1
|
Nyakotey DA, Gamble GD, McKinlay CJD, Bloomfield FH, Harding JE. Associations between growth and childhood body composition in very preterm, late preterm and term children. Acta Paediatr 2025; 114:1030-1042. [PMID: 39652519 PMCID: PMC11978493 DOI: 10.1111/apa.17534] [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: 06/25/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 04/09/2025]
Abstract
AIM To determine how gestational age at birth and postnatal growth relate to body composition in childhood. METHODS We calculated conditional growth (birth-2 years, 2 years-6 years) and measured body composition at 2 and 6 years using bioelectrical impedance in cohorts of New Zealand children born very preterm (VPT; 23-31 weeks), late preterm (LPT; 35- <37 weeks) and term (≥37 weeks). We explored the relationships between growth and fat mass (FM) index and fat-free mass (FFM) index at 6 years using multivariable linear regression. RESULTS Of 1125 children (51% male), 202 were VPT, 114 LPT and 809 Term. Compared to Term, VPT but not LPT were lighter and shorter at 2 and 6 years and had lower FM index and FFM index. The association between weight growth from 2 to 6 years and both FM index and FFM index at 6 years was stronger than for weight growth from birth to 2 years or height growth at any period in all gestational age groups. CONCLUSIONS Size and body composition at 2 and 6 years are different between infants born VPT, but not LPT, and at term. Later weight growth is more strongly associated with childhood body composition than earlier growth.
Collapse
Affiliation(s)
| | - Greg D. Gamble
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Christopher J. D. McKinlay
- Counties Manukau Health, Kidz First Neonatal Care, Auckland, New Zealand
- Department of Paediatrics: Child and Youth Health, University of Auckland, Auckland, New Zealand
| | | | - Jane E. Harding
- Liggins Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
2
|
Kim JH, Kim SJ, Chung IH, Lim JS. Adiposity Rebound Timing in Small for Gestational Age Children Treated With Growth Hormone: Results From LG Growth Study Data. J Korean Med Sci 2025; 40:e12. [PMID: 39901524 PMCID: PMC11790395 DOI: 10.3346/jkms.2025.40.e12] [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: 04/19/2024] [Accepted: 09/30/2024] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Adiposity rebound (AR) refers to the period during growth when the body mass index reaches its lowest point before increasing again. The timing of AR is associated with the development of obesity and puberty onset. Although studies have evaluated AR timing in Korean children, none has focused on children born small for gestational age (SGA). METHODS This study analyzed data from a multicenter observational clinical trial (LG Growth Study) to determine AR timing in children born SGA without catch-up growth (CUG) who were treated with growth hormone (GH) therapy. The study also aimed to identify factors associated with AR timing, examine the influence of AR timing on puberty onset, and assess the effectiveness of GH therapy. RESULTS A total of 151 children born SGA without CUG were included. Of them, 15% experienced AR between 4 and 5 years of age, 42% between 5 and 6 years, 27% between 6 and 7 years, and 16% after 7 years of age. A significant positive correlation was noted between the height standard deviation score at the start of treatment and AR timing. However, no significant correlation was observed between AR timing and puberty onset or the effectiveness of GH therapy. CONCLUSION This study provides insights into AR timing in prepubertal children who meet the specific SGA criteria and its relationship with growth outcomes. The findings suggest that AR in children born SGA who do not experience CUG occurs later than in the general population, with no significant relationship between AR timing and puberty onset or growth outcomes. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01604395.
Collapse
Affiliation(s)
- Ji Hyun Kim
- Department of Pediatrics, Dongguk University Ilsan Hospital, Goyang, Korea
| | - Su Jin Kim
- Department of Pediatrics, Inha University Hospital, Inha University College of Medicine, Incheon, Korea
| | - In Hyuk Chung
- Department of Pediatrics, National Health Insurance Service Ilsan Hospital, Goyang, Korea
| | - Jung Sub Lim
- Department of Pediatrics, Korea Cancer Center Hospital, Seoul, Korea.
| |
Collapse
|
3
|
Choe Y, Kim KN, Lee YJ, Kim JI, Kim BN, Lim YH, Hong YC, Shin CH, Lee YA. Prenatal and childhood exposure to endocrine-disrupting chemicals and early thelarche in 8-year-old girls: A prospective study using Bayesian kernel regression. ENVIRONMENTAL RESEARCH 2024; 263:120056. [PMID: 39343343 DOI: 10.1016/j.envres.2024.120056] [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: 05/22/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Studies on the combined effects of persistent and non-persistent endocrine-disrupting chemicals (EDCs) on puberty are insufficient. To date, no studies have analyzed breast development at age 8 years, a key criterion for determining precocious puberty. We investigated the relationship between prenatal or childhood exposure to EDC mixtures and early thelarche, defined as breast development before age 8 years in girls. METHODS This prospective study included 211 girls with data on prenatal and 8-year-old exposure of cadmium (Cd), lead, mercury, bisphenol-A (BPA), 3-phenoxybenzoic acid, and three phthalate metabolites from the Environment and Development of Children cohort. Prenatal exposure was assessed through samples from pregnant women at 14-27th weeks of gestation. Tanner staging was assessed by a pediatric endocrinologist. The relationship between single and mixed chemical exposures and outcomes was assessed using logistic regression, generalized additive models (GAM), and Bayesian kernel machine regression (BKMR) models. RESULTS Early thelarche was observed in 42 (19.9%) girls at age 8 years. In the logistic regression models, the risk of early thelarche increased with increased exposure to Cd in their mothers (adjusted odds ratio [aOR] per interquartile range [IQR] = 1.80, 95% confidence interval [CI] 1.23-2.65) but decreased with prenatal BPA exposure (aOR per IQR = 0.57, 95% CI 0.35-0.92). None of the 8-year-old chemical exposures was associated with early thelarche. In the GAM, early thelarche was positively correlated with prenatal Cd and inversely associated with prenatal BPA exposure (p = 0.004 for Cd and p = 0.036 for BPA). In the BKMR models, an increase in log-transformed prenatal Cd concentrations from the 25th to 75th percentile was associated with an increase in the estimated probability of early thelarche at age 8 years (risk difference: 0.46 [95% credible interval: 0.04-0.88]) when other chemicals were set at their median values. CONCLUSIONS Considering the combined effects of persistent and non-persistent chemical mixtures, maternal Cd exposure during the second trimester may be associated with early thelarche in 8-year-old girls.
Collapse
Affiliation(s)
- Yunsoo Choe
- Department of Pediatrics, Hanyang University Guri Hospital, Guri, South Korea; Department of Pediatrics, Hanyang University College of Medicine, Seoul, South Korea
| | - Kyoung-Nam Kim
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Yun Jeong Lee
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, South Korea
| | - Johanna Inhyang Kim
- Department of Psychiatry, Hanyang University College of Medicine, Seoul, South Korea
| | - Bung-Nyun Kim
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, South Korea
| | - Youn-Hee Lim
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, South Korea; Section of Environmental Health, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Yun-Chul Hong
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Human Systems Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Choong Ho Shin
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, South Korea
| | - Young Ah Lee
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
4
|
Benonisdottir S, Straub VJ, Kong A, Mills MC. Genetics of female and male reproductive traits and their relationship with health, longevity and consequences for offspring. NATURE AGING 2024; 4:1745-1759. [PMID: 39672892 DOI: 10.1038/s43587-024-00733-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/26/2024] [Indexed: 12/15/2024]
Abstract
Substantial shifts in reproductive behaviors have recently taken place in many high-income countries including earlier age at menarche, advanced age at childbearing, rising childlessness and a lower number of children. As reproduction shifts to later ages, genetic factors may become increasingly important. Although monogenic genetic effects are known, the genetics underlying human reproductive traits are complex, with both causal effects and statistical bias often confounded by socioeconomic factors. Here, we review genome-wide association studies (GWASs) of 44 reproductive traits of both female and male individuals from 2007 to early 2024, examining reproductive behavior, reproductive lifespan and aging, infertility and hormonal concentration. Using the GWAS Catalog as a basis, from 159 relevant studies, we isolate 37 genes that harbor association signals for four or more reproductive traits, more than half of which are linked to rare Mendelian disorders, including ten genes linked to reproductive-related disorders: FSHB, MCM8, DNAH2, WNT4, ESR1, IGSF1, THRB, BRWD1, CYP19A1 and PTPRF. We also review the relationship of reproductive genetics to related health and behavioral traits, aging and longevity and the effect of parental age on offspring outcomes as well as reflecting on limitations, open questions and challenges in this fast-moving field.
Collapse
Affiliation(s)
- Stefania Benonisdottir
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK
- Institute of Physical Science, University of Iceland, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Vincent J Straub
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK
| | - Augustine Kong
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK
| | - Melinda C Mills
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK.
- Department of Genetics, University Medical Centre Groningen, Groningen, the Netherlands.
- Department of Economics, Econometrics and Finance, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
5
|
Chen Y, Zhang M, Wang G, Hong X, Wang X, Mueller NT. Mother's age at menarche is associated with odds of preterm delivery: A case-control study. BJOG 2024; 131:424-432. [PMID: 37661294 PMCID: PMC10872971 DOI: 10.1111/1471-0528.17648] [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: 02/23/2023] [Revised: 07/31/2023] [Accepted: 08/15/2023] [Indexed: 09/05/2023]
Abstract
OBJECTIVE There is a secular trend towards earlier age of menarche in the US and globally. Earlier age at menarche (AAM) has been associated with metabolic disorders that increase risk for preterm delivery (PTD), yet no studies in the US have investigated whether AAM influences risk of PTD. This study tested the hypothesis that AAM is associated with PTD. DESIGN A case-control study. SETTING The Boston Medical Center (BMC) in Boston, Massachusetts. POPULATION OR SAMPLE 8264 mother-newborn dyads enrolled at birth at BMC between 1998 and 2019, of which 2242 mothers had PTD (cases) and 6022 did not have PTD (controls). METHODS Multivariable-adjusted logistic regression models and restricted cubic splines were used to examine the association between AAM and risk of PTD. The combined impact of AAM and age at delivery on the risk of PTD was also examined. MAIN OUTCOME MEASURES Preterm delivery and gestational age (GA) was defined by maternal last menstrual period and early ultrasound documented in medical records. RESULTS Maternal age at delivery was 28.1 ± 6.5 years and AAM was 12.85 ± 1.86 years. Multivariable-adjusted cubic spline suggested an inverse dose-response association of AAM with odds of PTD and, consistently, a positive association with GA. A 1-year earlier AAM was associated with 5% (95% CI 2%-8%) higher odds of PTD, after adjustment for maternal year of birth, parity, maternal place of birth, education, smoking status and Mediterranean-style diet score. The association between AAM and PTD was stronger among older mothers whose age at delivery was ≥35 years. CONCLUSIONS Earlier AAM is associated with higher odds for PTD, and this association is stronger among women at advanced reproductive age.
Collapse
Affiliation(s)
- Yingan Chen
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, MD
- Lifecourse Epidemiology of Adiposity & Diabetes Center, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Mingyu Zhang
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA
| | - Guoying Wang
- Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Xiumei Hong
- Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Xiaobin Wang
- Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Noel T. Mueller
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, MD
- Lifecourse Epidemiology of Adiposity & Diabetes Center, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Pediatrics, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
6
|
Yue M, Zhang L. Exploring the Mechanistic Interplay between Gut Microbiota and Precocious Puberty: A Narrative Review. Microorganisms 2024; 12:323. [PMID: 38399733 PMCID: PMC10892899 DOI: 10.3390/microorganisms12020323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
The gut microbiota has been implicated in the context of sexual maturation during puberty, with discernible differences in its composition before and after this critical developmental stage. Notably, there has been a global rise in the prevalence of precocious puberty in recent years, particularly among girls, where approximately 90% of central precocious puberty cases lack a clearly identifiable cause. While a link between precocious puberty and the gut microbiota has been observed, the precise causality and underlying mechanisms remain elusive. This narrative review aims to systematically elucidate the potential mechanisms that underlie the intricate relationship between the gut microbiota and precocious puberty. Potential avenues of exploration include investigating the impact of the gut microbiota on endocrine function, particularly in the regulation of hormones, such as gonadotropin-releasing hormone (GnRH), luteinizing hormone (LH), and follicle-stimulating hormone (FSH). Additionally, this review will delve into the intricate interplay between the gut microbiome, metabolism, and obesity, considering the known association between obesity and precocious puberty. This review will also explore how the microbiome's involvement in nutrient metabolism could impact precocious puberty. Finally, attention is given to the microbiota's ability to produce neurotransmitters and neuroactive compounds, potentially influencing the central nervous system components involved in regulating puberty. By exploring these mechanisms, this narrative review seeks to identify unexplored targets and emerging directions in understanding the role of the gut microbiome in relation to precocious puberty. The ultimate goal is to provide valuable insights for the development of non-invasive diagnostic methods and innovative therapeutic strategies for precocious puberty in the future, such as specific probiotic therapy.
