposure has been associated with negative effects on reproduction, the endocrine system, and liver metabolic health. However, limited research has explored the impact of MPR on the offspring's intestinal morphophysiology. This study investigated the effects of gestational and lactational MPR on the duodenum and colon of young male offspring rats at postnatal (PND)21. We hypothesize that MPR affects intestinal morphophysiology and development early in life. Our findings revealed tachygastria in offspring exposed to MPR. The ultrastructural analysis uncovered a reduction in goblet cell numbers and changes in collagen deposition in the duodenum and colon. We also identified imbalances in inflammatory markers (IL-6 and TGF-β1) and antioxidant enzymes (CAT and SOD). These results demonstrate that MPR significantly affects gastrointestinal morphophysiology early in life by disrupting gastric motility and altering duodenal and colonic histoarchitecture, antioxidant defense, and inflammatory pathways. Such alterations may predispose the descendants to long-term gastrointestinal disorders, underscoring the importance of further research on the developmental origins of intestinal health and disease.
Collapse
Affiliation(s)
| | - Matheus Naia Fioretto
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Miguel Silingardi Jorge
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luísa Annibal Barata
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Isabelle Tenori Ribeiro
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | | | - Erick Guilherme Stoppa
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Renato Mattos
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luiz Marcos Frediane Portela
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Maycon Tavares Emílio Silva
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Sérgio Alexandre Alcântara Dos Santos
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil; Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | - Clélia Akiko Hiruma Lima
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luis Antonio Justulin
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil.
| |
Collapse
|
14
|
Su YY, Chen CJ, Chen MH, Chang H, Chen CM, Lin HC, Guo YL, Hsieh WS, Chen PC. Long-term effects on growth in preterm and small for gestational age infants: A national birth cohort study. Pediatr Neonatol 2025; 66:168-175. [PMID: 39107217 DOI: 10.1016/j.pedneo.2024.06.007] [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] [Received: 10/16/2023] [Revised: 01/29/2024] [Accepted: 06/25/2024] [Indexed: 08/09/2024] Open
Abstract
BACKGROUND Premature and small-for-gestational-age (SGA) infants tend to have long-term growth morbidities such as short stature, failure to thrive, and obesity. Although most of these infants show catch-up growth at 2-4 years of age, they are still more susceptible to childhood obesity and related metabolic disorders. Those who fail to achieve catch-up will suffer from pathological short stature and neurodevelopmental impairment through adulthood. This study aims to depict the growth pattern of premature or SGA infants and their growth morbidities in Taiwan. METHODS Data were obtained from a nationally representative cohort of 24,200 pairs of postpartum women and newborns in the Taiwan Birth Cohort Study (TBCS), using structured questionnaire interviews. A total of 16,358 infants were included and three follow-up surveys were completed at 6, 18, and 36 months after the deliveries. We constructed growth curves to conduct an in-depth investigation into anthropometric data, applying a linear mixed model. Logistic regression was used to model the relevant outcomes, with adjustment for various potential confounding factors. RESULTS Despite being born shorter and lighter, preterm and SGA infants generally showed catch-up growth and had no higher odds ratios (ORs) of developing short stature or failure to thrive compared to appropriate-for-gestational-age (AGA) term infants before 3 years of age. Preterm SGA infants, particularly females, had higher ORs for obesity at the 36-month follow-up. CONCLUSION This is the first nationwide population-based study depicting the growth of SGA infants in Taiwan. The growth patterns of preterm and term SGA infants are different from those of preterm and term AGA infants. Further research is necessary to understand the growth trajectories of preterm and SGA infants and their associations with later diseases.
Collapse
Affiliation(s)
- Yi-Yu Su
- Institute of Environmental and Occupational Health Sciences, National Taiwan University, College of Public Health, Taipei, Taiwan; Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chi-Jen Chen
- Institute of Epidemiology and Preventive Medicine, National Taiwan University College of Public Health, Taipei, Taiwan
| | - Mei-Huei Chen
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan; Department of Pediatrics, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Hsi Chang
- Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chung-Ming Chen
- Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiu-Chen Lin
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Clinical Pathology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yueliang Leon Guo
- Institute of Environmental and Occupational Health Sciences, National Taiwan University, College of Public Health, Taipei, Taiwan; Department of Environmental and Occupational Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan; National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Wu-Shiun Hsieh
- Department of Pediatrics, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan; Department of Pediatrics, Cathay General Hospital, Taipei, Taiwan
| | - Pau-Chung Chen
- Institute of Environmental and Occupational Health Sciences, National Taiwan University, College of Public Health, Taipei, Taiwan; Department of Environmental and Occupational Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan; National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan; Department of Public Health, National Taiwan University College of Public Health, Taipei, Taiwan.
| |
Collapse
|
15
|
Bizerea-Moga TO, Moga TV, Stroescu R, Chisavu L, Mărginean O, Chisavu F. Associations Between Birth Characteristics, Weaning Practices, and the Metabolic Syndrome in Children: A Descriptive Study. Metabolites 2025; 15:148. [PMID: 40137113 PMCID: PMC11943707 DOI: 10.3390/metabo15030148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Childhood obesity has seen an important rise in recent decades, in both the pediatric and adult populations. Excess weight can cause various health complications, such as the metabolic syndrome (MetS), a cluster of medical conditions linked to adverse cardiometabolic outcomes. Although MetS may be attributed mainly to adults, early life factors, such as birth characteristics and feeding practices, may influence its development in obese children. Aim: This study aims to investigate the relationships between birth metrics, early feeding practices, and the prevalence of MetS and its components among obese children. Methods: A retrospective observational study was conducted on 800 obese patients aged 0-18 years, admitted to the "Louis Țurcanu" Children's Clinical and Emergency Hospital in Timișoara, Romania, from 1 January 2013 to 31 December 2023. Patients were divided according to gestational age: small for gestational age (SGA), appropriate for gestational age (AGA), and large for gestational age (LGA). Results: Type 2 diabetes (18.2%), hypercholesterolemia (24.6%), IR (41.3%), and MetS (39.2%) were more prevalent among oSGA patients included in the study. These patients were breastfed for longer periods but weaned at a younger age. oLGA patients had the highest BMI values (28.4 ± 4.2) and, in this study group, hypertriglyceridemia (29.4%), arterial hypertension (26.8%), and lower HDL-C (41.7 ± 6.3 mg/dL) were more prevalent. The incidence of MetS increased with age (12.6 ± 3.1 years). Among these patients, IR (52.3%) was more prevalent. The introduction of flour-based energy-dense foods before six months was more frequent in MetS patients, but not statistically significant. Logistic regression showed oSGA patients had a 4.49-fold higher MetS risk (p < 0.001). Older age at diagnosis increased the risk of developing MetS by 37%, a diagnosis of impaired glucose tolerance by 19-fold, and a family history of diabetes by 2.7-fold. ROC analysis showed strong predictability (AUC = 0.905, sensitivity = 82%, specificity = 88%). Conclusions: Obese children born SGA had a higher risk for developing MetS. The incidence of MetS and its components increases with age among obese patients. Monitoring growth patterns and dietary habits in early life is paramount to mitigate future metabolic complications.
Collapse
Affiliation(s)
- Teofana Otilia Bizerea-Moga
- Department XI of Pediatrics-1st Pediatric Discipline, Center for Research on Growth and Developmental Disorders in Children, ‘Victor Babeș’ University of Medicine and Pharmacy Timișoara, Eftimie Murgu Sq No. 2, 300041 Timișoara, Romania; (T.O.B.-M.); (R.S.); (O.M.)
- 1st Pediatric Clinic from ‘Louis Țurcanu’ Children’s Clinical and Emergency Hospital, Iosif Nemoianu 2, 300011 Timișoara, Romania
| | - Tudor Voicu Moga
- Department VII of Internal Medicine-Gastroenterology Discipline, Advanced Regional Research Center in Gastroenterology and Hepatology, ‘Victor Babeș’ University of Medicine and Pharmacy Timișoara, Eftimie Murgu Sq No. 2, 300041 Timișoara, Romania;
- Gastroenterology and Hepatology Clinic from ‘Pius Brînzeu’ County Emergency Clinical Hospital, Liviu Rebreanu 156, 300723 Timișoara, Romania
| | - Ramona Stroescu
- Department XI of Pediatrics-1st Pediatric Discipline, Center for Research on Growth and Developmental Disorders in Children, ‘Victor Babeș’ University of Medicine and Pharmacy Timișoara, Eftimie Murgu Sq No. 2, 300041 Timișoara, Romania; (T.O.B.-M.); (R.S.); (O.M.)
- 4th Pediatric Clinic from ‘Louis Țurcanu’ Children’s Clinical and Emergency Hospital, Iosif Nemoianu 2, 300011 Timișoara, Romania;
| | - Lazar Chisavu
- Centre for Molecular Research in Nephrology and Vascular Disease from “Victor Babes” University of Medicine and Pharmacy-Faculty of Medicine, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Nephrology Discipline, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Otilia Mărginean
- Department XI of Pediatrics-1st Pediatric Discipline, Center for Research on Growth and Developmental Disorders in Children, ‘Victor Babeș’ University of Medicine and Pharmacy Timișoara, Eftimie Murgu Sq No. 2, 300041 Timișoara, Romania; (T.O.B.-M.); (R.S.); (O.M.)
- 1st Pediatric Clinic from ‘Louis Țurcanu’ Children’s Clinical and Emergency Hospital, Iosif Nemoianu 2, 300011 Timișoara, Romania
| | - Flavia Chisavu
- 4th Pediatric Clinic from ‘Louis Țurcanu’ Children’s Clinical and Emergency Hospital, Iosif Nemoianu 2, 300011 Timișoara, Romania;
- Centre for Molecular Research in Nephrology and Vascular Disease from “Victor Babes” University of Medicine and Pharmacy-Faculty of Medicine, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| |
Collapse
|
16
|
Luo X, Hou L, Zhong Y, Zhao S, Chen X, Dong Q, Du H, Lu H, Yang Y, Wu X, Luo F, Chen R, Xu Z, Ma Y, Song W, Feng M, Gu X, Qiu W. A Phase 2 Study of PEGylated Recombinant Human Growth Hormone for 52 Weeks in Short Children Born Small for Gestational Age in China. Clin Endocrinol (Oxf) 2025; 102:136-146. [PMID: 39513569 DOI: 10.1111/cen.15156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 11/15/2024]
Abstract
OBJECTIVE Children born small for gestational age (SGA) are at increased risk of health issues. This study evaluated the efficacy, safety and optimal dose of PEGylated-recombinant human growth hormone (PEG-rhGH) in these children. DESIGN In this multicentre, randomised, open-label, Phase 2 trial conducted at nine clinical sites in China, patients were randomised 1:1 to receive subcutaneous injections of PEG-rhGH at 0.1 mg/kg/week (low dose) or 0.2 mg/kg/week (high dose) for 52 weeks. PATIENTS Ninety-six children were born SGA. MEASUREMENTS The primary endpoint was the change in height standard deviation score (HT-SDS) at Week 52. RESULTS At Week 52, the change in HT-SDS in the high- and low-dose groups was 0.923 ± 0.352 (p < 0.0001) and 0.511 ± 0.336 (p < 0.0001), respectively (least-squares means difference, 0.410; 95% confidence interval 0.270-0.551; p < 0.0001). Height velocity (9.94 ± 1.55 vs. 8.37 ± 1.50 cm/year) was also significantly higher in the high-dose than in the low-dose group (p < 0.0001). Change in insulin-like growth factor (IGF)-1 SDS was 1.867 ± 1.747 and 1.168 ± 1.193 in the high- and low-dose groups, respectively (p = 0.0189). IGF-1/IGF binding protein-3 and bone maturity were improved in both groups at Week 52. Most treatment-emergent adverse events were mild to moderate; the safety profile was similar in both groups. CONCLUSIONS PEG-rhGH at either dose for 52 weeks was effective and well tolerated in children born SGA. Patients in the high-dose group achieved greater improvement in HT-SDS than in the low-dose group. TRIAL REGISTRATION ClinicalTrials. gov identifier: NCT02375620.