Collapse
Affiliation(s)
- Min Yue
- Microbiome-X, National Institute of Health Data Science of China & Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lei Zhang
- Microbiome-X, National Institute of Health Data Science of China & Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| |
Collapse
|
7
|
Oyama S, Duckham RL, Pomer A, Rivara AC, Kershaw EE, Wood A, Fidow UT, Naseri T, Reupena MS, Viali S, McGarvey ST, Hawley NL. Association between age at menarche and cardiometabolic risk among Samoan adults. Am J Hum Biol 2024; 36:e23982. [PMID: 37668413 PMCID: PMC10845161 DOI: 10.1002/ajhb.23982] [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: 05/10/2023] [Revised: 07/19/2023] [Accepted: 08/15/2023] [Indexed: 09/06/2023] Open
Abstract
OBJECTIVES Recent studies suggest that early menarche may increase cardiometabolic morbidity and mortality. Yet few studies have examined this association in the Pacific Islands, where obesity prevalence is among the highest globally. We sought to examine associations between age at menarche and cardiometabolic risk in Samoa. METHODS Participants were from the Soifua Manuia study (n = 285, age 32-72 years) conducted in Samoa from 2017 to 2019. Logistic regressions were conducted to estimate odds of obesity, hypertension, diabetes, dyslipidemia, and metabolic syndrome per one-year increase in age at menarche. Linear regressions were conducted to examine associations between age at menarche and continuous measures of adiposity, blood pressure, insulin resistance, and serum lipids. RESULTS Median age at menarche was 14 years (IQR = 2). After controlling for relevant covariates, each one-year increase in age at menarche was associated with a 15% decrease (OR = 0.85, 95% CI: 0.72-1.01, p = .067) in odds of hypertension, but a 21% increase (OR = 1.21, 95% CI: 1.01-1.45, p = .044) in odds of diabetes and 18% increase (OR = 1.18, 95% CI: 0.98-1.42, p = .081) in odds of high total cholesterol. Each additional year in age at menarche was associated with a 1.60 ± 0.52 kg (p = .002) decrease in lean mass and 1.56 ± 0.51 kg (p = .003) decrease in fat-free mass. CONCLUSIONS Associations between age at menarche and cardiometabolic risk may be population-specific and are likely influenced by both current and historical nutritional and epidemiological contexts. Prospective studies are needed to clarify the role of childhood adiposity and other early life exposures on age at menarche and subsequent cardiometabolic risk.
Collapse
Affiliation(s)
- Sakurako Oyama
- Yale School of Medicine, New Haven, Connecticut, USA
- Department of Anthropology, Yale University, New Haven, Connecticut, USA
| | - Rachel L Duckham
- Institute for Physical Activity and Nutrition (IPAN), Deakin University, Burwood, Victoria, Australia
- Australian Institute for Musculoskeletal Sciences, Department of Medicine, Western Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Alysa Pomer
- Center for Surgery and Public Health, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Anna C Rivara
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut, USA
| | - Erin E Kershaw
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ashlee Wood
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ulai T Fidow
- Department of Obstetrics & Gynecology, Tupua Tamasese Meaole Hospital, Apia, Samoa
| | | | | | | | - Stephen T McGarvey
- International Health Institute, Department of Epidemiology, Department of Anthropology, Brown University, Providence, Rhode Island, USA
| | - Nicola L Hawley
- Department of Anthropology, Yale University, New Haven, Connecticut, USA
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut, USA
| |
Collapse
|
8
|
Calcaterra V, Cena H, Sottotetti F, Rossi V, Loperfido F, Zuccotti G. Breast and Formula Milk and Early Puberty Onset. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1686. [PMID: 37892349 PMCID: PMC10605833 DOI: 10.3390/children10101686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/12/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023]
Abstract
Nutrients have an enormous impact on many hormonal systems and aspects of health, and nutrition status is a crucial regulator of growth and pubertal development in children and adolescents. In this narrative review, we explore the connection between these feeding methods and the timing of puberty to provide a clearer understanding of how infant nutrition might contribute to the early development of puberty. Puberty is a key stage in the transition from childhood to adulthood and the timing of puberty represents a significant biological milestone of growth. Breast milk seems to have a pivotal role in puberty onset, mainly due to its dynamism, which shape indirectly the gut microbiota in early life, besides direct exposure of the baby to the milk microbiota through gut-breast axis. Concerning breast and formula milk and their effects on the onset of puberty, a protective role of the former occurs. As for the potential harmful effects of soy-based formulas and the isoflavones that they contain, the studies reported demonstrate conflicting opinions, underlining the need for further research on this topic. A healthy and well-nourished diet from the earliest stages of life has significant preventive potential for overall well-being, reducing the risk of many health problems later in life.
Collapse
Affiliation(s)
- Valeria Calcaterra
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy;
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.R.); (G.Z.)
| | - Hellas Cena
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (F.S.); (F.L.)
- Clinical Nutrition Unit, General Medicine, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | - Francesca Sottotetti
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (F.S.); (F.L.)
| | - Virginia Rossi
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.R.); (G.Z.)
| | - Federica Loperfido
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (F.S.); (F.L.)
| | - Gianvincenzo Zuccotti
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.R.); (G.Z.)
- Department of Biomedical and Clinical Science, University of Milano, 20157 Milano, Italy
| |
Collapse
|
9
|
Xu Y, Xiong J, Shan S, Wang X, He F, Cheng G. Age-Dependent and Body Composition-Dependent Association of Child Gut Microbial Enterotype With Puberty Timing: A Chinese Cohort. J Clin Endocrinol Metab 2023; 108:2363-2370. [PMID: 36840481 PMCID: PMC10438909 DOI: 10.1210/clinem/dgad090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/20/2023] [Accepted: 02/10/2023] [Indexed: 02/26/2023]
Abstract
CONTEXT Puberty timing, which is vital for adult well-being, has recently been suggested to be linked to specific gut taxa. However, the impact of comprehensive gut microbiome structure assessed by enterotype on puberty timing remains unknown. OBJECTIVE Investigate the prospective association of gut microbial enterotype with puberty timing and the potential interaction of age and body composition. METHODS This study included 1826 children from the Chinese Adolescent Cohort Study, a cohort that has collected information on sociodemographics, dietary intake, physical activity, anthropometry, and pubertal development of children aged 6-8 years since 2013 and follows them up annually until the age of 15 years. Fecal samples have been collected annually since 2019 and analyzed for 16S rRNA sequencing and targeted fecal metabolomics. Cox proportional hazard regression models were used to investigate the prospective association of enterotype with puberty timing and the impact of age and body mass index (BMI) sex- and age-independent standard deviation score (SDS). RESULTS 592 (32.4%) and 1234 (67.6%) children belonged to the Prevotella-rich enterotype and the Bacteroides-rich enterotype, respectively. Children with the Bacteroides-rich enterotype experienced their menarche/voice break later than those with the Prevotella enterotype (hazard ratio 0.53, 95% CI 0.28-0.98), P = .02). Moreover, this association was more pronounced among younger children with higher BMI SDS (P for interaction = .006). CONCLUSION Our findings supported a role for gut microbial communities in pubertal development, in which younger children with higher body mass seems more sensitive.
Collapse
Affiliation(s)
- Yujie Xu
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Jingyuan Xiong
- West China School of Public Health and West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610041, P.R. China
| | - Shufang Shan
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Xiaoyu Wang
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Fang He
- West China School of Public Health and West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610041, P.R. China
| | - Guo Cheng
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| |
Collapse
|
10
|
Gilley SP, Harrall KK, Friedman C, Glueck DH, Cohen CC, Perng W, Sauder KA, Krebs NF, Shankar K, Dabelea D. Association of Maternal BMI and Rapid Infant Weight Gain With Childhood Body Size and Composition. Pediatrics 2023; 151:e2022059244. [PMID: 37016999 PMCID: PMC11033707 DOI: 10.1542/peds.2022-059244] [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] [Accepted: 01/27/2023] [Indexed: 04/06/2023] Open
Abstract
OBJECTIVES Maternal prepregnancy BMI (ppBMI) and an infant's rapid weight gain (RWG) are each associated with increased risk for childhood obesity. We hypothesized that ppBMI and RWG interact to further raise childhood obesity risk. METHODS Mother-infant dyads (n = 414) from the Healthy Start Study, an observational prebirth cohort, were included. RWG was defined as a weight-for-age z score increase of ≥0.67 from birth to 3 to 7 months. Body composition was measured by air displacement plethysmography at age 4 to 7 years. General linear regression models were fit to characterize associations between ppBMI, RWG, and their interaction with the outcomes of childhood BMI-for-age z score and percent fat mass (%FM). RESULTS A total of 18.6% (n = 77) of offspring experienced RWG. Maternal ppBMI and RWG were both positively associated with offspring BMI z score and %FM. RWG amplified the association between ppBMI and BMI z score, especially among females. Females exposed to maternal obesity and RWG had an average BMI at the 94th percentile (1.50 increase in childhood BMI z score) compared with those exposed to normal ppBMI and no RWG (average childhood BMI at the 51st percentile). RWG had a weaker effect on the association between ppBMI and %FM. Adjustment for breastfeeding status or childhood daily caloric intake did not significantly alter findings. CONCLUSIONS Rapid infant weight gain interacts with maternal ppBMI to jointly exacerbate risk of childhood obesity. Pediatric providers should monitor infants for RWG, especially in the context of maternal obesity, to reduce future risk of obesity.
Collapse
Affiliation(s)
- Stephanie P. Gilley
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, Colorado
| | - Kylie K. Harrall
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado Anschutz Medical Campus, and Aurora, Colorado
| | - Chloe Friedman
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado Anschutz Medical Campus, and Aurora, Colorado
| | - Deborah H. Glueck
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, Colorado
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado Anschutz Medical Campus, and Aurora, Colorado
| | - Catherine C. Cohen
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, Colorado
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado Anschutz Medical Campus, and Aurora, Colorado
| | - Wei Perng
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado Anschutz Medical Campus, and Aurora, Colorado
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Denver, Colorado
| | - Katherine A. Sauder
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, Colorado
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado Anschutz Medical Campus, and Aurora, Colorado
| | - Nancy F. Krebs
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, Colorado
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, Colorado
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado Anschutz Medical Campus, and Aurora, Colorado
| | - Dana Dabelea
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, Colorado
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado Anschutz Medical Campus, and Aurora, Colorado
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Denver, Colorado
| |
Collapse
|
11
|
Feig DS, Sanchez JJ, Murphy KE, Asztalos E, Zinman B, Simmons D, Haqq AM, Fantus IG, Lipscombe L, Armson A, Barrett J, Donovan L, Karanicolas P, Tobin S, Mangoff K, Klein G, Jiang Y, Tomlinson G, Hamilton J. Outcomes in children of women with type 2 diabetes exposed to metformin versus placebo during pregnancy (MiTy Kids): a 24-month follow-up of the MiTy randomised controlled trial. Lancet Diabetes Endocrinol 2023; 11:191-202. [PMID: 36746160 DOI: 10.1016/s2213-8587(23)00004-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Metformin is increasingly being used during pregnancy, with potentially adverse long-term effects on children. We aimed to examine adiposity in children of women with type 2 diabetes from the Metformin in Women with Type 2 Diabetes in Pregnancy (MiTy) trial, with and without in-utero exposure to metformin, up to 24 months of age. METHODS MiTy Kids is a follow-up study that included infants of women who participated in the MiTy randomised controlled trial, receiving either oral 1000 mg metformin twice daily or placebo. Caregivers and researchers remained masked to the type of medication (metformin or placebo) mothers received during their pregnancy. Anthropometric measurements, including weight, height, and skinfold thicknesses, were taken at 3, 6, 12, 18, and 24 months. At 24 months, linear regression was used to compare the BMI Z score and sum of skinfolds in the metformin versus placebo groups, adjusted for confounders. Fractional polynomials were used to assess growth trajectories. This study is registered with ClinicalTrials.gov, NCT01832181. FINDINGS Of the 465 eligible children, 283 (61%) were included from 19 centres in Canada and Australia. At 24 months, there was no difference between groups in mean BMI Z score (0·84 [SD 1·52] with metformin vs 0·91 [1·38] with placebo; mean difference 0·07 [95% CI -0·31 to 0·45], p=0·72) or mean sum of skinfolds (23·0 mm [5·2] vs 23·8 mm [5·4]; mean difference 0·8 mm [-0·7 to 2·3], p=0·31). Metformin was not a predictor of BMI Z score at 24 months of age (mean difference -0·01 [95% CI -0·42 to 0·37], p=0·92). There was no overall difference in BMI trajectory but, in males, trajectories were significantly different by treatment (p=0·048); BMI in the metformin group was higher between 6 and 24 months. Children of women with type 2 diabetes were approximately 1 SD heavier than the WHO reference population. INTERPRETATION Anthropometrics were similar in children exposed and those not exposed to metformin in utero; hence, overall, data are reassuring with regard to the use of metformin during pregnancy in women with type 2 diabetes and the long-term health of their children. FUNDING Canadian Institute for Health Research.
Collapse
Affiliation(s)
- Denice S Feig
- Department of Medicine, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada; Sinai Health System, Mount Sinai Hospital, Toronto, ON, Canada.
| | | | - Kellie E Murphy
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada; Sinai Health System, Mount Sinai Hospital, Toronto, ON, Canada
| | - Elizabeth Asztalos
- Sunnybrook Research Institute, Toronto, ON, Canada; Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Bernard Zinman
- Department of Medicine, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada; Sinai Health System, Mount Sinai Hospital, Toronto, ON, Canada
| | - David Simmons
- Department of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Andrea M Haqq
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - I George Fantus
- Department of Medicine, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada; Sinai Health System, Mount Sinai Hospital, Toronto, ON, Canada; Department of Medicine and Research Institute, McGill University Health Center, McGill University, Montreal, QC, Canada
| | - Lorraine Lipscombe
- Department of Medicine, University of Toronto, Toronto, ON, Canada; Department of Medicine, Women's College Hospital, Toronto, ON, Canada
| | - Anthony Armson
- Department of Obstetrics and Gynecology, Dalhousie University, Halifax, NS, Canada
| | - Jon Barrett
- Sunnybrook Research Institute, Toronto, ON, Canada; Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Lois Donovan
- Department of Medicine and Department of Obstetrics and Gynecology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, Calgary, AB, Canada
| | - Paul Karanicolas
- Sunnybrook Research Institute, Toronto, ON, Canada; Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | | | - Gail Klein
- Sunnybrook Research Institute, Toronto, ON, Canada
| | - Yidi Jiang
- Sunnybrook Research Institute, Toronto, ON, Canada
| | - George Tomlinson
- Department of Medicine, University of Toronto, Toronto, ON, Canada; Department of Medicine, University Health Network, Toronto, ON, Canada
| | - Jill Hamilton
- Department of Pediatrics, Division of Endocrinology, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
12
|
Stimulation of GHRH Neuron Axon Growth by Leptin and Impact of Nutrition during Suckling in Mice. Nutrients 2023; 15:nu15051077. [PMID: 36904077 PMCID: PMC10005278 DOI: 10.3390/nu15051077] [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: 12/29/2022] [Revised: 02/12/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Nutrition during the early postnatal period can program the growth trajectory and adult size. Nutritionally regulated hormones are strongly suspected to be involved in this physiological regulation. Linear growth during the postnatal period is regulated by the neuroendocrine somatotropic axis, whose development is first controlled by GHRH neurons of the hypothalamus. Leptin that is secreted by adipocytes in proportion to fat mass is one of the most widely studied nutritional factors, with a programming effect in the hypothalamus. However, it remains unclear whether leptin stimulates the development of GHRH neurons directly. Using a Ghrh-eGFP mouse model, we show here that leptin can directly stimulate the axonal growth of GHRH neurons in vitro in arcuate explant cultures. Moreover, GHRH neurons in arcuate explants harvested from underfed pups were insensitive to the induction of axonal growth by leptin, whereas AgRP neurons in these explants were responsive to leptin treatment. This insensitivity was associated with altered activating capacities of the three JAK2, AKT and ERK signaling pathways. These results suggest that leptin may be a direct effector of linear growth programming by nutrition, and that the GHRH neuronal subpopulation may display a specific response to leptin in cases of underfeeding.
Collapse
|
13
|
Koohmanaee S, Badeli H, Rad AH, Novin MH, Mostofizadeh N, Dalili S, Kazemnejad-Leili E. Predicting childhood overweight status by accelerated weight gain from neonatal period to infancy. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2023; 28:2. [PMID: 36974114 PMCID: PMC10039107 DOI: 10.4103/jrms.jrms_1041_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/21/2022] [Accepted: 10/12/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND The increased prevalence of obesity in early childhood is a public health problem. Childhood obesity may affect cardiorespiratory fitness and can induce obesity and its comorbidities in adulthood. We aimed to assess childhood overweight status by accelerated weight gain during infancy. MATERIALS AND METHODS This is a historical cohort that was conducted on 637 7-year-old students of Guilan province, north of Iran. Data were collected, including demographic characteristics, weight at 4, 6, 12, and 18 months, and clinical examination. The ROC curve was designated based on the standardized z-scores, and the most appropriate cutoff point by sensitivity and specificity was noted for predicting obesity at 7 years. Rapid weight gain (RWG) was also assessed. RESULTS Among participants, 334 (53.3%) were female. In this study, the mean and standard deviation of RWG in 0-4 months, 0-6 months, 0-12 months, and 0-18 months were 3.50 ± 0.89, 4.64 ± 1.02, 6.54 ± 1.21, and 8.00 ± 1.46 kg, respectively. The highest AUC was dedicated to 0-18 months (0.7 ± 0.05) and the suitable cut-off for RWG in this interval was 8.55 kg with 65.5% and 72.0% sensitivity and specificity, respectively. CONCLUSION Although in the previous investigations, the changes in the first 3 years of life had a significant role in further complications, regarding our results, it seems that even earlier consideration of excess weight gain may be necessary.
Collapse
Affiliation(s)
- Shahin Koohmanaee
- Pediatric Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Hamidreza Badeli
- Pediatric Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Afagh Hassanzadeh Rad
- Pediatric Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Hassan Novin
- Pediatric Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Neda Mostofizadeh
- Metabolic Liver Disease Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Setila Dalili
- Pediatric Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | | |
Collapse
|
14
|
Robertson OC, Marceau K, Moding KJ, Knopik VS. Developmental pathways linking obesity risk and early puberty: The thrifty phenotype and fetal overnutrition hypotheses. DEVELOPMENTAL REVIEW 2022. [DOI: 10.1016/j.dr.2022.101048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
15
|
Dynamics of human milk oligosaccharides in early lactation and relation with growth and appetitive traits of Filipino breastfed infants. Sci Rep 2022; 12:17304. [PMID: 36243744 PMCID: PMC9569346 DOI: 10.1038/s41598-022-22244-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/12/2022] [Indexed: 01/10/2023] Open
Abstract
Human milk oligosaccharides play a key role in the maturation of the infant gut microbiome and immune system and are hypothesized to affect growth. This study examined the temporal changes of 24 HMOs and their associations to infant growth and appetitive traits in an exploratory, prospective, observational, study of 41 Filipino mother-infant dyads. Exclusively breastfed, healthy, term infants were enrolled at 21-26 days of age (≈ 0.75 mo) and followed for 6 months. Infant growth measures and appetitive traits were collected at visit 1 (V1) (≈ 0.75 mo), V2 (≈ 1.5 mo), V3 (2.5 mo), V4 (2.75 mo), V5 (4 mo), and V6 (6 mo), while HMOs were measured at V1, V2, V3 and V5. Overall exposure to each HMO was summarized as area under the curve from baseline to 4 months of age and examined in association with each measure of growth at 6 months using linear regression adjusted for maternal age at birth, infant sex, birth weight, and mode of delivery. We saw modest associations between several HMOs and infant growth parameters. Our results suggest that specific HMOs, partly as proxy for milk groups (defined by Secretor and Lewis status), may be associated with head circumference and length, increasing their relevance especially in populations at the lower end of the WHO growth curve. We did not identify the same HMOs associated with infant appetitive traits, indicating that at least in our cohort, changes in appetite were not driving the observed associations between HMOs and growth.Clinical trial registration: NCT03387124.
Collapse
|
16
|
Zheng Y, Liang J, Zeng D, Tan W, Yang L, Lu S, Yao W, Yang Y, Liu L. Association of body composition with pubertal timing in children and adolescents from Guangzhou, China. Front Public Health 2022; 10:943886. [PMID: 36062089 PMCID: PMC9428289 DOI: 10.3389/fpubh.2022.943886] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/20/2022] [Indexed: 01/21/2023] Open
Abstract
Objective An ongoing debate surrounds the relationship between body composition and pubertal timing, in particular for boys. This cross-sectional study aimed to investigate the association of body composition with pubertal timing among children and adolescents. Methods A total of 1,493 boys and 1,261 girls who entered puberty were enrolled in Guangzhou, China. Tanner stages were evaluated by examination of breast development for girls and testicular volume for boys. Fat mass (FM) and fat-free mass (FFM) were determined by bioelectrical impedance analysis. Parameters for body composition were transformed into age-and gender-specific Z-scores. The association of body composition with pubertal timing was examined using multinomial logistic regression with inverse probability weighting (IPW) based on the propensity score. Results For boys, IPW analysis showed Z-scores of body fat percentage (BF%) and FM index (FMI) were negatively associated with early puberty (OR = 0.75, 95% CI = 0.64-0.87; OR = 0.74, 95% CI = 0.63-0.88). As for girls, in contrast to boys, positive associations were seen between BF% and FMI with early puberty (OR = 1.39, 95% CI = 1.19-1.64; OR = 1.59, 95% CI = 1.33-1.90). With respect to appendicular skeletal muscle mass index (ASMI), there was a positive association with early puberty and a negative one with late puberty in boys (OR = 1.26, 95% CI = 1.07-1.49; OR = 0.82, 95% CI = 0.69-0.99). Conclusion There is a positive association of FM with early puberty for girls while negative for boys. FFM yields a positive association with early puberty and a negative one with late puberty in boys, but not in girls. Our findings highlight the gender differences in the connection between body composition and pubertal onset.
Collapse
Affiliation(s)
- Yijin Zheng
- Department of Epidemiology and Biostatistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jianping Liang
- Guangzhou Health Care Promotion Center for Primary and Middle Schools, Guangzhou, China
| | - Ding Zeng
- Department of Epidemiology and Biostatistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Weiqing Tan
- Guangzhou Health Care Promotion Center for Primary and Middle Schools, Guangzhou, China
| | - Lun Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shuang Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wanwen Yao
- Department of Epidemiology and Biostatistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yi Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Li Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China,*Correspondence: Li Liu
| |
Collapse
|
17
|
Dimas A, Politi A, Bargiota A, Panoskaltsis T, Vlahos NF, Valsamakis G. The Gestational Effects of Maternal Bone Marker Molecules on Fetal Growth, Metabolism and Long-Term Metabolic Health: A Systematic Review. Int J Mol Sci 2022; 23:ijms23158328. [PMID: 35955462 PMCID: PMC9368754 DOI: 10.3390/ijms23158328] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
Fetal exposure in adverse environmental factors during intrauterine life can lead to various biological adjustments, affecting not only in utero development of the conceptus, but also its later metabolic and endocrine wellbeing. During human gestation, maternal bone turnover increases, as reflected by molecules involved in bone metabolism, such as vitamin D, osteocalcin, sclerostin, sRANKL, and osteoprotegerin; however, recent studies support their emerging role in endocrine functions and glucose homeostasis regulation. Herein, we sought to systematically review current knowledge on the effects of aforementioned maternal bone biomarkers during pregnancy on fetal intrauterine growth and metabolism, neonatal anthropometric measures at birth, as well as on future endocrine and metabolic wellbeing of the offspring. A growing body of literature converges on the view that maternal bone turnover is likely implicated in fetal growth, and at least to some extent, in neonatal and childhood body composition and metabolic wellbeing. Maternal sclerostin and sRANKL are positively linked with fetal abdominal circumference and subcutaneous fat deposition, contributing to greater birthweights. Vitamin D deficiency correlates with lower birthweights, while research is still needed on intrauterine fetal metabolism, as well as on vitamin D dosing supplementation during pregnancy, to diminish the risks of low birthweight or SGA neonates in high-risk populations.