Collapse
Affiliation(s)
- Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Hou
- Department of Pediatrics, Tongji Hospital, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Zhong
- Department of Child Health Care, Hunan Children's Hospital, Hunan, China
| | - Sha Zhao
- Department of Child Health Care, Hunan Children's Hospital, Hunan, China
| | - Xiaobo Chen
- Department of Endocrinology, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Qian Dong
- Department of Endocrinology, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Hongwei Du
- Department of Paediatrics, The First Hospital of Jilin University, Jilin, China
| | - Honghua Lu
- Department of Paediatrics, The First Hospital of Jilin University, Jilin, China
| | - Yu Yang
- Department of Endocrinology and Genetics, Jiangxi Provincial Children's Hospital, Jiangxi, China
| | - Xian Wu
- Department of Endocrinology and Genetics, Jiangxi Provincial Children's Hospital, Jiangxi, China
| | - Feihong Luo
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Ruoqian Chen
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Zhuangjian Xu
- Department of Pediatrics, Wuxi Fourth People's Hospital, Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Yaping Ma
- Department of Pediatrics, Wuxi Fourth People's Hospital, Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Wenhui Song
- Department of Endocrinology, Children's Hospital of Shanxi, Women's Health Center of Shanxi, Shanxi, China
| | - Mei Feng
- Department of Endocrinology, Children's Hospital of Shanxi, Women's Health Center of Shanxi, Shanxi, China
| | - Xuefan Gu
- Department of Pediatric Endocrinology and Genetics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjuan Qiu
- Department of Pediatric Endocrinology and Genetics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Zhang B, Zhan Z, Xi S, Zhang Y, Yuan X. Alkaline phosphatase of late pregnancy promotes the prediction of adverse birth outcomes. J Glob Health 2025; 15:04028. [PMID: 39849974 PMCID: PMC11758466 DOI: 10.7189/jogh.15.04028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2025] Open
Abstract
Background Adverse birth outcomes (ABO), such as preterm birth (PTB), small and large for gestational age (SGA/LGA), can compromise both the short- and long-term health of mothers and their foetuses. The purpose of this observational study was to investigate the association between maternal serum alkaline phosphatase (ALP) levels in late pregnancy and the risk of ABO, and to evaluate its predictive value of maternal ALP levels for ABO in women with singleton pregnancies. Methods A total of 11 853 consecutive pregnant women underwent hepatic and renal function tests, lipid profile assessments, ALP and high-sensitivity C-reactive protein levels measurements upon admission for labour. Their clinical perinatal parameters and outcomes were also analysed. Results The prevalence of PTB, SGA, and LGA in this study was 7.2% (n = 849), 8.9% (n = 1053), and 15.6% (n = 1844), respectively. With increasing quartiles of maternal serum ALP levels, the foetal gestational age increased by 0.58 weeks (95% confidence interval (CI) = 0.50-0.66), 0.78 weeks (95% CI = 0.70-0.86), and 0.98 weeks (95% CI = 0.90-1.06), respectively, and the birth weight increased by 62.91 g (95% CI = 43.96-81.86), 91.54 g (95% CI = 72.41-110.67), and 117.92 g (95% CI = 98.18-137.67), respectively. Compared to women in the bottom quartile of ALP, those in the top quartile had a lower risk of PTB (adjusted odds ratio (OR) = 0.14; 95% CI = 0.11-0.18), a lower risk of SGA (adjusted OR = 0.65; 95% CI = 0.53-0.80), and a higher risk of LGA (adjusted OR = 1.92; 95% CI = 1.62-2.28). Sensitivity analyses conducted among individuals without advanced maternal age, obesity, multiparity, pregnancy complications, and PTB (for SGA/LGA) validated the consistency of these results. More importantly, adding ALP to the established model significantly increased the area under the curve (AUC) for predicting adverse birth outcomes: for PTB, the AUC increased from 0.761 to 0.809 (P < 0.001); for SGA, it increased from 0.754 to 0.759 (P = 0.014); and for LGA, it increased from 0.750 to 0.755 (P < 0.001). Conclusions Maternal serum ALP levels in late pregnancy are significantly associated with the risk of ABO. When combined with clinical characteristics and routine laboratory results, ALP has incremental predictive value for ABO, particularly for PTB.
Collapse
|
18
|
Sompolinsky Y, Lipschuetz M, Cohen-Cymberknoh M, Cohen SM, Kabiri D, Walfisch A, Yagel S, Gordon S, Haklai Z, Applbaum Y. Early childhood respiratory morbidity according to gestational age at birth: A nationwide cohort study. Respir Med 2025; 236:107913. [PMID: 39689734 DOI: 10.1016/j.rmed.2024.107913] [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] [Received: 06/24/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Preterm birth survivors are at risk for short- and long-term respiratory morbidity. This includes increased rates of chronic obstructive pulmonary disease and infectious morbidity. Previous studies showed increased utilization of healthcare services throughout early childhood. However, only a few large-scale studies showed the effect on respiratory morbidity throughout the full spectrum of gestational age at birth. The aim of this study was to show the healthcare burden associated with prematurity, in a large nationwide cohort. STUDY DESIGN Data regarding gestational age at birth, month and year of birth, and infant sex were gathered for all 1,762,149 infants born in Israel between January 1, 2010, and December 31, 2019. Rates of hospitalization, length of hospitalization, and emergency department visits were calculated per 1000 live births and stratified by gestational age. Poisson regression was constructed to adjust for infant sex, year and month of birth. RESULTS Preterm birth occurred in 6.43 % of deliveries (n = 109,405). A negative association was found between gestational age at birth and respiratory morbidity. As gestational age at birth advances, rates of respiratory hospitalization decrease, and length of hospitalization shortens. This association continues even after full term is reached. CONCLUSION The short- and long-term effect of preterm birth poses a significant burden on healthcare systems globally, not only at birth or in infancy, but well into early childhood. These results are a call for action to stakeholders and professional organizations to increase efforts in preventing and treating preterm and early term labor.
Collapse
Affiliation(s)
- Yishai Sompolinsky
- Department of Obstetrics & Gynecology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Michal Lipschuetz
- Department of Obstetrics & Gynecology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel; Henrietta Szold Hadassah Hebrew University School of Nursing in the Faculty of Medicine, Jerusalem, Israel
| | - Malena Cohen-Cymberknoh
- Pediatric Pulmonary Unit and Cystic Fibrosis Center, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Sarah M Cohen
- Department of Obstetrics & Gynecology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Doron Kabiri
- Department of Obstetrics & Gynecology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Asnat Walfisch
- Department of Obstetrics and Gynecology, Rabin Medical Center, Petah-Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Simcha Yagel
- Department of Obstetrics & Gynecology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel.
| | - Shulamit Gordon
- Division of Health Information, Ministry of Health, Jerusalem, Israel
| | - Ziona Haklai
- Division of Health Information, Ministry of Health, Jerusalem, Israel
| | - Yael Applbaum
- Division of Health Information, Ministry of Health, Jerusalem, Israel
| |
Collapse
|
19
|
Zhang B, Zhan Z, Xi S, Wang F, Yuan X. Impact of serum retinol-binding protein 4 levels in late pregnancy on the incidence of small/large for gestational age infants among 11,854 pregnant women: A retrospective study. J Epidemiol 2024; 35:287-296. [PMID: 39710423 PMCID: PMC12066191 DOI: 10.2188/jea.je20240275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/01/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND There is still uncertainty regarding the association between maternal serum levels of retinol-binding protein 4 (RBP4) and pregnancy outcomes. This study aimed to determine the association between RBP4 levels and incidence of small/large for gestational age (SGA/LGA) infants among the Chinese population. METHODS This was a retrospective study of 11,854 pregnant women who delivered at Changzhou Maternal and Child Health Care Hospital between 2016 and 2017 and whose serum RBP4 levels were measured at the time of admission. The incidence of SGA/LGA deliveries was retrieved from the medical records of the participants. RESULTS Maternal RBP4 levels in the second, third and fourth quartiles (28.8-34.0, 34.1-40.0, and >40.0 mg/L, respectively) were associated with lower birthweights relative to those in the first quartile (<28.8 mg/L), with estimated average decreases of 51.30 g (95% CI: -70.51, -32.10), 86.86 g (95% CI: -106.50, -67.22) and 124.08 g (95% CI: -144.51, -103.64), respectively (P for trend <0.01). Pregnant women in the fourth quartile for RBP4 levels had a greater SGA risk (OR: 2.14, 95% CI: 1.72, 2.65) and lower LGA risk (OR: 0.53, 95% CI: 0.45, 0.63) than those in the first quartile after controlling for demographic variables, gestational age, pregnancy complications and other laboratory results. The sensitivity analysis indicated the consistency of these findings. CONCLUSION High RBP4 levels in late pregnancy are associated with an increased SGA risk and decreased LGA risk, indicating that serum RBP4 levels at the time of admission for delivery could be a promising predictor of SGA/LGA delivery.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Zhaolong Zhan
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Sijie Xi
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Feng Wang
- Department of Obstetrics and Gynecology, Yancheng Third People’s Hospital, Yancheng, China
| | - Xiaosong Yuan
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| |
Collapse
|
20
|
van Klink JMM, van Zwet EW, Lopriore E, Roest AAW, Haak MC, Slaghekke F, de Bruin C, Groene SG. Does Catch-Up Growth Come with a Cognitive Cost? Cognitive Outcome and Growth Patterns in Growth Discordant Identical Twins. J Pediatr 2024; 275:114223. [PMID: 39097263 DOI: 10.1016/j.jpeds.2024.114223] [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] [Received: 03/18/2024] [Revised: 07/17/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
OBJECTIVE To determine whether it is the magnitude of early postnatal catch-up growth (CUG) in response to fetal growth restriction (FGR) or the FGR itself that negatively impacts cognitive outcome in a model of monochorionic twins discordant for fetal growth. STUDY DESIGN This analysis is part of the LEMON study, a cohort study including all monochorionic twins with selective FGR aged 3 through 17 years. Growth measurements as documented by our primary care system were collected retrospectively. An age-appropriate neurodevelopmental test was performed generating a full-scale IQ (FSIQ). CUG at 2 years was calculated as (weight [kg] at 2 years-birth weight [kg]). We used a multivariable regression model investigating the association between FSIQ (outcome) and birth weight zscore, gestational age at birth and CUG at 2 years (predictors). Generalized estimating equations accounted for the fact that observations between cotwins are not independent. RESULTS Median age at follow-up of the 46 included twin pairs was 11 (IQR 8-13) years. Birth weight z score and gestational age at birth were significantly associated with FSIQ, with β-coefficients of 5.897 (95% CI 3.382-8.411), and 2.589 (95% CI 1.227-3.951), respectively (P < .0001). Adjusted for birth weight z score and gestational age, CUG in the first 2 years after birth was not significantly associated with FSIQ (β-coefficient 0.108 [95% CI -1.373 to 1.590], P = .886). CONCLUSIONS Our results, combining detailed growth measurements and neurodevelopmental follow-up in a discordant identical twin model, demonstrate that FGR itself rather than early postnatal CUG has negative consequences for cognitive development.
Collapse
Affiliation(s)
- Jeanine M M van Klink
- Neonatology, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik W van Zwet
- Medical Statistics, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Enrico Lopriore
- Neonatology, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Arno A W Roest
- Pediatric Cardiology, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Monique C Haak
- Fetal Therapy, Department of Obstetrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Femke Slaghekke
- Fetal Therapy, Department of Obstetrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Christiaan de Bruin
- Pediatric Endocrinology, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Sophie G Groene
- Neonatology, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
21
|
Zhang X, Zhu S, Zhang F, Zhao G, Zhang X, Zhu W, Li D. Utilizing daily excessive concentration hours to estimate small for gestational age infants attributable to fine particulate matter in Wuhan, China. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2024; 34:4047-4058. [PMID: 38576314 DOI: 10.1080/09603123.2024.2337835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024]
Abstract
Limited studies in China have explored the association between gravidae exposure to PM2.5 and small for gestational age infants (SGA), yielding inconsistent results. This study in Wuhan utilized daily excessive concentration hours (DECH) as a novel measure to assess PM2.5's impact on SGA. Data on air pollutants and pregnant women were collected from the Wuhan Municipal Ecology and Environmental Bureau and Wuhan Children's Hospital, respectively. Logistic regression models were employed to evaluate the contribution of PM2.5-DECH and PM2.5-mean to SGA. Significant correlations were observed between PM2.5-mean and SGA during the second trimester [OR = 1.23 (95% CI: 1.14-1.32)] and the entire pregnancy [OR = 1.15 (95% CI: 1.07-1.24)]. Similar correlations were found between PM2.5-DECH and SGA. These findings suggest that increased PM2.5 exposure is associated with a higher risk of SGA, and DECH may be used as a prospective substitute indicator for daily average concentration in similar studies.