Collapse
Affiliation(s)
- Angelos Dimas
- 3rd University Department of Obstetrics & Gynecology, Attikon University Hospital, Medical School of Athens, Ethnikon and Kapodistriakon University of Athens, 12462 Athens, Greece
- Obst & Gynae Department, University Hospital of Ioannina, Stavros Niarchos Ave., 45500 Ioannina, Greece
- Correspondence: (A.D.); (G.V.)
| | - Anastasia Politi
- Nephrology Department, University Hospital of Ioannina, Stavros Niarchos Ave., 45500 Ioannina, Greece;
| | - Alexandra Bargiota
- Department of Endocrinology and Metabolic Diseases, Medical School, Larissa University Hospital, University of Thessaly, 41334 Larissa, Greece;
| | - Theodoros Panoskaltsis
- 2nd University Department of Obstetrics & Gynecology, “Aretaieion” University Hospital, Medical School of Athens, Ethnikon and Kapodistriakon University of Athens, 12462 Athens, Greece; (T.P.); (N.F.V.)
| | - Nikolaos F. Vlahos
- 2nd University Department of Obstetrics & Gynecology, “Aretaieion” University Hospital, Medical School of Athens, Ethnikon and Kapodistriakon University of Athens, 12462 Athens, Greece; (T.P.); (N.F.V.)
| | - Georgios Valsamakis
- Endocrine Unit, 2nd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, “Aretaieion” University Hospital, 11528 Athens, Greece
- Correspondence: (A.D.); (G.V.)
| |
Collapse
|
18
|
Preterm birth and subsequent timing of pubertal growth, menarche, and voice break. Pediatr Res 2022; 92:199-205. [PMID: 34429512 PMCID: PMC9411060 DOI: 10.1038/s41390-021-01690-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/10/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND We evaluated pubertal growth and pubertal timing of participants born preterm compared to those born at term. METHODS In the ESTER Preterm Birth Study, we collected growth data and measured final height of men/women born very or moderately preterm (<34 gestational weeks, n = 52/55), late preterm (34-<37 weeks, 94/106), and term (≥37 weeks, 131/151), resulting in median 9 measurements at ≥6 years. Timing of menarche or voice break was self-reported. Peak height velocity (PHV, cm/year) and age at PHV (years) were compared with SuperImposition by Translation And Rotation (SITAR) model (sexes separately). RESULTS Age at PHV (years) and PHV (cm/year) were similar in all gestational age groups. Compared to term controls, insignificant differences in age at PHV were 0.1 (95% CI: -0.2 to 0.4) years/0.2 (-0.1 to 0.4) for very or moderately/late preterm born men and -0.0 (-0.3 to 0.3)/-0.0 (-0.3 to 0.2) for women, respectively. Being born small for gestational age was not associated with pubertal growth. Age at menarche or voice break was similar in all the gestational age groups. CONCLUSIONS Timing of pubertal growth and age at menarche or voice break were similar in participants born preterm and at term. IMPACT Pubertal growth and pubertal timing were similar in preterm and term participants in a relatively large cohort with a wide range of gestational ages. Previous literature indicates that small for gestational age is a risk for early puberty in term born children. This was not shown in preterm children. While our study had limited power for children born very preterm, all children born preterm were not at increased risk for early puberty.
Collapse
|
19
|
Shalitin S, Gat-Yablonski G. Associations of Obesity with Linear Growth and Puberty. Horm Res Paediatr 2022; 95:120-136. [PMID: 34130293 DOI: 10.1159/000516171] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 03/27/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The prevalence of obesity in childhood has increased dramatically in recent decades with increased risk of developing cardiometabolic and other comorbidities. Childhood adiposity may also influence processes of growth and puberty. SUMMARY Growth patterns of obesity during childhood have been shown to be associated with increased linear growth in early childhood, leading to accelerated epiphyseal growth plate (EGP) maturation. Several hormones secreted by the adipose tissue may affect linear growth in the context of obesity, both via the growth hormone IGF-1 axis and via a direct effect on the EGP. The observation that children with obesity tend to mature earlier than lean children has led to the assumption that the degree of body fatness may trigger the neuroendocrine events that lead to pubertal onset. The most probable link between obesity and puberty is leptin and its interaction with the kisspeptin system, which is an important regulator of puberty. However, peripheral action of adipose tissue could also be involved in changes in the onset of puberty. In addition, nutritional factors, epigenetics, and endocrine-disrupting chemicals are potential mediators linking pubertal onset to obesity. In this review, we focused on interactions of obesity with linear growth and pubertal processes, based on basic research and clinical data in humans. KEY MESSAGE Children with obesity are subject to accelerated linear growth with risk of impaired adult height and early puberty, with its psychological consequences. The data highlight another important objective in combatting childhood obesity, for the prevention of abnormal growth and pubertal patterns.
Collapse
Affiliation(s)
- Shlomit Shalitin
- National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Galia Gat-Yablonski
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Felsenstein Medical Research Center, Petach Tikva, Israel
| |
Collapse
|
20
|
Dai W, Liu X, Su H, Li X, Xu Y, Yu Y. Influence of adipose tissue immune dysfunction on childhood obesity. Cytokine Growth Factor Rev 2022; 65:27-38. [PMID: 35595599 DOI: 10.1016/j.cytogfr.2022.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 11/17/2022]
Abstract
In recent decades, a dramatic rise has been observed in the prevalence of obesity in childhood and adolescence, along with an increase in fetal microsomia rates. The increased risk of obesity during this key period in development negatively affects the health of the individual later in life. Immune cells residing and recruited to white adipose tissue have been highlighted as important factors contributing to the pathogenesis of childhood obesity. Immune dysfunction in the context of obesity begins early in childhood, which is different from the pathological characteristics and influencing factors of adipose immunity in adults. Here, we explore the current understanding of the roles of childhood and early life events that result in high risks for obesity by influencing adipose tissue immune dysfunction under the pathological condition of obesity. Such knowledge will help in determining the mechanisms of childhood and early life obesity in efforts to ameliorate chronic inflammation-related metabolic diseases.
Collapse
Affiliation(s)
- Wanlin Dai
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China; Innovation Institute, China Medical University, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiyan Liu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Han Su
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Xuan Li
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China; Innovation Institute, China Medical University, China Medical University, Shenyang 110122, Liaoning, China
| | - Yingxi Xu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Yang Yu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
21
|
Aghaee S, Quesenberry CP, Deardorff J, Kushi LH, Greenspan LC, Ferrara A, Kubo A. Associations between infant growth and pubertal onset timing in a multiethnic prospective cohort of girls. BMC Pediatr 2022; 22:171. [PMID: 35361165 PMCID: PMC8969386 DOI: 10.1186/s12887-022-03242-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Early puberty increases risk of adverse health conditions throughout the life course. US girls are experiencing earlier puberty without clear reasons. Studies suggest early life factors, such as infant growth, may influence pubertal timing. We assessed the associations between infant growth and onset of breast development (thelarche), pubic hair development (pubarche), and menarche in girls. METHODS A prospective cohort of girls born at a Kaiser Permanente Northern California medical facility in 2005-11 was used. Weight-for-age z-scores were calculated at birth and 24 months. Difference in z-scores greater than 0.67 represent rapid "catch-up" growth, less than -0.67 represent delayed "catch-down" growth, and between -0.67 and 0.67 represent "normal" growth. Pubertal onset was measured using clinician-assessed sexual maturity ratings (SMRs) and defined as the age at transition from SMR 1 to SMR 2 + for both thelarche and pubarche. SMR data was collected through June 2020. Menarche was analyzed as a secondary outcome. Weibull and modified Poisson regression models were used. Models were adjusted for potential confounders. RESULTS There were 15,196 girls included in the study. Approximately 30.2% experienced catch-up growth, 25.8% experienced catch-down growth, and 44% had normal growth. Girls with catch-up growth had increased risk of earlier thelarche (hazard ratio = 1.26, 95% confidence interval (CI): 1.18, 1.35), pubarche (1.38, 95% CI: 1.28, 1.48), and menarche (< 12y, relative risk = 1.52, 95% CI: 1.36, 1.69) compared to those with normal growth, after adjusting for covariates. These associations were partially mediated by childhood body mass index. Catch-down growth was associated with later pubertal onset. CONCLUSIONS Girls who experience infant catch-up growth have higher risk of earlier pubertal development compared to girls with normal growth and the associations are partially explained by childhood obesity. This information may help clinicians to monitor girls who are at high risk of developing earlier.
Collapse
Affiliation(s)
- Sara Aghaee
- Kaiser Permanente Northern California Division of Research, 2000 Broadway, Oakland, CA, 94612, USA
| | - Charles P Quesenberry
- Kaiser Permanente Northern California Division of Research, 2000 Broadway, Oakland, CA, 94612, USA
| | - Julianna Deardorff
- Division of Maternal and Child Health, University of California, School of Public Health, 2121 Berkeley Way #5302, Berkeley, CA, 94720, USA
| | - Lawrence H Kushi
- Kaiser Permanente Northern California Division of Research, 2000 Broadway, Oakland, CA, 94612, USA
| | - Louise C Greenspan
- Kaiser Permanente San Francisco Medical Center, 2425 Geary Boulevard, San Francisco, CA, 94115, USA
| | - Assiamira Ferrara
- Kaiser Permanente Northern California Division of Research, 2000 Broadway, Oakland, CA, 94612, USA
| | - Ai Kubo
- Kaiser Permanente Northern California Division of Research, 2000 Broadway, Oakland, CA, 94612, USA.
| |
Collapse
|
22
|
Rasmussen JM, Thompson PM, Entringer S, Buss C, Wadhwa PD. Fetal programming of human energy homeostasis brain networks: Issues and considerations. Obes Rev 2022; 23:e13392. [PMID: 34845821 DOI: 10.1111/obr.13392] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/29/2021] [Accepted: 10/24/2021] [Indexed: 02/07/2023]
Abstract
In this paper, we present a transdisciplinary framework and testable hypotheses regarding the process of fetal programming of energy homeostasis brain circuitry. Our model proposes that key aspects of energy homeostasis brain circuitry already are functional by the time of birth (with substantial interindividual variation); that this phenotypic variation at birth is an important determinant of subsequent susceptibility for energy imbalance and childhood obesity risk; and that this brain circuitry exhibits developmental plasticity, in that it is influenced by conditions during intrauterine life, particularly maternal-placental-fetal endocrine, immune/inflammatory, and metabolic processes and their upstream determinants. We review evidence that supports the scientific premise for each element of this formulation, identify future research directions, particularly recent advances that may facilitate a better quantification of the ontogeny of energy homeostasis brain networks, highlight animal and in vitro-based approaches that may better address the determinants of interindividual variation in energy homeostasis brain networks, and discuss the implications of this formulation for the development of strategies targeted towards the primary prevention of childhood obesity.