Collapse
Affiliation(s)
- Xiaowei Zhang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, China
| | - Shijie Zhu
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, China
| | - Faxue Zhang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, China
| | - Gaichan Zhao
- Department of Public Health, School of Public Health, Wuhan University, Wuhan, China
| | - Xupeng Zhang
- Department of Public Health, School of Public Health, Wuhan University, Wuhan, China
| | - Wei Zhu
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, China
| | - Dejia Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, China
| |
Collapse
|
22
|
Yang R, Shi Z, Li Y, Huang X, Li Y, Li X, Chen Q, Hu Y, Li X. Research focus and emerging trends of the gut microbiome and infant: a bibliometric analysis from 2004 to 2024. Front Microbiol 2024; 15:1459867. [PMID: 39633813 PMCID: PMC11615055 DOI: 10.3389/fmicb.2024.1459867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
Background Over the past two decades, gut microbiota has demonstrated unprecedented potential in human diseases and health. The gut microbiota in early life is crucial for later health outcomes. This study aims to reveal the knowledge collaboration network, research hotspots, and explore the emerging trends in the fields of infant and gut microbiome using bibliometric analysis. Method We searched the literature on infant and gut microbiome in the Web of Science Core Collection (WOSCC) database from 2004 to 2024. CiteSpace V (version: 6.3.R1) and VOSview (version: 1.6.20) were used to display the top authors, journals, institutions, countries, authors, keywords, co-cited articles, and potential trends. Results A total of 9,899 documents were retrieved from the Web of Science Core Collection. The United States, China, and Italy were the three most productive countries with 3,163, 1,510, and 660 publications. The University of California System was the most prolific institution (524 publications). Van Sinderen, Douwe from University College Cork of Ireland was the most impactful author. Many studies have focused on atopic dermatitis (AD), necrotizing enterocolitis (NEC), as well as the immune mechanisms and microbial treatments for these diseases, such as probiotic strains mixtures and human milk oligosaccharides (HMOs). The mother-to-infant microbiome transmission, chain fatty acids, and butyrate maybe the emerging trends. Conclusion This study provided an overview of the knowledge structure of infant and gut microbiome, as well as a reference for future research.
Collapse
Affiliation(s)
- Ru Yang
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Zeyao Shi
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Yuan Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xi Huang
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Yingxin Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xia Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Qiong Chen
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Yanling Hu
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xiaowen Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
23
|
Zhang B, Han X, Long W, Xi S, Yu B, Yuan X. Association Between Red Blood Cell Distribution Width in Late Pregnancy and the Incidence of Adverse Perinatal Outcomes: A Retrospective Cohort Study. Arch Med Res 2024; 55:103057. [PMID: 39067407 DOI: 10.1016/j.arcmed.2024.103057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/06/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND AND AIMS This study aimed to quantify adverse perinatal outcomes (APO), including small/large for gestational age (SGA/LGA) and preterm birth (PTB), in pregnant women with abnormal red cell distribution width (RDW) and explore the related mechanisms. METHODS This study included 11,659 pregnant women who delivered in a specialized hospital. At the time of admission, the lipid profiles and whole blood cell counts were assessed, and APO was analyzed. RESULTS Women with high RDW (>18.5% [the 97.5th percentile]) in late pregnancy had a higher risk of LGA compared with those with low RDW (<12.3% [the 2.5th percentile]), whereas women with low RDW had a higher risk of SGA and PTB, compared with those with high RDW. A 1% increase in RDW was associated with an increased risk of LGA and a decreased risk of SGA and PTB. Consistent associations were observed in sensitivity analysis among pregnant women of non-advanced age, non-obesity, non-pregnancy complications, and non-PTB (for SGA/LGA only). Increased RDW was significantly associated with increased triglycerides and decreased high-density lipoprotein cholesterol (HDL-C). Triglycerides and HDL-C significantly mediated 10.63 and 15.8% of RDW-associated LGA, 9.51% and 9.40 of RDW-associated SGA, and 8.44 and -8.25% of RDW-associated PTB, respectively. CONCLUSION Abnormal RDW was associated with an increased risk of APO, and the RDW-associated APO risk could be partially mediated by triglycerides and HDL-C, suggesting that RDW may be a promising APO predictor.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Xiaoya Han
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Wei Long
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Sijie Xi
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Bin Yu
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Xiaosong Yuan
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China.
| |
Collapse
|
24
|
He Q, Liu H, Lu L, Zhang Q, Wang Q, Wang B, Wu X, Guan L, Mao J, Xue Y, Zhang C, Cao X, He Y, Peng X, Peng H, Zhao K, Li H, Jin X, Zhao L, Zhang J, Wang T. A genome-wide association study of neonatal metabolites. CELL GENOMICS 2024; 4:100668. [PMID: 39389019 PMCID: PMC11602626 DOI: 10.1016/j.xgen.2024.100668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 12/16/2023] [Accepted: 09/11/2024] [Indexed: 10/12/2024]
Abstract
Genetic factors significantly influence the concentration of metabolites in adults. Nevertheless, the genetic influence on neonatal metabolites remains uncertain. To bridge this gap, we employed genotype imputation techniques on large-scale low-pass genome data obtained from non-invasive prenatal testing. Subsequently, we conducted association studies on a total of 75 metabolic components in neonates. The study identified 19 previously reported associations and 11 novel associations between single-nucleotide polymorphisms and metabolic components. These associations were initially found in the discovery cohort (8,744 participants) and subsequently confirmed in a replication cohort (19,041 participants). The average heritability of metabolic components was estimated to be 76.2%, with a range of 69%-78.8%. These findings offer valuable insights into the genetic architecture of neonatal metabolism.
Collapse
Affiliation(s)
- Quanze He
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province 215000, China; Suzhou Municipal Hospital, Suzhou Jiangsu 215000, China
| | - Hankui Liu
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Clin Lab, BGI Genomics, Shijiazhuang 050035, China; BGI Genomics, Shenzhen 518083, China
| | - Lu Lu
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province 215000, China
| | - Qin Zhang
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province 215000, China
| | - Qi Wang
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province 215000, China
| | - Benjing Wang
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province 215000, China
| | - Xiaojuan Wu
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province 215000, China
| | - Liping Guan
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Clin Lab, BGI Genomics, Shijiazhuang 050035, China; BGI Genomics, Shenzhen 518083, China
| | - Jun Mao
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province 215000, China
| | - Ying Xue
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province 215000, China
| | - Chunhua Zhang
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province 215000, China
| | - Xinye Cao
- Clinical Medicine Department, Xinjiang Medical University, Urumqi, Xinjiang Province 830054, China
| | - Yuxing He
- Clinical Medicine Department, Xinjiang Medical University, Urumqi, Xinjiang Province 830054, China
| | - Xiangwen Peng
- Changsha Hospital for Maternal and Child Health Care of Hunan Normal University, Changsha, Hunan Province 431005, China
| | | | - Kangrong Zhao
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province 215000, China
| | - Hong Li
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province 215000, China
| | - Xin Jin
- BGI Research, Shenzhen 518083, China; The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China.
| | - Lijian Zhao
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Clin Lab, BGI Genomics, Shijiazhuang 050035, China; BGI Genomics, Shenzhen 518083, China; Medical Technology College, Hebei Medical University, Shijiazhuang 050000, China.
| | - Jianguo Zhang
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Clin Lab, BGI Genomics, Shijiazhuang 050035, China; BGI Research, Shenzhen 518083, China; School of Public Health, Hebei Medical University, Shijiazhuang 050000, China.
| | - Ting Wang
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province 215000, China; Suzhou Municipal Hospital, Suzhou Jiangsu 215000, China.
| |
Collapse
|
25
|
Ozarslan N, Mong C, Ategeka J, Li L, Buarpung S, Robinson JF, Kizza J, Kakuru A, Kamya MR, Dorsey G, Rosenthal PJ, Gaw SL. Placental Malaria Induces a Unique Methylation Profile Associated with Fetal Growth Restriction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593431. [PMID: 38798500 PMCID: PMC11118523 DOI: 10.1101/2024.05.09.593431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
BACKGROUND Fetal growth restriction (FGR) is associated with perinatal death and adverse birth outcomes, as well as long-term complications, including increased childhood morbidity, abnormal neurodevelopment, and cardio-metabolic diseases in adulthood. Placental epigenetic reprogramming associated with FGR may mediate these long-term outcomes. Placental malaria (PM), characterized by sequestration of Plasmodium falciparum-infected erythrocytes in placental intervillous space, is the leading global cause of FGR, but its impact on placental epigenetics is unknown. We hypothesized that placental methylomic profiling would reveal common and distinct mechanistic pathways of non-malarial and PM-associated FGR. RESULTS We analyzed placentas from a US cohort with no malaria exposure (n = 12) and a cohort from eastern Uganda, a region with a high prevalence of malaria (n = 12). From each site, 8 cases of FGR (defined as birth weight <10%ile for gestational age by Intergrowth-21 standard curves) and 4 healthy controls with normal weight were analyzed. PM was diagnosed by placental histopathology. We compared the methylation levels of over 850K CpGs of the placentas using Infinium MethylationEPIC v1 microarray. Non-malarial FGR was associated with 65 differentially methylated CpGs (DMCs), whereas PM-FGR was associated with 133 DMCs, compared to their corresponding controls without FGR. One DMC (cg16389901, located in the promoter region of BMP4) was commonly hypomethylated in both groups. We identified 522 DMCs between non-malarial FGR vs. PM-FGR placentas, which was independent of differing geographic location or cellular composition. CONCLUSION Placentas with PM-associated FGR have distinct methylation profiles as compared to placentas with non-malarial FGR, suggesting novel epigenetic reprogramming in response to malaria. Larger cohort studies are needed to determine the distinct long-term health outcomes in PM-associated FGR pregnancies.
Collapse
|
26
|
Ribeiro IT, Fioretto MN, Dos Santos SAA, Colombelli KT, Portela LMF, Niz Alvarez MV, de Magalhães Padilha P, Delgado AQ, Marques MVLSG, Bosqueiro JR, Seiva FRF, Barbisan LF, de Andrade Paes AM, Zambrano E, Justulin LA. Maternal protein restriction combined with postnatal sugar consumption alters liver proteomic profile and metabolic pathways in adult male offspring rats. Mol Cell Endocrinol 2024; 592:112316. [PMID: 38880278 DOI: 10.1016/j.mce.2024.112316] [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] [Received: 04/18/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/18/2024]
Abstract
This study investigated the impact of maternal protein restriction (MPR) and early postnatal sugar consumption (SUG) on the liver health of adult male descendant rats. Male offspring of mothers fed a normal protein diet (NPD) or a low protein diet (LPD) were divided into four groups: Control (CTR), Sugar Control (CTR + SUG), LPD during gestation and lactation (GLLP), and LPD with sugar (GLLP + SUG). Sugar consumption (10% glucose diluted in water) began after weaning on day 21 (PND 21), and at 90 days (PND 90), rats were sacrificed for analysis. Sugar intake reduced food intake and increased water consumption in CTR + SUG and GLLP + SUG compared to CTR and GLLP. GLLP and GLLP + SUG groups showed lower body weight and total and retroperitoneal fat compared to CTR and CTR + SUG. CTR + SUG and GLLP + SUG groups exhibited hepatocyte vacuolization associated with increased hepatic glycogen content compared to CTR and GLLP. Hepatic catalase activity increased in GLLP compared to CTR. Proteomic analysis identified 223 differentially expressed proteins (DEPs) among experimental groups. While in the GLLP group, the DEPs enriched molecular pathways related to cellular stress, glycogen metabolic pathways were enriched in the GLLP + SUG and CTR + SUG groups. The association of sugar consumption amplifies the effects of MPR, deregulating molecular mechanisms related to metabolism and the antioxidant system.
Collapse
Affiliation(s)
- Isabelle Tenori Ribeiro
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Matheus Naia Fioretto
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Sérgio Alexandre Alcantara Dos Santos
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil; Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Ketlin Thassiani Colombelli
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Luiz Marcos Frediani Portela
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | | | - Pedro de Magalhães Padilha
- Department of Chemical and Biological Sciences, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Aislan Quintiliano Delgado
- Department of Physical Education, Institute of Biosciences, Sao Paulo State University, Bauru, SP, Brazil
| | | | - José Roberto Bosqueiro
- Department of Physical Education, Institute of Biosciences, Sao Paulo State University, Bauru, SP, Brazil
| | - Fábio Rodrigues Ferreira Seiva
- Department of Chemical and Biological Sciences, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luís Fernando Barbisan
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | | | - Elena Zambrano
- Department Reproductive Biology, Salvador Zubirán National Institute of Medical Sciences and Nutrition, Mexico City, Mexico; Facultad de Química, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - Luis Antonio Justulin
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil.
| |
Collapse
|
27
|
Takatani T, Shiohama T, Takatani R, Hattori S, Yokota H, Hamada H. Brain morphometric changes in children born as small for gestational age without catch up growth. Front Neurosci 2024; 18:1441563. [PMID: 39268030 PMCID: PMC11390431 DOI: 10.3389/fnins.2024.1441563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/01/2024] [Indexed: 09/15/2024] Open
Abstract
Introduction Most infants born as small for gestational age (SGA) demonstrate catch up growth by 2-4 years, but some fail to do so. This failure is associated with several health risks, including neuropsychological development issues. However, data on the morphological characteristics of the brains of infants born as SGA without achieving catch up growth are lacking. This study aims to determine the structural aspects of the brains of children born as SGA without catch up growth. Methods We conducted voxel- and surface-based morphometric analyses of 1.5-T T1-weighted brain images scanned from eight infants born as SGA who could not achieve catch up growth by 3 years and sixteen individuals with idiopathic short stature (ISS) to exclude body size effects. Growth hormone (GH) secretion stimulation tests were used to rule out GH deficiency in all SGA and ISS cases. The magnetic resonance imaging data were assessed using Levene's test for equality of variances and a two-tailed unpaired t-test for equality of means. The Benjamini-Hochberg procedure was used to apply discovery rate correction for multiple comparisons. Results Morphometric analyses of both t-statical map and surface-based analyses using general linear multiple analysis determined decreased left insula thickness and volume in SGA without catch up growth compared with ISS. Conclusion The brain scans of patients with SGA who lack catch up growth indicated distinct morphological disparities when compared to those with ISS. The discernible features of brain morphology observed in patients born as SGA without catch up growth may improve understanding of the association of SGA without catch up growth with both intellectual and psychological outcomes.