Collapse
Affiliation(s)
- Jerod M Rasmussen
- Development, Health and Disease Research Program, University of California, Irvine, California, USA.,Department of Pediatrics, University of California, Irvine, California, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging and Informatics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Sonja Entringer
- Development, Health and Disease Research Program, University of California, Irvine, California, USA.,Department of Pediatrics, University of California, Irvine, California, USA.,Department of Medical Psychology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Departments of Psychiatry and Human Behavior, Obstetrics and Gynecology, Epidemiology, University of California, Irvine, California, USA
| | - Claudia Buss
- Development, Health and Disease Research Program, University of California, Irvine, California, USA.,Department of Pediatrics, University of California, Irvine, California, USA.,Department of Medical Psychology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Departments of Psychiatry and Human Behavior, Obstetrics and Gynecology, Epidemiology, University of California, Irvine, California, USA
| | - Pathik D Wadhwa
- Development, Health and Disease Research Program, University of California, Irvine, California, USA.,Department of Pediatrics, University of California, Irvine, California, USA.,Departments of Psychiatry and Human Behavior, Obstetrics and Gynecology, Epidemiology, University of California, Irvine, California, USA.,Department of Obstetrics and Gynecology, University of California, Irvine, California, USA.,Department of Epidemiology, University of California, Irvine, California, USA
| |
Collapse
|
23
|
Pereira A, Merino PM, Santos JL, Iñiguez G, Cutler GB, Corvalan C, Mericq V. High DHEAS in girls and metabolic features throughout pubertal maturation. Clin Endocrinol (Oxf) 2022; 96:419-427. [PMID: 34904249 DOI: 10.1111/cen.14654] [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: 09/02/2021] [Revised: 11/23/2021] [Accepted: 11/27/2021] [Indexed: 01/19/2023]
Abstract
CONTEXT An association between premature adrenarche and metabolic syndrome at presentation has been described. Our aim was to assess whether the presence of high dehydroepiandrosterone sulphate (DHEAS [HD]) at the adrenarche determines the risk of metabolic syndrome during puberty, taking into account body mass index (BMI) and birth weight. DESIGN Prospective observational. PATIENTS Five hundred four girls from the Growth and Obesity Chilean Cohort Study were followed from birth through puberty. At age ~7, subjects were classified by DHEAS concentrations into the HD (>75th percentile) or normal DHEAS (ND, ≤75th percentile) subgroups. MEASUREMENTS Anthropometrics, semiannual clinical pubertal staging and hormonal and metabolic levels. The relationships among DHEAS at age ~7, metabolic syndrome, and each of its components independently, were analyzed by linear and logistic regression models during puberty and 1-year postmenarche, adjusted by confounders. RESULTS Girls with HD at 7 years exhibited higher BMI, more central fat and higher serum androgen and insulin like growth factor (IGF)-I levels throughout puberty. Also, girls with HD had a greater prevalence of hyperglycemia at B2 and B4 breast stages, and of low HDL at B4. At 1 year after menarche, HD girls had a higher prevalence of metabolic syndrome, and those with BMI > 1 SD score had a higher metabolic score and insulin levels than ND girls with similar BMI. CONCLUSIONS Our observations suggest that girls with HD at the age of adrenarche may be at greater risk for metabolic syndrome at adolescence, especially in those who are overweight or obese. Our results emphasize the importance of lifestyle interventions for childhood overweight and obesity among girls with HD.
Collapse
Affiliation(s)
- Ana Pereira
- Institute of Nutrition and Food Technology (INTA), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Paulina M Merino
- Institute of Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Jose L Santos
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - German Iñiguez
- Institute of Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile
| | | | - Camila Corvalan
- Institute of Nutrition and Food Technology (INTA), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Veronica Mericq
- Institute of Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
24
|
Houghton LC, Wei Y, Wang T, Goldberg M, Paniagua-Avila A, Sweeden RL, Bradbury A, Daly M, Schwartz LA, Keegan T, John EM, Knight JA, Andrulis IL, Buys SS, Frost CJ, O’Toole K, White ML, Chung WK, Terry MB. Body mass index rebound and pubertal timing in girls with and without a family history of breast cancer: the LEGACY girls study. Int J Epidemiol 2022; 51:1546-1555. [PMID: 35157067 PMCID: PMC9799198 DOI: 10.1093/ije/dyac021] [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: 06/04/2021] [Accepted: 02/02/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Heavier body mass index (BMI) is the most established predictor of earlier age at puberty. However, it is unknown whether the timing of the childhood switch to heavier BMI (age at BMI rebound) also matters for puberty. METHODS In the LEGACY Girls Study (n = 1040), a longitudinal cohort enriched with girls with a family history of breast cancer, we collected paediatric growth chart data from 852 girls and assessed pubertal development every 6 months. Using constrained splines, we interpolated individual growth curves and then predicted BMI at ages 2, 4, 6, 8 and 9 years for 591 girls. We defined age at BMI rebound as the age at the lowest BMI between ages 2 and 8 years and assessed its association with onset of thelarche, pubarche and menarche using Weibull survival models. RESULTS The median age at BMI rebound was 5.3 years (interquartile range: 3.6-6.7 years). A 1-year increase in age at BMI rebound was associated with delayed thelarche (HR = 0.90; 95% CI = 0.83-0.97) and menarche (HR = 0.86; 95% CI = 0.79-0.94). The magnitude of these associations remained after adjusting for weight between birth and 2 years, was stronger after adjusting for BMI at age 9, and was stronger in a subset of girls with clinically assessed breast development. CONCLUSIONS Earlier BMI rebound is associated with earlier pubertal timing. Our observation that BMI rebound may be a driver of pubertal timing in girls with and without a family history of breast cancer provides insight into how growth and pubertal timing are associated with breast cancer risk.
Collapse
Affiliation(s)
- Lauren C Houghton
- Corresponding author. Department of Epidemiology, Mailman School of Public Health, 722 West 168th Street, Room 706, New York, NY 10032, USA. E-mail:
| | - Ying Wei
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Tianying Wang
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Mandy Goldberg
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Alejandra Paniagua-Avila
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Rachel L Sweeden
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Angela Bradbury
- Departments of Medicine and Hematology/Oncology and of Medical Ethics and Health Policy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mary Daly
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Lisa A Schwartz
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Theresa Keegan
- Department of Internal Medicine, Center for Oncology Hematology Outcomes Research and Training (COHORT), University of California, Davis, Sacramento, CA, USA
| | - Esther M John
- Departments of Epidemiology & Population Health and Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Julia A Knight
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Irene L Andrulis
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Departments of Molecular Genetics and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Saundra S Buys
- Department of Medicine, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, UT, USA
| | - Caren J Frost
- College of Social Work, University of Utah, Salt Lake City, UT, USA
| | - Karen O’Toole
- Department of Medicine, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, UT, USA
| | - Melissa L White
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Wendy K Chung
- Departments of Pediatrics and Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Mary Beth Terry
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
25
|
Bar-Sadeh B, Amichai OE, Pnueli L, Begum K, Leeman G, Emes RD, Stöger R, Bentley GR, Melamed P. Epigenetic regulation of 5α reductase-1 underlies adaptive plasticity of reproductive function and pubertal timing. BMC Biol 2022; 20:11. [PMID: 34996447 PMCID: PMC8742331 DOI: 10.1186/s12915-021-01219-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 12/16/2021] [Indexed: 12/30/2022] Open
Abstract
Background Women facing increased energetic demands in childhood commonly have altered adult ovarian activity and shorter reproductive lifespan, possibly comprising a strategy to optimize reproductive success. Here, we sought to understand the mechanisms of early-life programming of reproductive function, by integrating analysis of reproductive tissues in an appropriate mouse model with methylation analysis of proxy tissue DNA in a well-characterized population of Bangladeshi migrants in the UK. Bangladeshi women whose childhood was in Bangladesh were found to have later pubertal onset and lower age-matched ovarian reserve than Bangladeshi women who grew-up in England. Subsequently, we aimed to explore the potential relevance to the altered reproductive phenotype of one of the genes that emerged from the screens. Results Of the genes associated with differential methylation in the Bangladeshi women whose childhood was in Bangladesh as compared to Bangladeshi women who grew up in the UK, 13 correlated with altered expression of the orthologous gene in the mouse model ovaries. These mice had delayed pubertal onset and a smaller ovarian reserve compared to controls. The most relevant of these genes for reproductive function appeared to be SRD5A1, which encodes the steroidogenic enzyme 5α reductase-1. SRD5A1 was more methylated at the same transcriptional enhancer in mice ovaries as in the women’s buccal DNA, and its expression was lower in the hypothalamus of the mice as well, suggesting a possible role in the central control of reproduction. The expression of Kiss1 and Gnrh was also lower in these mice compared to controls, and inhibition of 5α reductase-1 reduced Kiss1 and Gnrh mRNA levels and blocked GnRH release in GnRH neuronal cell cultures. Crucially, we show that inhibition of this enzyme in female mice in vivo delayed pubertal onset. Conclusions SRD5A1/5α reductase-1 responds epigenetically to the environment and its downregulation appears to alter the reproductive phenotype. These findings help to explain diversity in reproductive characteristics and how they are shaped by early-life environment and reveal novel pathways that might be targeted to mitigate health issues caused by life-history trade-offs. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01219-6.
Collapse
Affiliation(s)
- Ben Bar-Sadeh
- Faculty of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Or E Amichai
- Faculty of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Lilach Pnueli
- Faculty of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Khurshida Begum
- Department of Anthropology, Durham University, Durham, DH1 3LE, UK
| | - Gregory Leeman
- School of Biosciences, University of Nottingham, Nottingham, LE12 5RD, UK
| | - Richard D Emes
- School of Veterinary Medicine and Sciences, University of Nottingham, Nottingham, LE12 5RD, UK
| | - Reinhard Stöger
- School of Biosciences, University of Nottingham, Nottingham, LE12 5RD, UK
| | | | - Philippa Melamed
- Faculty of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel.
| |
Collapse
|
26
|
Marphatia AA, Wells JCK, Reid AM, Yajnik CS. Biosocial life-course factors associated with women's early marriage in rural India: The prospective longitudinal Pune Maternal Nutrition Study. AMERICAN JOURNAL OF BIOLOGICAL ANTHROPOLOGY 2022; 177:147-161. [PMID: 36787733 DOI: 10.1002/ajpa.24408] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/20/2021] [Accepted: 08/30/2021] [Indexed: 11/09/2022]
Abstract
OBJECTIVES By convention, women's early marriage is considered a sociocultural decision sensitive to factors acting during adolescence such as poverty, early menarche, and less education. Few studies have examined broader risk factors in the natal household prior to marriage. We investigated whether biosocial markers of parental investment through the daughters' life-course were associated with early marriage risk in rural India. We used an evolutionary perspective to interpret our findings. MATERIALS AND METHODS A prospective cohort recruited mothers at preconception. Children were followed from birth to age 21 years. Multivariable logistic regression models estimated odds ratios of marrying early (<19 years) associated first with wealth, age at menarche and education, and then with broader markers of maternal phenotype, natal household characteristics, and girls' growth trajectories. Models adjusted for confounders. RESULTS Of 305 girls, 71 (23%) had married early. Early married girls showed different patterns of growth compared to unmarried girls. Neither poverty nor early menarche predicted early marriage. Girls' non-completion of lower secondary school predicted early marriage, explaining 19% of the variance. Independent of girls' lower schooling, nuclear household, low paternal education, shorter gestation, and girls' poor infant weight gain were associated with marrying early, explaining in combination 35% of the variance. DISCUSSION Early marriage reflects "future discounting," where reduced parental investment in daughters' somatic and educational capital from early in her life favors an earlier transition to the life-course stage when reproduction can occur. Interventions initiated in adolescence may occur too late in the life-course to effectively delay women's marriage.
Collapse
Affiliation(s)
| | - Jonathan C K Wells
- Population, Policy and Practice Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Alice M Reid
- Department of Geography, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
27
|
Mennella JA, Smethers AD, Decker JE, Delahanty MT, Stallings VA, Trabulsi JC. Effects of Early Weight Gain Velocity, Diet Quality, and Snack Food Access on Toddler Weight Status at 1.5 Years: Follow-Up of a Randomized Controlled Infant Formula Trial. Nutrients 2021; 13:nu13113946. [PMID: 34836199 PMCID: PMC8625308 DOI: 10.3390/nu13113946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 12/22/2022] Open
Abstract
This study followed children who participated in a feeding trial in which the type of randomized infant formula fed from 2 weeks significantly affected weight gain velocity during the first 4 months and weight-for-length Z (WLZ) scores up to 11.5 months. We focused on measures of anthropometry, dietary intakes, and parenting related to the provision of snack foods that were collected at the end of the trial (1 year) and the 1.5 years follow-up visit. We not only describe what toddlers are eating, but we also determined the independent and/or interactive effects of randomized formula group, early weight gain velocity, the nutrient content of the post-formula diet, and maternal snack food practices, on toddlers’ weight status. Diet quality underwent drastic changes during this 6-month period. As infant formula disappeared from the diet, fruit and 100% fruit juice intake increased slightly, while intake of “What We Eat in America” food categories sweetened beverages and snacks and sweets more than doubled. Added sugars accounted for 5% of energy needs at 1 year and 9% at 1.5 years. Generalized linear mixed models revealed that, independent of the randomized formula group, greater velocities of weight gain during early infancy and lower access to snacks as toddlers predicted higher WLZ and a greater proportion of toddlers with overweight at 1.5 years. Energy and added sugar intake had no significant effects. These findings add to the growing body of evidence that unhealthy dietary habits are formed even before formula weaning and that, along with improving early diet, transient rapid weight gain and parental feeding practices are modifiable determinants that may reduce risks for obesity.