Collapse
Affiliation(s)
- Tomozumi Takatani
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tadashi Shiohama
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Rieko Takatani
- Faculty of Education, Graduate School of Education, Chiba University, Chiba, Japan
| | - Shinya Hattori
- Department of Radiology, Chiba University Hospital, Chiba, Japan
| | - Hajime Yokota
- Department of Diagnostic Radiology and Radiation Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiromichi Hamada
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
28
|
Yuan W, Jiang J, Song SS, Liu J, Chen M, Shi Q, Zhang Y, Chen L, Qin Y, Wang R, Guo T, Wang X, Ma Q, Dong Y, Ma J. The association of breastfeeding duration on metabolic syndrome among children and adolescents, stratified by birth weight for gestational age. Pediatr Obes 2024; 19:e13145. [PMID: 38890760 DOI: 10.1111/ijpo.13145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/19/2024] [Accepted: 05/17/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND AND OBJECTIVES This study aimed to examine the associations between breastfeeding duration and metabolic syndrome (MetS) in adolescents and to further investigate the role of birth weight for gestational age (GA) on these associations. METHODS A total of 10 275 participants aged 7 to 18 years were included applying multistage cluster random sampling from a Chinese national survey. Birth weight was classified into small for GA (SGA), appropriate for GA (AGA) and large for GA (LGA). Information was collected through a self-administered questionnaire, physical examination and blood biochemical examination. Multivariable linear regression, logistic regression models, restricted cubic spline models were applied to assess the relationships of breastfeeding duration and MetS with different birth weight for GA. RESULTS The prevalence of non-breastfeeding, 0-5, 6-12 and >12 months groups were 16.2%, 23.1%, 42.5% and 18.2%, and the prevalence of SGA and LGA was 11.9% and 12.7%, respectively. Prolonged breastfeeding duration was associated with higher odds of MetS (β: 0.08, 95% CI: 0.03, 0.13), WC (β: 3.49, 95% CI: 2.82, 4.16) and SBP (β: 2.34, 95% CI: 1.80, 2.89). SGA and prolonged breastfeeding synergistically increased MetS risks, but LGA appeared to offset the adverse effects of prolonged breastfeeding. CONCLUSION Prolonged breastfeeding may increase children's MetS risks. SGA synergies with prolonged breastfeeding increased MetS burden in children and adolescents, while LGA mitigated the risks. This reminds us that intensive attention should be paid to both early birth weight and subsequent living environment for children and adolescents' lifelong health.
Collapse
Affiliation(s)
- Wen Yuan
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, PR China
| | - Jianuo Jiang
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, PR China
| | - Sha-Sha Song
- Pathology Department, Yantai Fushan People's Hospital, Yantai, PR China
| | - Jieyu Liu
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, PR China
| | - Manman Chen
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Qiaoxin Shi
- School of Public Health, The University of Hong Kong Li Ka Shing Faculty of Medicine, Hong Kong, PR China
| | - Yi Zhang
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, PR China
| | - Li Chen
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, PR China
| | - Yang Qin
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, PR China
| | - Ruolin Wang
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, PR China
| | - Tongjun Guo
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, PR China
| | | | - Qi Ma
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, PR China
| | - Yanhui Dong
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, PR China
| | - Jun Ma
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, PR China
| |
Collapse
|
29
|
Díez López I, Cernada M, Galán L, Boix H, Ibañez L, Couce ML. Small for gestational age: concept, diagnosis and neonatal characterization, follow-up and recommendations. An Pediatr (Barc) 2024; 101:124-131. [PMID: 39127580 DOI: 10.1016/j.anpede.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 06/12/2024] [Indexed: 08/12/2024] Open
Abstract
Newborns who do not reach a weight appropriate for their gestational age and sex can be classified in different ways. This article defines the concepts of small for gestational age (SGA) and intrauterine growth restriction, as well as the underlying causes of these conditions, with the goal of establishing consensus definitions for these patients, in whom treatment with growth hormone throughout childhood may be indicated and who may be at risk of developing endocrine or metabolic disorders in puberty and adulthood. Most SGA children experience spontaneous catch-up growth that is usually completed by age 2 years. In SGA children who remain short, treatment with recombinant human growth hormone is effective, increasing adult height. Small for gestational age infants with rapid catch-up growth and marked weight gain are at increased risk of premature adrenarche, early puberty, polycystic ovary syndrome (girls), insulin resistance and obesity, all of which are risk factors for type 2 diabetes and metabolic syndrome in adulthood. The SGA status can affect different areas of neurodevelopment and manifest at different stages in life; neurodevelopmental outcomes are better in SGA infants with spontaneous catch-up growth. Due to the potential risks associated with SGA, adequate characterization of these patients at birth is imperative, as it allows initiation of appropriate follow-up and early detection of abnormalities.
Collapse
Affiliation(s)
- Ignacio Díez López
- Departamento de Pediatría, UPV-EHU, Servicio de Pediatría. HU Araba, OSI Araba, BIOARABA, Vitoria, Spain
| | - María Cernada
- Servicio de Neonatología, Hospital Universitario y Politécnico La Fe, Grupo de Investigación en Perinatología, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Laura Galán
- Servicio de Pediatria, Hospital San Pedro de Alcántara, Cáceres, Spain
| | - Hector Boix
- Unidad de Cuidados Intensivos Neonatales, Hospital Quironsalud, Barcelona, Spain
| | - Lourdes Ibañez
- Servicio de Endocrinología, Hospital Sant Joan de Déu, Universidad de Barcelona, CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain.
| | - Maria L Couce
- Servicio de Neonatología, Hospital Clínico Universitario de Santiago, IDIS, RICORS-SAMID, Universidad de Santiago de Compostela, Santiago de Compostela, A Coruña, Spain.
| |
Collapse
|
30
|
Meesters M, Van Eetvelde M, Verdru K, Govaere J, Opsomer G. Small for Gestational Age Calves: Part II-Reduced Fertility, Productive Performance, and Survival in Holstein Friesian Heifers Born Small for Their Gestational Age. Animals (Basel) 2024; 14:2157. [PMID: 39123682 PMCID: PMC11311054 DOI: 10.3390/ani14152157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Recently: more attention has been given to low-birth-weight calves, often without considering gestation length. Calves can be classified as small for gestational age (SGA) when their birth weight is below the 10th percentile, similar to the definition in human medicine. While SGA babies face various health risks, it remains unclear if SGA calves face similar long-term health consequences. This study aimed to investigate the long-term effects on fertility, productive performance, and overall survival in Holstein Friesian (HF) heifers born SGA. Chi-squared analysis assessed culling and survival rates, and linear mixed-effect models evaluated the impact of gestational age group (small, average, or large for gestational age, respectively, SGA, AGA, and LGA) on growth, fertility, milk yield, and lifespan. SGA calves showed catch-up growth at six months but weighed significantly less at twelve months (p = 0.003). Age at first insemination and calving did not differ significantly, although SGA heifers required more inseminations (2.3 ± 1.50) compared to AGA and LGA heifers (1.7 ± 0.98 and 1.5 ± 0.89, respectively, p = 0.006). SGA calves tended to be culled more during the first lactation than AGA calves (25.0% vs. 11.9%, p = 0.078) and showed lower survival to second calving (p = 0.019) compared to AGA and LGA heifers. The Kaplan-Meier analysis indicated a tendency for gestational age to affect overall survival (p = 0.1), with SGA heifers having a higher risk of leaving the herd prematurely (p = 0.035, hazard ratio = 1.53). Milk yield per productive day was significantly lower in SGA heifers (21.2 ± 8.73 kg) compared to AGA and LGA heifers (26.9 ± 5.01 kg and 26.3 ± 3.38 kg, respectively, p = 0.006). This study reveals that HF calves born SGA suffer long-term consequences, although further research is needed to understand the economic impact of rearing SGA heifers.
Collapse
Affiliation(s)
- Maya Meesters
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | | | | | | | | |
Collapse
|
31
|
Ding L, Weger BD, Liu J, Zhou L, Lim Y, Wang D, Xie Z, Liu J, Ren J, Zheng J, Zhang Q, Yu M, Weger M, Morrison M, Xiao X, Gachon F. Maternal high fat diet induces circadian clock-independent endocrine alterations impacting the metabolism of the offspring. iScience 2024; 27:110343. [PMID: 39045103 PMCID: PMC11263959 DOI: 10.1016/j.isci.2024.110343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 05/02/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Maternal obesity has long-term effects on offspring metabolic health. Among the potential mechanisms, prior research has indicated potential disruptions in circadian rhythms and gut microbiota in the offspring. To challenge this hypothesis, we implemented a maternal high fat diet regimen before and during pregnancy, followed by a standard diet after birth. Our findings confirm that maternal obesity impacts offspring birth weight and glucose and lipid metabolisms. However, we found minimal impact on circadian rhythms and microbiota that are predominantly driven by the feeding/fasting cycle. Notably, maternal obesity altered rhythmic liver gene expression, affecting mitochondrial function and inflammatory response without disrupting the hepatic circadian clock. These changes could be explained by a masculinization of liver gene expression similar to the changes observed in polycystic ovarian syndrome. Intriguingly, such alterations seem to provide the first-generation offspring with a degree of protection against obesity when exposed to a high fat diet.
Collapse
Affiliation(s)
- Lu Ding
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Benjamin D. Weger
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Jieying Liu
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Liyuan Zhou
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100022, China
| | - Yenkai Lim
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Dongmei Wang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ziyan Xie
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jing Liu
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jing Ren
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jia Zheng
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, China
| | - Qian Zhang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Miao Yu
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Meltem Weger
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Mark Morrison
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia
- Australian Infectious Diseases Research Centre, St. Lucia, QLD 4072, Australia
| | - Xinhua Xiao
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Frédéric Gachon
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| |
Collapse
|
32
|
Stinson SE, Kromann Reim P, Lund MAV, Lausten-Thomsen U, Aas Holm L, Huang Y, Brøns C, Vaag A, Thiele M, Krag A, Fonvig CE, Grarup N, Pedersen O, Christiansen M, Ängquist L, Sørensen TIA, Holm JC, Hansen T. The interplay between birth weight and obesity in determining childhood and adolescent cardiometabolic risk. EBioMedicine 2024; 105:105205. [PMID: 38918147 PMCID: PMC11293585 DOI: 10.1016/j.ebiom.2024.105205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/29/2024] [Accepted: 06/02/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Birth weight (BW) is associated with risk of cardiometabolic disease (CMD) in adulthood, which may depend on the state of obesity, in particular if developed at a young age. We hypothesised that BW and a polygenic score (PGS) for BW were associated with cardiometabolic risk and related plasma protein levels in children and adolescents. We aimed to determine the modifying effect of childhood obesity on these associations. METHODS We used data from The cross-sectional HOLBAEK Study with 4263 participants (median [IQR] age, 11.7 [9.2, 14.3] years; 57.1% girls and 42.9% boys; 48.6% from an obesity clinic and 51.4% from a population-based group). We gathered information on BW and gestational age, anthropometrics, cardiometabolic risk factors, calculated a PGS for BW, and measured plasma proteins using Olink Inflammation and Cardiovascular II panels. We employed multiple linear regression to examine the associations with BW as a continuous variable and performed interaction analyses to assess the effect of childhood obesity on cardiometabolic risk and plasma protein levels. FINDINGS BW and a PGS for BW associated with cardiometabolic risk and plasma protein levels in childhood and adolescence. Childhood obesity modified the associations between BW and measures of insulin resistance, including HOMA-IR (βadj [95% CI per SD] for obesity: -0.12 [-0.15, -0.08]; normal weight: -0.04 [-0.08, 0.00]; Pinteraction = 0.004), c-peptide (obesity: -0.11 [-0.14, -0.08]; normal weight: -0.02 [-0.06, 0.02]; Pinteraction = 5.05E-04), and SBP SDS (obesity: -0.12 [-0.16, -0.08]; normal weight: -0.06 [-0.11, -0.01]; Pinteraction = 0.0479). Childhood obesity also modified the associations between BW and plasma levels of 14 proteins (e.g., IL15RA, MCP1, and XCL1; Pinteraction < 0.05). INTERPRETATION We identified associations between lower BW and adverse metabolic phenotypes, particularly insulin resistance, blood pressure, and altered plasma protein levels, which were more pronounced in children with obesity. Developing effective prevention and treatment strategies for this group is needed to reduce the risk of future CMD. FUNDING Novo Nordisk Foundation (NNF15OC0016544, NNF0064142 to T.H., NNF15OC0016692 to T.H. and A.K., NNF18CC0033668 to S.E.S, NNF18SA0034956 to C.E.F., NNF20SA0067242 to DCA, NNF18CC0034900 to NNF CBMR), The Innovation Fund Denmark (0603-00484B to T.H.), The Danish Cardiovascular Academy (DCA) and the Danish Heart Foundation (HF) (PhD2021007-DCA to P.K.R, 18-R125-A8447-22088 (HF) and 21-R149-A10071-22193 (HF) to M.A.V.L., PhD2023009-HF to L.A.H), EU Horizon (668031, 847989, 825694, 964590 to A.K.), Innovative Health Initiative (101132901 for A.K.), A.P. Møller Foundation (19-L-0366 to T.H.), The Danish National Research Foundation, Steno Diabetes Center Sjælland, and The Region Zealand and Southern Denmark Health Scientific Research Foundation.
Collapse
Affiliation(s)
- Sara Elizabeth Stinson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pauline Kromann Reim
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Asp Vonsild Lund
- The Children's Obesity Clinic, Accredited European Centre for Obesity Management, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Lausten-Thomsen
- Department of Neonatology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Louise Aas Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Children's Obesity Clinic, Accredited European Centre for Obesity Management, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark
| | - Yun Huang
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Allan Vaag
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Maja Thiele
- Center for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark; Department for Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Aleksander Krag
- Center for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark; Department for Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Cilius Esmann Fonvig
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Children's Obesity Clinic, Accredited European Centre for Obesity Management, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Center for Clinical Metabolic Research, Herlev-Gentofte University Hospital, Denmark
| | - Michael Christiansen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department for Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark
| | - Lars Ängquist
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thorkild I A Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Center for Childhood Health, Copenhagen, Denmark
| | - Jens-Christian Holm
- The Children's Obesity Clinic, Accredited European Centre for Obesity Management, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
33
|
Maw AA, Thwin T, Owino VO, Ward LC. Development of a bioelectrical impedance analysis-based prediction equation for body composition of rural children aged 4-8 years in Myanmar. Nutr Health 2024:2601060241260983. [PMID: 38860329 DOI: 10.1177/02601060241260983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Background: Reliable and accurate estimates of body composition are essential when studying the various health correlates of disease. Bioelectrical impedance analysis (BIA) is an affordable and feasible body composition assessment technique for clinical and field settings. Total body water (TBW) and hence fat-free mass is estimated by predictive regression algorithms using anthropometric measurements plus the resistance index. Aim: The study aimed to develop a BIA prediction equation for TBW in children in Myanmar using the deuterium dilution technique as the reference method. Methods: The study design was cross-sectional in a school setting with convenience sampling of participants. One hundred and two healthy children (57 boys and 45 girls) with aged 4 and 8 years participated; randomly divided into the prediction group (29 boys and 22 girls) and cross-validation group (28 boys and 23 girls). Whole-body impedance, anthropometric and TBW (by D2O dilution) measurements. The prediction equation was cross-validated using a split-group design and compared to published equations for contemporaneous populations. Results: TBW could be predicted by the following equation. TBW = 0.4597 * Weight (kg) + 0.1564 * Impedance index + 0.6075 (R2 = 0.891, P < 0.0001) with a correlation coefficient of 0.942 and limits of agreement of 0.98 kg TBW on cross-validation. Conclusions: This equation can be used to predict body composition in young (aged 4-8 years) children in Myanmar but because the age range of the participants in the present study was relatively narrow, more research in different age groups is required to establish its broader applicability.
Collapse
Affiliation(s)
- Aye Aye Maw
- Department of Medical Research, Myanmar Ministry of Health and Sports, Yangon, Myanmar
| | - Theingi Thwin
- Department of Medical Research, Myanmar Ministry of Health and Sports, Yangon, Myanmar
| | - Victor O Owino
- Division of Human Health, International Atomic Energy Agency, Vienna, Austria
| | - Leigh C Ward
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| |
Collapse
|
34
|
Singh S, Bortoletto P, Wylie BJ, Melnick AP, Prabhu M. The impact of reference growth standards on small- and large-for-gestational age outcomes among pregnancies conceived by fresh and frozen embryo transfers. F S Rep 2024; 5:164-169. [PMID: 38983739 PMCID: PMC11228793 DOI: 10.1016/j.xfre.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 07/11/2024] Open
Abstract
Objective To describe differences in the frequency of small-for-gestational age (SGA) and large-for-gestational age (LGA) driven by different birth weight curves in assisted reproductive technology (ART)-conceived pregnancies. Design Retrospective cohort study. Setting Single academic medical center. Patients Singleton live births between the gestational ages of 36 weeks and 0 days and 42 weeks and 6 days from fresh or frozen embryo transfer (ET). Interventions None. Main Outcome Measures SGA (<10th percentile) and LGA (>90th percentile) classified by Fenton, INTERGROWTH-21, World Health Organization, Duryea, and Oken curves. Results The median birth weight and gestational age at birth among fresh ET pregnancies were 3,289g (interquartile range [IQR], 2,977-3,600g) and 39.4 (IQR, 38.6-40.3) weeks, respectively, and those among frozen ET pregnancies were 3,399g (IQR, 3,065-3,685g) and 39.4 (IQR, 38.7-40.1) weeks, respectively. The frequencies of SGA neonates using each birth weight standard ranged from 5.8% to 13.4% for fresh ET and from 3.5% to 8.7% for frozen ET. Those of LGA neonates ranged from 5.3% to 14.3% for fresh ET and from 6.6% to 21.2% for frozen ET. Conclusion The frequency of SGA and LGA neonates among ART-conceived gestations is partially driven by the birth weight standard. Selecting an appropriate standard that best reflects the patient population is critical to quantifying the risk of ART-conceived pregnancies.
Collapse
Affiliation(s)
- Sunidhi Singh
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, New York
- Department of Obstetrics, Gynecology, and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pietro Bortoletto
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medical College, New York, New York
- Boston IVF, Waltham, Massachusetts
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Blair J Wylie
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| | - Alexis P Melnick
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medical College, New York, New York
| | - Malavika Prabhu
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, New York
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
35
|
Knuth MM, Xue J, Elnagheeb M, Gharaibeh RZ, Schoenrock SA, McRitchie S, Brouwer C, Sumner SJ, Tarantino L, Valdar W, Rector RS, Simon JM, Ideraabdullah F. Early life exposure to vitamin D deficiency impairs molecular mechanisms that regulate liver cholesterol biosynthesis, energy metabolism, inflammation, and detoxification. Front Endocrinol (Lausanne) 2024; 15:1335855. [PMID: 38800476 PMCID: PMC11116800 DOI: 10.3389/fendo.2024.1335855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction Emerging data suggests liver disease may be initiated during development when there is high genome plasticity and the molecular pathways supporting liver function are being developed. Methods Here, we leveraged our Collaborative Cross mouse model of developmental vitamin D deficiency (DVD) to investigate the role of DVD in dysregulating the molecular mechanisms underlying liver disease. We defined the effects on the adult liver transcriptome and metabolome and examined the role of epigenetic dysregulation. Given that the parental origin of the genome (POG) influences response to DVD, we used our established POG model [POG1-(CC011xCC001)F1 and POG2-(CC001xCC011)F1] to identify interindividual differences. Results We found that DVD altered the adult liver transcriptome, primarily downregulating genes controlling liver development, response to injury/infection (detoxification & inflammation), cholesterol biosynthesis, and energy production. In concordance with these transcriptional changes, we found that DVD decreased liver cell membrane-associated lipids (including cholesterol) and pentose phosphate pathway metabolites. Each POG also exhibited distinct responses. POG1 exhibited almost 2X more differentially expressed genes (DEGs) with effects indicative of increased energy utilization. This included upregulation of lipid and amino acid metabolism genes and increased intermediate lipid and amino acid metabolites, increased energy cofactors, and decreased energy substrates. POG2 exhibited broader downregulation of cholesterol biosynthesis genes with a metabolomics profile indicative of decreased energy utilization. Although DVD primarily caused loss of liver DNA methylation for both POGs, only one epimutation was shared, and POG2 had 6.5X more differentially methylated genes. Differential methylation was detected at DEGs regulating developmental processes such as amino acid transport (POG1) and cell growth & differentiation (e.g., Wnt & cadherin signaling, POG2). Conclusions These findings implicate a novel role for maternal vitamin D in programming essential offspring liver functions that are dysregulated in liver disease. Importantly, impairment of these processes was not rescued by vitamin D treatment at weaning, suggesting these effects require preventative measures. Substantial differences in POG response to DVD demonstrate that the parental genomic context of exposure determines offspring susceptibility.
Collapse
Affiliation(s)
- Megan M. Knuth
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jing Xue
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
| | - Marwa Elnagheeb
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Raad Z. Gharaibeh
- Department of Medicine, Division of Gastroenterology, University of Florida, Gainesville, FL, United States
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, United States
| | - Sarah A. Schoenrock
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Susan McRitchie
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
| | - Cory Brouwer
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, United States
- University of North Carolina at Charlotte Bioinformatics Service Division, North Carolina Research Campus, Kannapolis, NC, United States
| | - Susan J. Sumner
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
- Department of Nutrition, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lisa Tarantino
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - William Valdar
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - R. Scott Rector
- Research Service, Harry S. Truman Memorial Veterans Medical Center, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, United States
| | - Jeremy M. Simon
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Neuroscience Center Bioinformatics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Folami Ideraabdullah
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
- Department of Nutrition, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
36
|
Rancière F, Wafo O, Perrot X, Momas I. Associations between heat wave during pregnancy and term birth weight outcomes: The PARIS birth cohort. ENVIRONMENT INTERNATIONAL 2024; 188:108730. [PMID: 38776654 DOI: 10.1016/j.envint.2024.108730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/28/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Climate change will make extreme weather events more frequent in the 21st century. Extreme ambient temperatures during the prenatal period have been associated with adverse pregnancy outcomes such as preterm birth. It is unclear, however, whether heat waves during pregnancy impact fetal growth in apparently healthy term newborns. OBJECTIVES We aimed to investigate associations between heat wave during pregnancy and birth weight outcomes in term newborns from the PARIS birth cohort, and to explore meteorological conditions and air pollution as possible intermediate factors. METHODS We examined data on 3,359 newborns born between 37 and 42 weeks in Paris, France, between 2003 and 2006. Associations of maternal exposure to heat wave (during whole pregnancy and each trimester) with birth weight and small for gestational age (SGA) at term were studied using linear and logistic regression models adjusted for potential confounders. Maternal characteristics were investigated as possible modifiers. We explored the mediating role of ambient temperature, relative humidity, and air pollution levels in the relationship between heat wave during the first trimester and term SGA. RESULTS Mothers who were pregnant during the 2003 French heat wave (n = 506, 15 %) were more likely to have a term SGA baby (aOR = 2.70; 95 %CI: 1.38, 5.28) compared to mothers who did not experience heat wave during pregnancy. The association was stronger when heat wave occurred during the first trimester (aOR = 4.18; 95 %CI: 1.69, 10.35). Primiparous women were identified as more vulnerable than multiparous women. Average ambient temperature and air quality index explained about 36 % and 56 % of the association between heat wave during the first trimester and term SGA, respectively. CONCLUSIONS This study suggests prenatal exposure to heat wave, especially during the first trimester, may adversely affect fetal growth of term newborns, which could be explained by both increasing ambient temperatures and worsening air quality.