Collapse
Affiliation(s)
- Julie A. Mennella
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA;
- Correspondence:
| | | | - Jessica E. Decker
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE 19173, USA; (J.E.D.); (M.T.D.); (J.C.T.)
| | - Michelle T. Delahanty
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE 19173, USA; (J.E.D.); (M.T.D.); (J.C.T.)
| | | | - Jillian C. Trabulsi
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE 19173, USA; (J.E.D.); (M.T.D.); (J.C.T.)
| |
Collapse
|
28
|
Bleil ME, Appelhans BM, Gregorich SE, Thomas AS, Hiatt RA, Roisman GI, Booth-LaForce C. Patterns of Early Life Weight Gain and Female Onset of Puberty. J Endocr Soc 2021; 5:bvab165. [PMID: 35274069 PMCID: PMC8900195 DOI: 10.1210/jendso/bvab165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Indexed: 11/19/2022] Open
Abstract
Context Prepubertal obesity is a well-established predictor of earlier pubertal onset, which is itself a risk factor for poor health and well-being. Identifying specific patterns of weight gain in early life may help explain differential risk for earlier pubertal onset. Objective The objective of the study was to examine patterns of weight gain across infancy and early childhood in relation to pubertal onset outcomes. Design, Setting, and Participants Participants were 426 girls in the National Institute of Child Health and Human Development Study of Early Child Care and Youth Development, a longitudinal birth cohort of children and their families followed between birth and adolescence. Main Outcome Measures Three pubertal onset outcomes were examined, including age at menarche and ages at Tanner stage II for dimensions of breast and pubic hair development. Results In infancy (birth to 15 months), greater percent weight gain and higher birthweight predicted earlier pubertal onset for all outcomes (Ps < 0.05). In early childhood (24 months to grade 1), body mass index (BMI) trajectories reflecting BMI values that were persistently high or changed from low to high over time (vs BMI values that were stable at median or low levels), predicted younger ages at menarche and the onset of breast (Ps < 0.05), but not pubic hair (Ps > 0.05), development. All associations were independent of breastfeeding, maternal menarcheal age, and race/ethnicity. Conclusions Distinct patterns of early life weight gain predict differential risk for earlier onset puberty. Focusing on these patterns for earlier and more targeted intervention may help lessen life course linkages between prepubertal obesity, accelerated pubertal development, and negative postpubertal outcomes.
Collapse
Affiliation(s)
- Maria E Bleil
- Child, Family, & Population Health Nursing, University of Washington, Seattle, WA 98195, USA
| | - Bradley M Appelhans
- Department of Preventive Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Steven E Gregorich
- Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Alexis S Thomas
- Child, Family, & Population Health Nursing, University of Washington, Seattle, WA 98195, USA
| | - Robert A Hiatt
- Department of Epidemiology & Biostatistics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Glenn I Roisman
- Institute of Child Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Cathryn Booth-LaForce
- Child, Family, & Population Health Nursing, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
29
|
Hvidt JJ, Brix N, Ernst A, Lunddorf LLH, Ramlau-Hansen CH. Breast feeding and timing of puberty in boys and girls: A nationwide cohort study. Paediatr Perinat Epidemiol 2021; 35:578-589. [PMID: 34080724 DOI: 10.1111/ppe.12767] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Breast feeding has been associated with improved infant health, but its impact on pubertal timing remains uncertain, particularly in boys. OBJECTIVE The objective of this study was to investigate the association between duration of breast feeding and pubertal timing in boys and girls. METHODS This population-based cohort study included 13 511 boys and girls from the Puberty Cohort nested within the Danish National Birth Cohort. The children gave half-yearly, self-reported information on pubertal development through questionnaires (Tanner stages, age at menarche, first ejaculation, voice break, axillary hair growth, and acne). Information on breast feeding was provided by the mothers when the children were 6 months of age. We estimated mean differences (in months) in age at attaining each pubertal marker and for overall timing of puberty (combined estimate) for every additional month of exclusive breast feeding. Furthermore, we estimated differences in pubertal age when comparing children never exclusively breastfed and exclusively breastfed <4 months using children exclusively breastfed ≥4 months as reference. In sub-analyses, we further adjusted for infant weight gain and childhood BMI at 7 years to investigate whether these variables mediated the association. RESULTS Boys tended to reach pubertal markers later for every additional month of exclusive breast feeding (combined estimate: 0.2 (95% confidence interval [CI] 0.0, 0.4 months). Never exclusively breastfed boys reached pubertal markers earlier than the boys exclusively breastfed ≥4 months (combined estimate: -4.1 (95% CI -6.7, -1.6) months). Boys exclusively breastfed <4 months also reached pubertal markers earlier than those never exclusively breastfed but with smaller differences. In girls, duration of breast feeding was not associated with pubertal development. When including infant weight gain or childhood BMI, the results remained essentially unchanged. CONCLUSIONS Shorter duration of exclusive breast feeding was associated with earlier pubertal development in boys but not in girls.
Collapse
Affiliation(s)
- Julie J Hvidt
- Department of Public Health, Research Unit for Epidemiology, Aarhus University, Aarhus, Denmark
| | - Nis Brix
- Department of Public Health, Research Unit for Epidemiology, Aarhus University, Aarhus, Denmark.,Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| | - Andreas Ernst
- Department of Public Health, Research Unit for Epidemiology, Aarhus University, Aarhus, Denmark.,Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Lea L H Lunddorf
- Department of Public Health, Research Unit for Epidemiology, Aarhus University, Aarhus, Denmark
| | - Cecilia H Ramlau-Hansen
- Department of Public Health, Research Unit for Epidemiology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
30
|
Perez-Miguelsanz J, Jiménez-Ortega V, Cano-Barquilla P, Garaulet M, Esquifino AI, Varela-Moreiras G, Fernández-Mateos P. Early Appearance of Epicardial Adipose Tissue through Human Development. Nutrients 2021; 13:nu13092906. [PMID: 34578784 PMCID: PMC8469969 DOI: 10.3390/nu13092906] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 12/21/2022] Open
Abstract
Background: Epicardial adipose tissue (EAT) is a visceral fat depot with unique anatomic, biomolecular and genetic features. Due to its proximity to the coronary arteries and myocardium, dysfunctional EAT may contribute to the development and progression of cardiovascular and metabolic-related adiposity-based chronic diseases. The aim of this work was to describe, by morphological techniques, the early origin of EAT. Methods: EAT adipogenesis was studied in 41 embryos from 32 gestational days (GD) to 8 gestational weeks (GW) and in 23 fetuses until full term (from 9 to 36 GW). Results: This process comprises five stages. Stage 1 appears as mesenchyme at 33-35 GD. Stage 2 is characterized by angiogenesis at 42-45 GD. Stage 3 covers up to 34 GW with the appearance of small fibers in the extracellular matrix. Stage 4 is visible around the coronary arteries, as multilocular adipocytes in primitive fat lobules, and Stage 5 is present with unilocular adipocytes in the definitive fat lobules. EAT precursor tissue appears as early as the end of the first gestational month in the atrioventricular grooves. Unilocular adipocytes appear at the eighth gestational month. Conclusions: Due to its early origin, plasticity and clinical implications, factors such as maternal health and nutrition might influence EAT early development in consequence.
Collapse
Affiliation(s)
- Juliana Perez-Miguelsanz
- Departamento de Anatomía y Embriología, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (V.J.-O.); (P.C.-B.); (A.I.E.)
| | - Vanesa Jiménez-Ortega
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (V.J.-O.); (P.C.-B.); (A.I.E.)
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Pilar Cano-Barquilla
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (V.J.-O.); (P.C.-B.); (A.I.E.)
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Marta Garaulet
- Departamento de Fisiología, Universidad de Murcia, IMIB-Arrixaca, 30120 Murcia, Spain;
| | - Ana I. Esquifino
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (V.J.-O.); (P.C.-B.); (A.I.E.)
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Gregorio Varela-Moreiras
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Boadilla del Monte, 28668 Madrid, Spain;
| | - Pilar Fernández-Mateos
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (V.J.-O.); (P.C.-B.); (A.I.E.)
- Departamento de Biología Celular, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain
- Correspondence: ; Tel.: +34-913-947-256
| |
Collapse
|
31
|
Bräuner EV, Koch T, Juul A, Doherty DA, Hart R, Hickey M. Prenatal exposure to maternal stressful life events and earlier age at menarche: the Raine Study. Hum Reprod 2021; 36:1959-1969. [PMID: 33744952 DOI: 10.1093/humrep/deab039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 12/21/2020] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION Is there an association between prenatal exposure to stressful life events and age at menarche, and does childhood BMI mediate this association? SUMMARY ANSWER Girls exposed to prenatal stress had a slightly earlier age at menarche, but this association did not show a dose-response effect and was not mediated by childhood offspring BMI. WHAT IS ALREADY KNOWN Prenatal stress may impact on reproductive function in females including age at menarche, but human data are very limited. High childhood BMI is known to be associated with earlier age at menarche. Only one small study has measured the association between maternal stress and age at menarche and reported that childhood BMI mediated the association between maternal stress and earlier age at menarche. However, neither maternal stress nor age at menarche was prospectively recorded and the study was limited to 31 mother-daughter pairs. STUDY DESIGN, SIZE, DURATION The Raine Study is a large prospective population-based pregnancy cohort study (n = 1414 mother-daughter pairs) continuously followed from prenatal life through to adolescence. In the present study, we examined the association between exposure to maternal stressful life events during early, late and total gestation and age at menarche in offspring using 753 mother-daughter pairs with complete case information. PARTICIPANTS/MATERIALS, SETTING, METHODS Mothers prospectively reported stressful life events during pregnancy at 18 and 34 weeks using a standardized 10-point questionnaire. Exact date of menarche was assessed using a purpose-designed questionnaire at 8, 10, 14 and 17 years of age. Complete information on exposure, outcome and confounding variables was obtained from 753 mothers-daughter pairs. Multivariate linear regression complete case analysis was used to examine associations between maternal stressful life event exposure and age at menarche. Potential selection bias was evaluated using multiple imputations (50 datasets). The mediating effects of offspring childhood BMI (ages 5, 8, or 10 years) on these associations were measured in separate sub-analyses. MAIN RESULTS AND ROLE OF CHANCE Most (580/753, 77%) daughters were exposed to at least one prenatal stressful life event. Exposure to maternal stressful life events during the entire pregnancy was associated with a non-linear earlier age at menarche. Exposure to one event and two or more psychological stressful events was associated with a 3.5 and 1.7-month earlier onset of puberty, respectively when compared to the reference group with no exposure maternal stressful life events. The estimates from multiple imputation with 50 datasets were comparable with complete case analysis confirming the existence of an underlying effect. No separate significant effects were observed for exposure during early or late gestation. The association between prenatal stressful events and age at menarche was not mediated by childhood BMI in the offspring. LIMITATIONS, REASONS FOR CAUTION Stressful life events may have affected pregnant women in different ways and self-perceived maternal stress severity may have provided a more precise estimate of gestational psychological stress. The observed non-linear U-shape of the association between maternal psychological stress and age at menarche did not reflect a dose-response. This suggests that the first exposure to prenatal stress exerts a greater effect on fetal reproductive development. A potential mechanism is via dramatic initial activation of the hypothalamic-pituitary-adrenal (HPA) axis following the first stressful life event which is greater than that observed following subsequent exposure to two or more maternal stressful life events. Whilst we adjusted for a priori chosen confounders, we cannot exclude residual confounding or confounding by factors we did not include. Maternal age at menarche was not available so the effects of familial history/genetics could not be assessed. There was a large loss due to the number of girls with no information on date of menarche and missing confounder information implying risk of selection bias and multiple imputation analyses did not fully exclude this risk (similar direction but slightly weaker estimate magnitude). WIDER IMPLICATIONS OF THE FINDINGS Menarche is a sentinel reproductive event and earlier age at menarche carries implications for psychological, social and reproductive health and for long-term risk of common non-communicable diseases. Understanding the factors regulating age at menarche has extensive health implications. This is the first population-based cohort study in humans to demonstrate that prenatal psychological stress might directly modify age at menarche. STUDY FUNDING/COMPETING INTEREST(S) Dr. Bräuner and Trine Koch's salaries were supported by Doctor Sofus Carl Emil Friis and spouse Olga Doris Friis foundation, The Danish Cancer Society (Kræftens Bekæmpelse, RP15468, R204-A12636, Denmark) and The Danish Health Foundation (Helsefonden, F-22181-23, Denmark). Martha Hickey was funded by NHMRC Practitioner Fellowships. The funding bodies played no role in the design, collection, analysis, or interpretation of data; in the writing of the manuscript; or in the decision to submit the manuscript for publication. Dr. Hart has received personal fees in his function as the Medical Director of Fertility Specialists of Western Australia and received educational sponsorship grants from MSD, Merck-Serono and from Ferring Pharmaceuticals. Dr Hart has also received personal fees from Shareholders in Western IVF outside the submitted work. TRIAL REGISTRATION NUMBER NA.