Collapse
Affiliation(s)
- Fanny Rancière
- Université Paris Cité, Inserm U1153 CRESS, Inrae, HERA team, Paris, France; Faculté de Pharmacie de Paris, Université Paris Cité, Paris, France.
| | - Octave Wafo
- Université Paris Cité, Inserm U1153 CRESS, Inrae, HERA team, Paris, France
| | - Xavier Perrot
- Laboratoire de Météorologie Dynamique, Ecole Normale Supérieure, CNRS, Paris, France
| | - Isabelle Momas
- Université Paris Cité, Inserm U1153 CRESS, Inrae, HERA team, Paris, France; Faculté de Pharmacie de Paris, Université Paris Cité, Paris, France; Cellule Cohorte, Direction de l'Action Sociale de l'Enfance et de la Santé, Mairie de Paris, Paris, France
| |
Collapse
|
37
|
Ye L, Huang Y, Chen K, Hang C, Ying Y, Zu L, Luo X, Du L. Early postnatal moderate catch‑up growth in rats with nutritional intrauterine growth restriction preserves pulmonary vascular and cognitive function in adulthood. Exp Ther Med 2024; 27:183. [PMID: 38515647 PMCID: PMC10952380 DOI: 10.3892/etm.2024.12471] [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: 10/25/2023] [Accepted: 02/09/2024] [Indexed: 03/23/2024] Open
Abstract
Intrauterine growth restriction (IUGR) with rapid postnatal catch-up growth is strongly associated with pulmonary vascular dysfunction in adulthood, whereas IUGR with delayed growth in early postnatal life results in long-term brain deficits. In the present study, it was hypothesized that IUGR with early moderate catch-up growth may alleviate pulmonary vascular remodeling in adulthood without affecting memory function. An IUGR model was established by restricting maternal nutrition during pregnancy. Different growth patterns were achieved by adjusting the litter size in each group during lactation. Rats meeting the weight requirement at weaning were selected for subsequent studies at three time points (3, 9 and 13 weeks). Cognitive function was evaluated using a Y-maze. Invasive hemodynamic measurements were conducted to measure the mean pulmonary arterial pressure (mPAP). In addition, primary pulmonary artery smooth muscle cells (PASMCs) and pulmonary vascular endothelial cells (PVECs) were cultured to investigate their role in the increase in mPAP following rapid catch-up growth. The results showed that memory function deficits in the rats in the delayed growth group were associated with reduced proliferation of neural stem cells in the subgranular zone of the hippocampus. Furthermore, moderate catch-up growth at the three time points improved memory function while maintaining a normal mPAP. In adult IUGR rats experiencing rapid catch-up growth, although memory function improved, elevated mPAP and medial thickening of pulmonary arterioles were observed. Additionally, PASMCs exhibited excessive proliferation, migration and anti-apoptotic activity in the rapid catch-up group, and PVECs also displayed excessive proliferation. These results suggested that moderate catch-up growth after IUGR is a better strategy for optimal cognition and cardiovascular health in adulthood compared with rapid catch-up growth or delayed growth.
Collapse
Affiliation(s)
- Lixia Ye
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Yajie Huang
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Kewei Chen
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Chengcheng Hang
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Yuhan Ying
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Lu Zu
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Xiaofei Luo
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Lizhong Du
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| |
Collapse
|
38
|
Brown LD, Rozance PJ, Wang D, Eroglu EC, Wilkening RB, Solmonson A, Wesolowski SR. Increased hepatic glucose production with lower oxidative metabolism in the growth-restricted fetus. JCI Insight 2024; 9:e176497. [PMID: 38687612 PMCID: PMC11141920 DOI: 10.1172/jci.insight.176497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 04/17/2024] [Indexed: 05/02/2024] Open
Abstract
Fetal growth restriction (FGR) is accompanied by early activation of hepatic glucose production (HGP), a hallmark of type 2 diabetes (T2D). Here, we used fetal hepatic catheterization to directly measure HGP and substrate flux in a sheep FGR model. We hypothesized that FGR fetuses would have increased hepatic lactate and amino acid uptake to support increased HGP. Indeed, FGR fetuses compared with normal (CON) fetuses had increased HGP and activation of gluconeogenic genes. Unexpectedly, hepatic pyruvate output was increased, while hepatic lactate and gluconeogenic amino acid uptake rates were decreased in FGR liver. Hepatic oxygen consumption and total substrate uptake rates were lower. In FGR liver tissue, metabolite abundance, 13C-metabolite labeling, enzymatic activity, and gene expression supported decreased pyruvate oxidation and increased lactate production. Isolated hepatocytes from FGR fetuses had greater intrinsic capacity for lactate-fueled glucose production. FGR livers also had lower energy (ATP) and redox state (NADH/NAD+ ratio). Thus, reduced hepatic oxidative metabolism may make carbons available for increased HGP, but also produces nutrient and energetic stress in FGR liver. Intrinsic programming of these pathways regulating HGP in the FGR fetus may underlie increased HGP and T2D risk postnatally.
Collapse
Affiliation(s)
- Laura D Brown
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Paul J Rozance
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Dong Wang
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Evren C Eroglu
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Randall B Wilkening
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ashley Solmonson
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Stephanie R Wesolowski
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
39
|
Cerasani J, Consales A, Gangi S, Macchi M, Morniroli D, Vizzari G, Tiraferri V, Petrelli A, Mosca F, Giannì ML. Exclusive human milk feeding and prevalence of early adiposity rebound in ELBW infants: a retrospective cohort study. Eur J Pediatr 2024; 183:1295-1303. [PMID: 38112801 PMCID: PMC10950974 DOI: 10.1007/s00431-023-05374-6] [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] [Received: 05/27/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023]
Abstract
The purpose of the present study was to evaluate the prevalence of early adiposity rebound (EAR) and factors associated with its occurrence in a cohort of extremely low birth weight infants (ELBW). We conducted a retrospective longitudinal study including ELBW infants followed-up for up to 10 years after discharge. EAR was defined as occurring before 5 years of age. A multivariate binary logistic regression analysis was performed to evaluate maternal and perinatal variables independently associated with EAR. Out of 212 ELBW infants included in the analysis, 40.6% developed EAR and 21.5% showed it before 4 years of age. Only formula milk feeding at discharge was independently associated with a higher risk of EAR. The mean BMI of children with EAR was higher than that of children without EAR. Furthermore, the prevalence of overweight and obesity was higher in the EAR group than in the timely AR group. Conclusions: ELBW infants in our cohort developed EAR in a relatively high percentage of cases. In this already at-risk population, EAR may represent a further risk factor for an adverse metabolic outcome. Monitoring preterm infants' growth within a long-term follow-up program and promoting and supporting human milk feeding is advisable. What is Known: • Preterm-born infants are at high risk for long-term adverse health outcomes, especially cardiovascular and metabolic. • The occurrence of early adiposity rebound (EAR) is associated with the risk of later obesity and metabolic syndrome. What is New: • The occurrence of EAR in ELBW infants may represent an additional risk factor for later adverse metabolic outcomes in an already vulnerable population. • Future preventive strategies should include a long-term follow-up and the promotion of exclusive breastfeeding.
Collapse
Affiliation(s)
- Jacopo Cerasani
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Alessandra Consales
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.
| | - Silvana Gangi
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marta Macchi
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Morniroli
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giulia Vizzari
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Tiraferri
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Angelo Petrelli
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Fabio Mosca
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Lorella Giannì
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
40
|
Meesters M, Van Eetvelde M, Beci B, Opsomer G. The importance of developmental programming in the dairy industry. Anim Reprod Sci 2024; 262:107428. [PMID: 38340370 DOI: 10.1016/j.anireprosci.2024.107428] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/18/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024]
Abstract
The concept of developmental programming suggests that environmental influences during pre- and early postnatal life that can have long-term effects on future health and performance. In dairy cattle, maternal body growth, age, parity and milk yield, as well as environmental factors during gestation, have the potential to create a suboptimal environment for the developing fetus. As a result, the calf's phenotype may undergo adaptations. Moreover, developmental programming can have long-term effects on subsequent birth weight, immunity and metabolism, as well as on postnatal growth, body composition, fertility, milk yield and even longevity of dairy cows. This review provides an overview of the impact of developmental programming on later health and performance in dairy cows.
Collapse
Affiliation(s)
- Maya Meesters
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium.
| | - Mieke Van Eetvelde
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Barbara Beci
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Geert Opsomer
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| |
Collapse
|
41
|
Cajachagua-Torres KN, Quezada-Pinedo HG, Guzman-Vilca WC, Tarazona-Meza C, Carrillo-Larco RM, Huicho L. Vulnerable newborn phenotypes in Peru: a population-based study of 3,841,531 births at national and subnational levels from 2012 to 2021. LANCET REGIONAL HEALTH. AMERICAS 2024; 31:100695. [PMID: 38500961 PMCID: PMC10945436 DOI: 10.1016/j.lana.2024.100695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/28/2024] [Accepted: 02/01/2024] [Indexed: 03/20/2024]
Abstract
Background We aimed to examine the national and subnational prevalence of vulnerable newborn phenotypes in Peru, 2012-2021. Methods Newborn phenotypes were defined using gestational age (preterm [PT], term [T]), birthweight for gestational age using INTERGROWTH-21st standards (small for gestational age [SGA], appropriate for gestational age [AGA] or large for gestational age [LGA]), and birthweight (low birthweight [LBW], non-LBW) using the Peruvian National Birth Registry as six (by excluding birthweight) and ten newborn phenotypes (using all three outcomes). Small phenotypes (with at least one classification of PT, SGA, or LBW) were further considered. Using individual-level data, we stratified the phenotypes by maternal educational level, maternal age, healthcare insurance, altitude of residence, and geographic region (Coast, Andes, and Amazon). Findings The prevalence of the five vulnerable newborn phenotypes for the study period was LGA+T (15.2%), AGA+PT (5.2%), SGA+T (4.6%), LGA+PT (0.8%), and SGA+PT (0.7%). The Coast had a higher prevalence of newborns with large phenotypes (19.4%) and the Highlands a higher prevalence of newborns with small phenotypes (12.5%). Mothers with poor socioeconomic status, extreme ages and living at high altitude had a higher prevalence of newborns with small phenotypes, and mothers who were wealthier, more educated, and older had a higher prevalence of infants with large phenotypes. Interpretation Our findings cautiously suggest that socioeconomic and geographic disparities may play a crucial role in shaping vulnerable newborn phenotypes at national and subnational level in Peru. Further studies using longitudinal data are needed to corroborate our findings and to identify individual-level risk factors. Funding Ter Meulen Grant from the KNAW Medical Sciences Fund of the Royal Netherlands Academy of Arts and Sciences (KNAWWF/1085/TMB406, KNAWWF/1327/TMB202116), Fogarty Program (D43TW011502).
Collapse
Affiliation(s)
- Kim N. Cajachagua-Torres
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Paediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, New York University Grossman of Medicine, New York University, New York, NY, USA
- Centro de Investigación en Salud Materna e Infantil and Centro de Investigación para el Desarrollo Integral y Sostenible, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Hugo G. Quezada-Pinedo
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Paediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Centro de Investigación en Salud Materna e Infantil and Centro de Investigación para el Desarrollo Integral y Sostenible, Universidad Peruana Cayetano Heredia, Lima, Peru
- Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
| | - Wilmer Cristobal Guzman-Vilca
- CRONICAS Centre of Excellence in Chronic Diseases, Universidad Peruana Cayetano Heredia, Lima, Peru
- School of Medicine “Alberto Hurtado”, Universidad Peruana Cayetano Heredia, Lima, Peru
- Sociedad Científica de Estudiantes de Medicina Cayetano Heredia (SOCEMCH), Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Carla Tarazona-Meza
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
- Nutrition and Dietetics, Universidad Científica del Sur, Lima, Perú
| | - Rodrigo M. Carrillo-Larco
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Emory Global Diabetes Research Center, Emory University, Atlanta, GA, USA
| | - Luis Huicho
- Centro de Investigación en Salud Materna e Infantil and Centro de Investigación para el Desarrollo Integral y Sostenible, Universidad Peruana Cayetano Heredia, Lima, Peru
- School of Medicine “Alberto Hurtado”, Universidad Peruana Cayetano Heredia, Lima, Peru
| |
Collapse
|
42
|
Fang X, Xie Y, Cao S, Liu J, Shi Y, Yu L, Zheng T, Liu H, Li Y, Xu S, Xia W. Associations between maternal urinary rare earth elements during pregnancy and birth weight-for-gestational age: Roles of cord blood vitamin D levels. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169222. [PMID: 38081430 DOI: 10.1016/j.scitotenv.2023.169222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/25/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Prenatal exposure to rare earth elements (REEs) may contribute to adverse birth outcomes in previous studies. Cord blood vitamin D has been suggested to modify or mediate the effects of environmental exposures. However, none has investigated these roles of cord blood vitamin D in the associations of prenatal exposure to REEs with fetal growth. Maternal trimester-specific urinary concentrations of 13 REEs, cord blood total 25-hydroxyvitamin D at delivery, and birth weight (BW)-for-gestational age (GA) were determined in 710 mother-newborn pairs from Wuhan, China. Higher maternal average urinary concentrations of europium (Eu), gadolinium (Gd), dysprosium (Dy), holmium (Ho), erbium (Er), and ytterbium (Yb) across three trimesters, either individually or jointly, were significantly associated with lower BW-for-GA Z-scores and higher odds of small for gestational age (SGA) [β = -0.092; 95 % confidence interval (CI): -0.149, -0.035 for BW-for-GA Z-scores, and odds ratio = 1.60; 95 % CI: 1.14, 2.24 for SGA involved in each unit increase in weighted quantile sum index of REEs mixture]. When stratified by cord blood vitamin D levels, the associations mentioned above persisted in participants with relatively low vitamin D levels (<13.94 μg/L, the first tertile of distribution), but not among those with relatively high levels (≥13.94 μg/L) (all p-values for interaction < 0.05). The mediation analyses taking account of exposure-mediator interaction showed that the relationships between REEs (as individual and mixture) exposure and lower BW-for-GA were partly mediated through decreasing cord blood vitamin D levels. The proportions mediated by cord blood vitamin D levels were 24.48 % for BW-for-GA Z-scores and 29.05 % for SGA corresponding to the REEs mixture exposure. Conclusively, our study revealed that prenatal exposures to Eu, Gd, Dy, Ho, Er, and Yb were related to fetal growth restriction. Cord blood vitamin D might alleviate toxic effects of these REEs and its reduction might partly mediate REE-induced fetal growth restriction.
Collapse
Affiliation(s)
- Xingjie Fang
- Key Laboratory of Environment and Health, Ministry of Education, Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ya Xie
- Key Laboratory of Environment and Health, Ministry of Education, Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuting Cao
- Key Laboratory of Environment and Health, Ministry of Education, Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiangtao Liu
- Key Laboratory of Environment and Health, Ministry of Education, Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yujie Shi
- Key Laboratory of Environment and Health, Ministry of Education, Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling Yu
- Key Laboratory of Environment and Health, Ministry of Education, Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tongzhang Zheng
- Department of Epidemiology, School of Public Health, Brown University, Providence, RI 02912, United States
| | - Hongxiu Liu
- Key Laboratory of Environment and Health, Ministry of Education, Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuanyuan Li
- Key Laboratory of Environment and Health, Ministry of Education, Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shunqing Xu
- Key Laboratory of Environment and Health, Ministry of Education, Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Xia
- Key Laboratory of Environment and Health, Ministry of Education, Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
43
|
Lee TK, Kim YM, Lim HH. Comparison of anthropometric, metabolic, and body compositional abnormalities in Korean children and adolescents born small, appropriate, and large for gestational age: a population-based study from KNHANES V (2010-2011). Ann Pediatr Endocrinol Metab 2024; 29:29-37. [PMID: 38461803 PMCID: PMC10925778 DOI: 10.6065/apem.2346044.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/27/2023] [Accepted: 05/17/2023] [Indexed: 03/12/2024] Open
Abstract
PURPOSE The impacts of growth restriction and programming in the fetal stage on metabolic and bone health in children and adolescents are poorly understood. Moreover, there is insufficient evidence for the relationship between current growth status and metabolic components. Herein, we compared the growth status, metabolic and body compositions, and bone mineral density in Korean children and adolescents based on birth weight at gestational age. METHODS We studied 1,748 subjects (272 small for gestational age [SGA], 1,286 appropriate for gestational age [AGA], and 190 large for gestational age [LGA]; 931 men and 817 women) aged 10-18 years from the Korean National Health and Nutrition Examination Survey (KNHANES) V (2010-2011). Anthropometric measurements, fasting blood biochemistry, and body composition data were analyzed according to birth weight and gestational age. RESULTS The prevalence of low birth weight (14.7% vs. 1.2% in AGA and 3.2% in LGA, p<0.001) and current short stature (2.237 [1.296-3.861] compared to AGA, p=0.004) in SGA subjects was greater than that in other groups; however, the prevalence of overweight and obesity risks, metabolic syndrome (MetS), and MetS component abnormalities was not. Moreover, no significant differences were found in age- and sex-adjusted lean mass ratio, fat mass ratio, truncal fat ratio, bone mineral content, or bone density among the SGA, AGA, and LGA groups in Korean children and adolescents. CONCLUSION Our data demonstrate that birth weight alone may not be a determining factor for body composition and bone mass in Korean children and adolescents. Further prospective and longitudinal studies in adults are necessary to confirm the impact of SGA on metabolic components and bone health.
Collapse
Affiliation(s)
- Tae Kwan Lee
- Department of Pediatrics, Chungnam National University Hospital, Daejeon, Korea
| | - Yoo Mi Kim
- Department of Pediatrics, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Pediatrics, Chungnam National University Sejong Hospital, Sejong, Korea
| | - Han Hyuk Lim
- Department of Pediatrics, Chungnam National University Hospital, Daejeon, Korea
- Department of Pediatrics, Chungnam National University College of Medicine, Daejeon, Korea
| |
Collapse
|
44
|
Rudar M, Suryawan A, Nguyen HV, Chacko SK, Vonderohe C, Stoll B, Burrin DG, Fiorotto ML, Davis TA. Pulsatile Leucine Administration during Continuous Enteral Feeding Enhances Skeletal Muscle Mechanistic Target of Rapamycin Complex 1 Signaling and Protein Synthesis in a Preterm Piglet Model. J Nutr 2024; 154:505-515. [PMID: 38141773 PMCID: PMC10900192 DOI: 10.1016/j.tjnut.2023.12.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/25/2023] Open
Abstract
BACKGROUND Continuous feeding does not elicit an optimal anabolic response in skeletal muscle but is required for some preterm infants. We reported previously that intermittent intravenous pulses of leucine (Leu; 800 μmol Leu·kg-1·h-1 every 4 h) to continuously fed pigs born at term promoted mechanistic target of rapamycin complex 1 (mTORC1) activation and protein synthesis in skeletal muscle. OBJECTIVES The aim was to determine the extent to which intravenous Leu pulses activate mTORC1 and enhance protein synthesis in the skeletal muscle of continuously fed pigs born preterm. METHODS Pigs delivered 10 d preterm was advanced to full oral feeding >4 d and then assigned to 1 of the following 4 treatments for 28 h: 1) ALA (continuous feeding; pulsed with 800 μmol alanine·kg-1·h-1 every 4 h; n = 8); 2) L1× (continuous feeding; pulsed with 800 μmol Leu·kg-1·h-1 every 4 h; n = 7); 3) L2× (continuous feeding; pulsed with 1600 μmol Leu·kg-1·h-1 every 4 h; n = 8); and 4) INT (intermittent feeding every 4 h; supplied with 800 μmol alanine·kg-1 per feeding; n = 7). Muscle protein synthesis rates were determined with L-[2H5-ring]Phenylalanine. The activation of insulin, amino acid, and translation initiation signaling pathways were assessed by Western blot. RESULTS Peak plasma Leu concentrations were 134% and 420% greater in the L2× compared to the L1× and ALA groups, respectively (P < 0.01). Protein synthesis was greater in the L2× than in the ALA and L1× groups in both the longissimus dorsi and gastrocnemius muscles (P < 0.05) but not different from the INT group (P > 0.10). Amino acid signaling upstream and translation initiation signaling downstream of mTORC1 largely corresponded to the differences in protein synthesis. CONCLUSIONS Intravenous Leu pulses potentiate mTORC1 activity and protein synthesis in the skeletal muscles of continuously fed preterm pigs, but the amount required is greater than in pigs born at term.
Collapse
Affiliation(s)
- Marko Rudar
- Department of Animal Sciences, Auburn University, Auburn, AL, United States
| | - Agus Suryawan
- Department of Pediatrics, USDA/Agricultural Research Service, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States
| | - Hanh V Nguyen
- Department of Pediatrics, USDA/Agricultural Research Service, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States
| | - Shaji K Chacko
- Department of Pediatrics, USDA/Agricultural Research Service, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States
| | - Caitlin Vonderohe
- Department of Pediatrics, USDA/Agricultural Research Service, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States
| | - Barbara Stoll
- Department of Pediatrics, USDA/Agricultural Research Service, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States
| | - Douglas G Burrin
- Department of Pediatrics, USDA/Agricultural Research Service, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States
| | - Marta L Fiorotto
- Department of Pediatrics, USDA/Agricultural Research Service, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States
| | - Teresa A Davis
- Department of Pediatrics, USDA/Agricultural Research Service, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States.
| |
Collapse
|
45
|
Silva-Jose C, May L, Sánchez-Polán M, Zhang D, Barrera-Garcimartín A, Refoyo I, Barakat R. Influence of Physical Activity during Pregnancy on Neonatal Complications: Systematic Review and Meta-Analysis. J Pers Med 2023; 14:6. [PMID: 38276221 PMCID: PMC10820764 DOI: 10.3390/jpm14010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/15/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Newborn hospitalisations after delivery are indicators of poor neonatal health with potential risks of future diseases for children. Interventions to promote a healthy environment have been used during pregnancy, with physical activity as a principal element. A systematic review and meta-analyses were performed to evaluate the effect of physical activity during pregnancy on neonatal intensive care unit (NICU) admissions and Apgar 1 and 5 scores (Registration No.: CRD42022372493). Fifty studies (11,492 pregnant women) were included. There were significantly different rates of NICU admissions between groups (RR = 0.76, 95% CI = 0.62, 0.93; Z = 2.65, p = 0.008; I2 = 0%, and Pheterogeneity = 0.78), and significant differences in Apgar 1 (Z = 2.04; p = 0.04) (MD = 0.08, 95% CI = 0.00, 0.17, I2 = 65%, Pheterogeneity = 0.00001) and Apgar 5 (Z = 3.15; p = 0.002) (MD = 0.09, 95% CI = 0.04, 0.15, I2 = 80%, and Pheterogeneity = 0.00001), favouring intervention groups. Physical activity during pregnancy could help to reduce the risk of NICU admissions that are related to neonatal complications.
Collapse
Affiliation(s)
- Cristina Silva-Jose
- AFIPE Research Group, Faculty of Physical Activity and Sport Sciences-INEF, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (C.S.-J.); (M.S.-P.); (D.Z.); (A.B.-G.)
| | - Linda May
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA;
| | - Miguel Sánchez-Polán
- AFIPE Research Group, Faculty of Physical Activity and Sport Sciences-INEF, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (C.S.-J.); (M.S.-P.); (D.Z.); (A.B.-G.)
| | - Dingfeng Zhang
- AFIPE Research Group, Faculty of Physical Activity and Sport Sciences-INEF, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (C.S.-J.); (M.S.-P.); (D.Z.); (A.B.-G.)
| | - Alejandro Barrera-Garcimartín
- AFIPE Research Group, Faculty of Physical Activity and Sport Sciences-INEF, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (C.S.-J.); (M.S.-P.); (D.Z.); (A.B.-G.)
| | - Ignacio Refoyo
- Sports Department, Faculty of Physical Activity and Sports Sciences-INEF, Universidad Politécnica de Madrid, 28040 Madrid, Spain;
| | - Rubén Barakat
- AFIPE Research Group, Faculty of Physical Activity and Sport Sciences-INEF, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (C.S.-J.); (M.S.-P.); (D.Z.); (A.B.-G.)
| |
Collapse
|
46
|
Xiao H, Chen H, Chen X, Lu Y, Wu B, Wang H, Cao Y, Hu L, Dong X, Zhou W, Yang L. Comprehensive assessment of the genetic characteristics of small for gestational age newborns in NICU: from diagnosis of genetic disorders to prediction of prognosis. Genome Med 2023; 15:112. [PMID: 38093364 PMCID: PMC10717355 DOI: 10.1186/s13073-023-01268-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND In China, ~1,072,100 small for gestational age (SGA) births occur annually. These SGA newborns are a high-risk population of developmental delay. Our study aimed to evaluate the genetic profile of SGA newborns in the newborn intensive care unit (NICU) and establish a prognosis prediction model by combining clinical and genetic factors. METHODS A cohort of 723 SGA and 1317 appropriate for gestational age (AGA) newborns were recruited between June 2018 and June 2020. Clinical exome sequencing was performed for each newborn. The gene-based rare-variant collapsing analyses and the gene burden test were applied to identify the risk genes for SGA and SGA with poor prognosis. The Gradient Boosting Machine framework was used to generate two models to predict the prognosis of SGA. The performance of two models were validated with an independent cohort of 115 SGA newborns without genetic diagnosis from July 2020 to April 2022. All newborns in this study were recruited through the China Neonatal Genomes Project (CNGP) and were hospitalized in NICU, Children's Hospital of Fudan University, Shanghai, China. RESULTS Among the 723 SGA newborns, 88(12.2%) received genetic diagnosis, including 42(47.7%) with monogenic diseases and 46(52.3%) with chromosomal abnormalities. SGA with genetic diagnosis showed higher rates in severe SGA(54.5% vs. 41.9%, P=0.0025) than SGA without genetic diagnosis. SGA with chromosomal abnormalities showed higher incidences of physical and neurodevelopmental delay compared to those with monogenic diseases (45.7% vs. 19.0%, P=0.012). We filtered out 3 genes (ITGB4, TXNRD2, RRM2B) as potential causative genes for SGA and 1 gene (ADIPOQ) as potential causative gene for SGA with poor prognosis. The model integrating clinical and genetic factors demonstrated a higher area under the receiver operating characteristic curve (AUC) over the model based solely on clinical factors in both the SGA-model generation dataset (AUC=0.9[95% confidence interval 0.84-0.96] vs. AUC=0.74 [0.64-0.84]; P=0.00196) and the independent SGA-validation dataset (AUC=0.76 [0.6-0.93] vs. AUC=0.53[0.29-0.76]; P=0.0117). CONCLUSION SGA newborns in NICU presented with roughly equal proportions of monogenic and chromosomal abnormalities. Chromosomal disorders were associated with poorer prognosis. The rare-variant collapsing analyses studies have the ability to identify potential causative factors associated with growth and development. The SGA prognosis prediction model integrating genetic and clinical factors outperformed that relying solely on clinical factors. The application of genetic sequencing in hospitalized SGA newborns may improve early genetic diagnosis and prognosis prediction.