Collapse
Affiliation(s)
- E V Bräuner
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,The International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - T Koch
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,The International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - A Juul
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,The International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - D A Doherty
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Western Australia, Australia
| | - R Hart
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Western Australia, Australia.,Fertility Specialists of Western Australia, Bethesda Hospital, Claremont, Western Australia, Australia
| | - M Hickey
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Combs E, Ickes M. Factors That Influence Maternal Feeding Decisions for Toddlers: Extending the Theory of Planned Behavior. JOURNAL OF NUTRITION EDUCATION AND BEHAVIOR 2021; 53:583-590. [PMID: 34034983 DOI: 10.1016/j.jneb.2021.02.009] [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: 07/17/2020] [Revised: 02/19/2021] [Accepted: 02/28/2021] [Indexed: 06/12/2023]
Abstract
OBJECTIVE To use the Theory of Planned Behavior (TPB) to assess mothers' behavioral intentions to provide toddlers with a healthy diet and see if the addition of the parental role construction (PRC) variable strengthened the TPB. DESIGN An online survey using data gathered from preliminary research and a previously validated survey. PARTICIPANTS The final sample consisted of 148 mothers. The mean age was 32.8 (SD = 6.16) years. Most participants were married (87.2%), had earned a college degree or higher (79.7%), held part-time or full-time employment (60.8%), and were White (90.3%). MAIN OUTCOME MEASURES The TPB constructs, PRC, and the mothers' behavioral intentions surrounding toddler feeding. ANALYSIS The determinants of intention per the TPB were explored using descriptive statistics and multiple linear regression. RESULTS The TPB model predicted 53% of the variance in mother's behavioral intention to provide their toddler with a healthy diet. The addition of the PRC variable added 6% more predictive power to the model. CONCLUSIONS AND IMPLICATIONS Mothers' behavioral intentions surrounding feeding their toddlers were strongly influenced by the TPB constructs and their perceptions of the maternal role. Health promotion efforts should aim to increase the mother's sense of behavioral control and parental responsibility rather than focusing on the benefits of healthy eating.
Collapse
Affiliation(s)
- Elizabeth Combs
- Department of Dietetics and Human Nutrition, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY.
| | - Melinda Ickes
- Department of Kinesiology and Health Promotion, College of Education, University of Kentucky, Lexington, KY
| |
Collapse
|
33
|
Wohlfahrt-Veje C, Tinggaard J, Juul A, Toppari J, Skakkebæk NE, Main KM. Pubarche and Gonadarche Onset and Progression Are Differently Associated With Birth Weight and Infancy Growth Patterns. J Endocr Soc 2021; 5:bvab108. [PMID: 34250379 PMCID: PMC8262798 DOI: 10.1210/jendso/bvab108] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Indexed: 11/19/2022] Open
Abstract
Context Controversy exists regarding associations between early-life growth patterns and timing of puberty. Objective This work aims to investigate associations between birth anthropometry, early growth patterns, and onset/progression of pubertal milestones in boys and girls. Methods Among children examined at birth (1997-2003) and at age 36 months in a mother-child cohort, pubertal Tanner stages (B1-5, PH1-5, G1-5) and testicular volume were examined by trained physicians at 1 to 5 follow-up examinations during childhood and adolescence (672 girls and 846 boys, 2006-2013).With parametric survival models we analyzed associations between birth weight, changes in SD scores (SDS) from birth to 36 months (ΔSDS 0-36 > 0.67 SD defining catch-up growth), and age at pubertal onset/attainment of late pubertal stages/menarche. Results A 1-kg higher birth weight was associated with earlier onset of B2+ (thelarche): -3.9 months (CI, -6.7 to -1.1 months), G2+ (gonadarche): -2.7 months (-5.3 to -0.1 months), Tvol3+ (testis size > 3 mL): -2.8 months (CI, -4.9 to -0.7 months), but with later G4+ and PH4+ in boys, and a slower progression from B2 to menarche (5.3 months [CI, 1.2 to 9.4 months]) in girls. Catch-up growth was associated with earlier PH2+ (pubarche) in girls (-4.1 months [CI, -7.6 to -0.6 months]), earlier PH2+ in boys (-3.4 months [CI, -6.6 to -0.2 months]), faster progression from B2 to menarche in girls (-9.1 months [CI, 14.6 to 3.5 months]), and earlier G4+ and PH4+ in boys. Conclusion Associations between birthweight and infancy catch-up growth differed for gonadarche and pubarche, and for early and late pubertal markers, with similar patterns in both sexes.
Collapse
Affiliation(s)
- Christine Wohlfahrt-Veje
- Department of Growth and Reproduction and EDMaRC, Copenhagen University Hospital-Rigshospitalet, DK-2100 Copenhagen, Denmark.,International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Jeanette Tinggaard
- Department of Growth and Reproduction and EDMaRC, Copenhagen University Hospital-Rigshospitalet, DK-2100 Copenhagen, Denmark.,International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction and EDMaRC, Copenhagen University Hospital-Rigshospitalet, DK-2100 Copenhagen, Denmark.,International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Jorma Toppari
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark.,Institute of Biomedicine, Research Center of Integrative Physiology and Pharmacology, University of Turku, and Department of Pediatrics, Turku University Hospital, 20521 Turku, Finland
| | - Niels E Skakkebæk
- Department of Growth and Reproduction and EDMaRC, Copenhagen University Hospital-Rigshospitalet, DK-2100 Copenhagen, Denmark.,International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Katharina M Main
- Department of Growth and Reproduction and EDMaRC, Copenhagen University Hospital-Rigshospitalet, DK-2100 Copenhagen, Denmark.,International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
34
|
Wei J, Liu S, Cheng Y, Yang W, Zhu Z, Zeng L. Association of Infant Physical Development and Rapid Growth With Pubertal Onset Among Girls in Rural China. JAMA Netw Open 2021; 4:e216831. [PMID: 33938939 PMCID: PMC8094009 DOI: 10.1001/jamanetworkopen.2021.6831] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/02/2021] [Indexed: 11/16/2022] Open
Abstract
IMPORTANCE Whether physical development and rapid growth during the first 2 years of life are associated with puberty onset in girls remains unclear. OBJECTIVE To examine the associations of infant physical development and rapid growth with the timing of puberty onset in girls. DESIGN, SETTING, AND PARTICIPANTS This prospective birth cohort study included girls whose mothers had participated in a randomized, double-blind cluster trial of micronutrient supplementation with a daily capsule of folic acid, iron and folic acid, or multiple micronutrients during pregnancy. A total of 547 singleton girls in rural western China who were born after 2004 were followed up at 3, 6, 12, and 24 months of age and were eligible for long-term follow-up. Of these, 294 adolescent girls (aged 10-12 years), representing 53.7% of the total sample, were included in the follow-up interview that was conducted from June 1 through December 31, 2016. Data analyses were conducted from November 1, 2019, to May 30, 2020. EXPOSURES Infant weight and length were assessed at birth and 3, 6, 12, and 24 months of age. Rapid growth, defined as a weight-for-age or length-for-age z score increase of greater than 0.67, was calculated during 5 periods: birth to 3 months, birth to 6 months, 6 to 12 months, 12 to 24 months, and birth to 24 months of age. MAIN OUTCOMES AND MEASURES Puberty stage was assessed using the standardized Tanner staging system. Onset of puberty was defined as attaining Tanner stage 2 of breast development or pubic hair growth, and the corresponding age at follow-up was recorded. RESULTS Of 294 girls included in the analysis (mean [SD] age, 11.25 [0.57] years), the median age of puberty onset was 11 (interquartile range, 10-11) years. A significant association with puberty onset was found for infancy weight-for-age z score at 12 months of age (adjusted hazard ratio [HR], 1.20; 95% CI, 1.01-1.44) and rapid weight gain from birth to 24 months of age (adjusted HR, 1.40; 95% CI, 1.01-1.93) and birth to 3 months of age (adjusted HR, 1.39; 95% CI, 1.02-1.91). However, length-related indicators during early life were not associated with adolescent puberty onset. CONCLUSIONS AND RELEVANCE This study found that a higher weight-for-age z score and rapid weight gain during infancy, particularly from birth to 3 months of age, were associated with an increased likelihood of puberty onset in girls. These findings suggest that weight-related indicators may serve as proxies of puberty onset among adolescent girls.
Collapse
Affiliation(s)
- Jing Wei
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Shuang Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yue Cheng
- Department of Nutrition and Food Safety Research, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Wenfang Yang
- Department of Obstetrics and Gynecology, Maternal and Child Health Center, The First Affiliated Hospital of Health Science Center in Xi’an Jiaotong University, Xi’an, China
| | - Zhonghai Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Lingxia Zeng
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, China
| |
Collapse
|
35
|
Flores-Barrantes P, Iglesia I, Miguel-Berges ML, Vučinić V, Moreno LA, Rodríguez G. Rapid infant weight gain is associated with excess adiposity at 7-years of age in children from Aragon, Spain. CHILD AND ADOLESCENT OBESITY 2021. [DOI: 10.1080/2574254x.2020.1865122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Affiliation(s)
- P. Flores-Barrantes
- GENUD (Growth, Exercise, NUtrition and Development) Research Group, Faculty of Health Sciences, University of Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (Ciberobn), Instituto de Salud Carlos III, Madrid, Spain
| | - I. Iglesia
- GENUD (Growth, Exercise, NUtrition and Development) Research Group, Faculty of Health Sciences, University of Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (Ciberobn), Instituto de Salud Carlos III, Madrid, Spain
- Instituto Agroalimentario de Aragón (IA2), Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
- Retics ISCIII, Madrid, Spain
| | - M. L. Miguel-Berges
- GENUD (Growth, Exercise, NUtrition and Development) Research Group, Faculty of Health Sciences, University of Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (Ciberobn), Instituto de Salud Carlos III, Madrid, Spain
- Instituto Agroalimentario de Aragón (IA2), Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
| | - V. Vučinić
- GENUD (Growth, Exercise, NUtrition and Development) Research Group, Faculty of Health Sciences, University of Zaragoza, Zaragoza, Spain
| | - L. A. Moreno
- GENUD (Growth, Exercise, NUtrition and Development) Research Group, Faculty of Health Sciences, University of Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (Ciberobn), Instituto de Salud Carlos III, Madrid, Spain
- Instituto Agroalimentario de Aragón (IA2), Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
| | - G. Rodríguez
- GENUD (Growth, Exercise, NUtrition and Development) Research Group, Faculty of Health Sciences, University of Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón (IA2), Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
- Retics ISCIII, Madrid, Spain
- Department of Pediatrics, Radiology and Physical Medicine, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
36
|
Braun T, Filleböck V, Metze B, Bührer C, Plagemann A, Henrich W. Long term alterations of growth after antenatal steroids in preterm twin pregnancies. J Perinat Med 2021; 49:127-137. [PMID: 33010142 DOI: 10.1515/jpm-2020-0204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/11/2020] [Indexed: 02/01/2023]
Abstract
OBJECTIVES To compare the long-term effects of antenatal betamethasone (ANS, ≤16 mg, =24 mg and >24 mg) in twins on infant and childhood growth. METHODS A retrospective cohort follow up study among 198 twins after ANS including three time points: U1 first neonatal examination after birth and in the neonatal period; U7 examination from the 21st to the 24th month of life and U9 examination from the 60th to the 64th month of life using data from copies of the children's examination booklets. Inclusion criteria are twin pregnancies with preterm labor, cervical shortening, preterm premature rupture of membranes, or vaginal bleeding, and exposure to ANS between 23+5 and 33+6 weeks. Outcome measures are dosage-dependent and sex-specific effects of ANS on growth (body weight, body length, head circumference, body mass index and ponderal index) up to 5.3 years. RESULTS Overall, 99 live-born twin pairs were included. Negative effects of ANS on fetal growth persisted beyond birth, altered infant and childhood growth, independent of possible confounding factors. Overall weight percentile significantly decreased between infancy and early childhood by 18.8%. Birth weight percentiles significantly changed in a dose dependent and sex specific manner, most obviously in female-female and mixed pairs. The ponderal index significantly decreased up to 42.9%, BMI index increased by up to 33.8%. CONCLUSIONS ANS results in long-term alterations in infant and childhood growth. Changes between infancy and early childhood in ponderal mass index and BMI, independent of dose or twin pair structure, might indicate an ANS associated increased risk for later life disease. SYNOPSIS First-time report on long-term ANS administration growth effects in twin pregnancies, showing persisting alterations beyond birth in infant and childhood growth up to 5.3 years as potential indicator of later life disease risk.