Collapse
Affiliation(s)
- Hui Xiao
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Huiyao Chen
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Xiang Chen
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Yulan Lu
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Bingbing Wu
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Huijun Wang
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Yun Cao
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Liyuan Hu
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Xinran Dong
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China.
| | - Wenhao Zhou
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China.
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China.
- Shanghai Key Laboratory of Birth Defects, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China.
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510005, China.
| | - Lin Yang
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China.
| |
Collapse
|
47
|
Zhang B, Xi S, Liu R, Han X, Long W, Yuan X, Yu B. Maternal fibrinogen/fibrin degradation products to high density lipoprotein cholesterol ratio for predicting delivery of small and large for gestational age infants: a pilot study. Lipids Health Dis 2023; 22:221. [PMID: 38087267 PMCID: PMC10714553 DOI: 10.1186/s12944-023-01986-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The purpose of this pilot study was to investigate associations between fibrinogen/fibrin degradation products (FDP) to high density lipoprotein-cholesterol (HDL-C) ratio (FHR) of mothers and the risk of delivering large/small for gestational age (LGA/SGA) infants and to evaluate the predictive power of FHR on LGA/SGA. METHODS This study retrospectively reviewed 11,657 consecutive women whose lipid profiles and FDP levels were investigated at the time of admission for delivery at a specialized hospital. The FHR was calculated, and perinatal outcomes, including clinical parameters, were analyzed. RESULTS The prevalence of SGA was 9% (n = 1034), and that of LGA was 15% (n = 1806) in this cohort study. FHR was significantly lower in women who delivered SGA infants (4.0 ± 3.2 vs. 4.7 ± 3.3 mg/mmol, P < 0.01) and higher in women who delivered LGA infants (5.7 ± 3.8 vs. 4.7 ± 3.3 mg/mmol, P < 0.01) compared with those who delivered infants of normal size for their gestational age. Women in the top quartile for FHR (> 5.9 mg/mmol) had a 2.9-fold higher risk of delivering LGA infants [adjusted odds ratio (OR) = 2.9, P < 0.01] and a 47% lower risk of delivering SGA infants (adjusted OR = 0.47, P < 0.01) than those in the bottom quartile (< 2.7 mg/mmol). In addition, adding FHR to the conventional models significantly improved the area under the curve for the prediction of delivering LGA (0.725 vs. 0.739, P < 0.01) and SGA (0.717 vs. 0.727, P < 0.01) infants. CONCLUSION These findings suggest that the FHR calculated in late pregnancy is an innovative predictor of delivering LGA and SGA infants. Combining FHR with perinatal parameters could thus enhance the predictive ability for predicting the delivery of LGA/SGA infants.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Medical Genetics, Changzhou Medical Center, Changzhou Maternal and Child Health Care Hospital, Nanjing Medical University, 16th Ding Xiang Road, Changzhou, 213023, Jiangsu, China
| | - Sijie Xi
- Department of Medical Genetics, Changzhou Medical Center, Changzhou Maternal and Child Health Care Hospital, Nanjing Medical University, 16th Ding Xiang Road, Changzhou, 213023, Jiangsu, China
| | - Renchen Liu
- General Education College, Anhui Institute of Information Technology, Wuhu, China
| | - Xiaoya Han
- Department of Medical Genetics, Changzhou Medical Center, Changzhou Maternal and Child Health Care Hospital, Nanjing Medical University, 16th Ding Xiang Road, Changzhou, 213023, Jiangsu, China
| | - Wei Long
- Department of Medical Genetics, Changzhou Medical Center, Changzhou Maternal and Child Health Care Hospital, Nanjing Medical University, 16th Ding Xiang Road, Changzhou, 213023, Jiangsu, China
| | - Xiaosong Yuan
- Department of Medical Genetics, Changzhou Medical Center, Changzhou Maternal and Child Health Care Hospital, Nanjing Medical University, 16th Ding Xiang Road, Changzhou, 213023, Jiangsu, China.
| | - Bin Yu
- Department of Medical Genetics, Changzhou Medical Center, Changzhou Maternal and Child Health Care Hospital, Nanjing Medical University, 16th Ding Xiang Road, Changzhou, 213023, Jiangsu, China.
| |
Collapse
|
48
|
Gleason JL, Yeung EH, Sundaram R, Putnick DL, Mendola P, Bell EM, Polinski KJ, Robinson SL, Grantz KL. Longitudinal Child Growth Patterns in Twins and Singletons in the Upstate KIDS Cohort. J Pediatr 2023; 263:113720. [PMID: 37660974 PMCID: PMC10872829 DOI: 10.1016/j.jpeds.2023.113720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/11/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
OBJECTIVES To investigate childhood growth patterns in twins and to determine whether they show the same signs of excess growth as singletons born small-for-gestational age (SGA), which may confer future cardiometabolic risk. STUDY DESIGN In the Upstate KIDS cohort of infants delivered from 2008 through 2010, we compared height, weight, and body mass index (BMI) z-scores at 0-3 and 7-9 years of age, as well as risk of rapid weight gain (RWG) in infancy and overweight/obesity beginning at 2 years, among appropriate-for-gestational age (AGA) twins (n = 1121), AGA singletons (n = 2684), and two groups of SGA twins: uncertain SGA twins (<10th percentile for birthweight by a singleton reference but >10th% by a population-based twin birthweight reference; n = 319) and true SGA twins (<10th% by a population-based twin reference; n = 144). RESULTS Compared with AGA twins, both SGA twin groups had lower weight and BMI z-scores at both time points. By 7-9 years, both groups caught up in height with AGA twins. Compared with AGA singletons, z-score differences decreased between 0-3 and 7-9 years for uncertain SGA and true SGA twins, though true SGA twins had the lowest z-scores for all measures. During infancy, twins were more likely to display RWG compared with AGA singletons (RR = 2.06 to 2.67), which may reflect normal catch-up growth, as no twin group had higher prevalence of overweight/obesity at either time point. CONCLUSIONS Though twins had lower height, weight, and BMI z-scores at birth and into toddlerhood, differences were reduced by 7-9 years, with no evidence of pathological growth and no group of twins showing elevated risk of overweight/obesity.
Collapse
Affiliation(s)
- Jessica L Gleason
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Edwina H Yeung
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Rajeshwari Sundaram
- Biostatistics and Bioinformatics Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Diane L Putnick
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Pauline Mendola
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY
| | - Erin M Bell
- Departments of Environmental Health Sciences and Epidemiology and Biostatistics University at Albany, School of Public Health, Rensselaer, NY
| | - Kristen J Polinski
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Sonia L Robinson
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Katherine L Grantz
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD.
| |
Collapse
|
49
|
Dominguez-Menendez G, Poggi H, Ochoa-Molina F, D'Apremont I, Moore R, Allende F, Solari S, Martinez-Aguayo A. Extremely and very preterm children who were born appropriate for gestational age show no differences in cortisol concentrations or diurnal rhythms compared to full-term children. J Pediatr Endocrinol Metab 2023; 36:1018-1027. [PMID: 37795843 DOI: 10.1515/jpem-2023-0194] [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] [Received: 04/29/2023] [Accepted: 09/18/2023] [Indexed: 10/06/2023]
Abstract
OBJECTIVES The objective of this study was to compare the diurnal variations in cortisol and cortisone concentrations in serum and saliva among extremely preterm (EPT), very preterm (VPT), and full-term (FT) children, all born appropriate for gestational age (AGA). METHODS EPT, VPT, and FT children, all born AGA, were recruited from two healthcare centers. Cortisol and cortisone concentrations in serum and saliva were measured by liquid chromatography-mass spectrometry (LC‒MS). Statistical analysis was performed using nonparametric tests. RESULTS A total of 101 children (5.0-8.9 years old) were included in this study: EPT=18, VPT=43 and FT=40. All groups had similar distributions in terms of age, birth weight standard deviation score (SDS) and BMI (SDS), showing no differences in serum ACTH, cortisol, or cortisone levels. Additionally, salivary cortisol and cortisone concentrations decreased significantly throughout the day (p-values<0.0001). Salivary cortisol concentrations were below the limit of detection (0.55 nmol/L) before dinner and before bedtime in approximately one-third and two-thirds of all children, respectively. Salivary cortisone was detectable in all but one sample. CONCLUSIONS The diurnal cortisol rhythm was preserved in all preterm children, regardless of their gestational age, and no differences in cortisol concentrations among the groups were found. This may have significant implications for the clinical management and follow-up of preterm individuals.
Collapse
Affiliation(s)
- Gonzalo Dominguez-Menendez
- Endocrinology Division, Department of Paediatric, University of British Columbia/BC Children's Hospital, Vancouver, BC, Canada
| | - Helena Poggi
- Endocrinology Units, Paediatrics Division, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Fernanda Ochoa-Molina
- Endocrinology Units, Paediatrics Division, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Ivonne D'Apremont
- Endocrinology Units, Paediatrics Division, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Rosario Moore
- Endocrinology Units, Paediatrics Division, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Fidel Allende
- Department of Clinical Laboratories, School of Medicine, Pontificia Universidad Catolica de Chile, Macul, Chile
| | - Sandra Solari
- Department of Clinical Laboratories, School of Medicine, Pontificia Universidad Catolica de Chile, Macul, Chile
| | - Alejandro Martinez-Aguayo
- Endocrinology Units, Paediatrics Division, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| |
Collapse
|
50
|
Wei Q, Lin W, Zhang H, Lai Y, Zhuang S, Han Z, Wang Q, Wang L, Li W, Wen L, Hou H, Hu Q. Role of antenatal anxiety in the relationship between maternal exposure to nitrogen dioxide and small for gestational age: A birth cohort study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 900:165812. [PMID: 37499810 DOI: 10.1016/j.scitotenv.2023.165812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/04/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Both Nitrogen dioxide (NO2) exposure and antenatal anxiety have individually been associated with small for gestational age (SGA). Little is known, however, about whether there is effect modification of antenatal anxiety on NO2-related SGA. METHODS The prospective birth cohort study included 1823 mother-newborn pairs in Guangzhou, China, from January 2017 to April 2020. Exposure to NO2 during the pre-conceptional and prenatal periods was estimated using an inverse distance weighted method. Antenatal anxiety was assessed by Trait Anxiety Inventory. SGA was determined by the Chinese gestational age- and sex-specific birthweight standards. Cox proportional hazards regression models was used to estimate hazard ratios (HRs) and 95 % confidence intervals (CIs) for SGA as per 10 μg/m3 increase in NO2. Modifying effects of trait anxiety on NO2-related SGA were identified by stratified analyses, and three-dimensional response surface plots and two-dimensional heat maps. RESULTS Each 10 μg/m3 increase in NO2 exposure during the third trimester was significantly associated with SGA risk among overall participants (HR = 1.221, 95 % CI: 1.014-1.471) and primipara (HR = 1.271, 95 % CI: 1.023-1.579). We found significant effect modification of anxiety level for NO2-related SGA in the third trimester (Pinteraction < 0.05). Pregnant women with higher levels of trait anxiety were more likely to deliver SGA newborns, particularly for those with high trait anxiety (HR = 1.781, 95 % CI: 1.007-2.945). Primiparous women were more susceptible. CONCLUSIONS This study provides evidence that antenatal trait anxiety may modify the effects of maternal NO2 exposure on SGA risk. The third trimester could be a critical window of susceptibility.
Collapse
Affiliation(s)
- Qiannan Wei
- Department of Occupational and Environmental Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Weiwei Lin
- Department of Occupational and Environmental Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Hedi Zhang
- Department of Occupational and Environmental Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yuming Lai
- Department of Occupational and Environmental Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Shuling Zhuang
- Department of Occupational and Environmental Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Zhenyan Han
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Qingqing Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Lijie Wang
- Department of Occupational and Environmental Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Wenzhuo Li
- Department of Occupational and Environmental Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Li Wen
- Department of Occupational and Environmental Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Hongying Hou
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Qiansheng Hu
- Department of Occupational and Environmental Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| |
Collapse
|