Collapse
Affiliation(s)
- Thorsten Braun
- Clinic of Obstetrics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Division of Experimental Obstetrics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Vivien Filleböck
- Clinic of Obstetrics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Boris Metze
- Clinic of Neonatology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Christoph Bührer
- Clinic of Neonatology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Andreas Plagemann
- Division of Experimental Obstetrics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Wolfgang Henrich
- Clinic of Obstetrics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
37
|
Kim JH, Lim JS. Early menarche and its consequence in Korean female: reducing fructose intake could be one solution. Clin Exp Pediatr 2021; 64:12-20. [PMID: 32403898 PMCID: PMC7806406 DOI: 10.3345/cep.2019.00353] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 12/27/2019] [Accepted: 02/28/2020] [Indexed: 12/31/2022] Open
Abstract
The mean age at menarche (AAM) of Korean females has been rapidly decreasing over the last 50 years; currently, the prevalence of early menarche (<12 years) is 22.3%. Female adolescents who experience early menarche are known to be at greater risk of psychosocial and behavioral problems along with several physical health problems such as menstrual problems. They also tend to achieve a shorter final height and develop obesity. Population-based Korean studies have shown a strong association between early menarche and the risk of obesity, insulin resistance, metabolic syndrome, nonalcoholic fatty liver disease, diabetes, breast cancer, and cardiovascular disease in adulthood. Although the exact mechanism of how early menarche causes cardiometabolic derangement in later adulthood is unknown, childhood obesity and insulin resistance might be major contributors. Recent studies demonstrated that an excessive consumption of fructose might underlie the development of obesity and insulin resistance along with an earlier AAM. A positive association was observed between sugar-sweetened beverages (a major source of fructose) intake and obesity, metabolic syndrome, insulin resistance, and cardiometabolic risk in Korean females. In pediatrics, establishing risk factors is important in preventing disease in later life. In this regard, early menarche is a simple and good marker for the management of cardiometabolic diseases in adulthood. Decreasing one's fructose intake might prevent early menarche as well as the development of obesity, insulin resistance, and cardiometabolic diseases.
Collapse
Affiliation(s)
- Ji Hyun Kim
- Department of Pediatrics, Dongguk University Ilsan Hospital, Goyang, Korea
| | - Jung Sub Lim
- Department of Pediatrics, Korea Cancer Center Hospital, Seoul, Korea
| |
Collapse
|
38
|
Peng X, Li J, Yan S, Chen J, Lane J, Malard P, Liu F. Xiang Study: an association of breastmilk composition with maternal body mass index and infant growth during the first 3 month of life. Nutr Res Pract 2020; 15:367-381. [PMID: 34093977 PMCID: PMC8155227 DOI: 10.4162/nrp.2021.15.3.367] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/02/2020] [Accepted: 08/25/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND/OBJECTIVES This study aimed to establish a mother and child cohort in the Chinese population, and investigate human breastmilk (HBM) composition and its relationship with maternal body mass index (BMI) and infant growth during the first 3 mon of life. SUBJECTS/METHODS A total of 101 Chinese mother and infant pairs were included in this prospective cohort. Alterations in the milk macronutrients of Chinese mothers at 1 mon (T1), 2 mon (T2), and 3 mon (T3) lactation were analyzed. HBM fatty acid (FA) profiles were measured by gas chromatography (GC), and HBM proteomic profiling was achieved by matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry (MS). RESULTS During the first 3 mon of lactation (P < 0.05), significant decreases were determined in the levels of total energy, fat, protein, and osteopontin (OPN), as well as ratios of long-chain saturated FA (including C16:0, C22:0 and C24:0), monounsaturated FA (including C16:1), and n-6 poly unsaturated FA (PUFA) (including C20:3n-6 and C20:4n-6, and n-6/n-3). Conversely, butyrate, C6:0 and n-3 PUFA C18:3n-3 (α-linolenic acid, ALA) were significantly increased during the first 3 mon (P < 0.05). HBM proteomic analyses distinguished compositional protein differences over time (P = 0.001). Personalized mother-infant analyses demonstrated that HBM from high BMI mothers presented increased total energy, fat, protein and OPN, and increased content of n-6 PUFA (including C18:3n-6, C20:3n-6 and n-6/n-3 ratio) as compared with low BMI mothers (P < 0.05). Furthermore, BMI of the mothers positively correlated with the head circumference (HC) of infants as well as the specific n-6 PUFA C20:3n-6 over the 3 time points examined. Infant HC was negatively associated with C18:0. CONCLUSION This study provides additional evidence to the Chinese HBM database, and further knowledge of FA function. It also helps to establish future maternal strategies that support the healthy growth and development of Chinese infants.
Collapse
Affiliation(s)
- Xuyi Peng
- H&H Group, Global Research and Technology Center, Guangzhou 510700, China.,School of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jie Li
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shuyuan Yan
- Child Health Care Center, Changsha Hospital for Maternal and Child Care, Changsha 410007, China
| | - Juchun Chen
- H&H Group, Global Research and Technology Center, Guangzhou 510700, China
| | - Jonathan Lane
- H&H Group, Global Research and Technology Center, Cork, P61 C996, Ireland
| | - Patrice Malard
- H&H Group, Global Research and Technology Center, Guangzhou 510700, China
| | - Feitong Liu
- H&H Group, Global Research and Technology Center, Guangzhou 510700, China
| |
Collapse
|
39
|
Orsso CE, Colin-Ramirez E, Field CJ, Madsen KL, Prado CM, Haqq AM. Adipose Tissue Development and Expansion from the Womb to Adolescence: An Overview. Nutrients 2020; 12:E2735. [PMID: 32911676 PMCID: PMC7551046 DOI: 10.3390/nu12092735] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/29/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023] Open
Abstract
Prevalence rates of pediatric obesity continue to rise worldwide. Adipose tissue (AT) development and expansion initiate in the fetus and extend throughout the lifespan. This paper presents an overview of the AT developmental trajectories from the intrauterine period to adolescence; factors determining adiposity expansion are also discussed. The greatest fetal increases in AT were observed in the third pregnancy trimester, with growing evidence suggesting that maternal health and nutrition, toxin exposure, and genetic defects impact AT development. From birth up to six months, healthy term newborns experience steep increases in AT; but a subsequent reduction in AT is observed during infancy. Important determinants of AT in infancy identified in this review included feeding practices and factors shaping the gut microbiome. Low AT accrual rates are maintained up to puberty onset, at which time, the pattern of adiposity expansion becomes sex dependent. As girls experience rapid increases and boys experience decreases in AT, sexual dimorphism in hormone secretion can be considered the main contributor for changes. Eating patterns/behaviors and interactions between dietary components, gut microbiome, and immune cells also influence AT expansion. Despite the plasticity of this tissue, substantial evidence supports that adiposity at birth and infancy highly influences its levels across subsequent life stages. Thus, a unique window of opportunity for the prevention and/or slowing down of the predisposition toward obesity, exists from pregnancy through childhood.
Collapse
Affiliation(s)
- Camila E. Orsso
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; (C.E.O.); (C.J.F.); (C.M.P.)
| | | | - Catherine J. Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; (C.E.O.); (C.J.F.); (C.M.P.)
| | - Karen L. Madsen
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2C2, Canada;
| | - Carla M. Prado
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; (C.E.O.); (C.J.F.); (C.M.P.)
| | - Andrea M. Haqq
- Department of Pediatrics and Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R7, Canada
| |
Collapse
|
40
|
Bar-Sadeh B, Rudnizky S, Pnueli L, Bentley GR, Stöger R, Kaplan A, Melamed P. Unravelling the role of epigenetics in reproductive adaptations to early-life environment. Nat Rev Endocrinol 2020; 16:519-533. [PMID: 32620937 DOI: 10.1038/s41574-020-0370-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/11/2020] [Indexed: 11/08/2022]
Abstract
Reproductive function adjusts in response to environmental conditions in order to optimize success. In humans, this plasticity includes age of pubertal onset, hormone levels and age at menopause. These reproductive characteristics vary across populations with distinct lifestyles and following specific childhood events, and point to a role for the early-life environment in shaping adult reproductive trajectories. Epigenetic mechanisms respond to external signals, exert long-term effects on gene expression and have been shown in animal and cellular studies to regulate normal reproductive function, strongly implicating their role in these adaptations. Moreover, human cohort data have revealed differential DNA methylation signatures in proxy tissues that are associated with reproductive phenotypic variation, although the cause-effect relationships are difficult to discern, calling for additional complementary approaches to establish functionality. In this Review, we summarize how adult reproductive function can be shaped by childhood events. We discuss why the influence of the childhood environment on adult reproductive function is an important consideration in understanding how reproduction is regulated and necessitates consideration by clinicians treating women with diverse life histories. The resolution of the molecular mechanisms responsible for human reproductive plasticity could also lead to new approaches for intervention by targeting these epigenetic modifications.
Collapse
Affiliation(s)
- Ben Bar-Sadeh
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Sergei Rudnizky
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Lilach Pnueli
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | | | - Reinhard Stöger
- Department of Biological Sciences, University of Nottingham, Nottingham, UK
| | - Ariel Kaplan
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Philippa Melamed
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
41
|
Kim JH, Kim DH, Lim JS. Growth status of children and adolescents born small for gestational age at full term in Korea: data from the KNHANES-V. J Pediatr Endocrinol Metab 2020; 33:743-750. [PMID: 32447332 DOI: 10.1515/jpem-2019-0471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 03/10/2020] [Indexed: 11/15/2022]
Abstract
Objectives Small for gestational age (SGA) status is known to show stunted growth and results in short stature in adults. The aim of this study was to describe the current short stature in subjects born SGA in Korea and to assess catch-up growth (CUG) or non-CUG. Methods We analyzed data from 3,524 subjects (1,831 male) aged 1-18 years who were born as full-term singletons and who participated in the Fifth Korean National Health and Nutrition Examination Survey (2010-2011). Results The prevalence of SGA was 13.4% (n=471). Subjects born SGA had fathers with shorter height, shorter mother's height, and mid-parental height than non-SGA subjects (p<0.05 for all). The odds ratios (ORs) for SGA birth of a short statured father and a short statured mother were 2.00 (95% CI; 1.15-3.47) and 2.11 (95% CI; 1.30-3.40), respectively. Among 471 SGA subjects, 28 subjects (5.9%) were non-CUG, which made up 36.4% of all subjects with short stature. The CUG subjects had a higher father's height, mother's height, mid-parental height, and current BMI (p<0.05 for all). The non-CUG subjects had a higher percentage of fathers being near-short stature (height<10th percentile; 33.3 vs. 12.7%; p=0.008) and mothers being near-short stature (39.3 vs. 13.9%; p<0.001). Conclusion Korean subjects born SGA had a higher risk of current short stature. This population-based nationwide survey also showed that both father's and mother's short stature are risk factors of not only SGA birth but also non-CUG in their children.
Collapse
Affiliation(s)
- Ji Hyun Kim
- Department of Pediatrics, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| | - Dong Ho Kim
- Department of Pediatrics, Korea Cancer Center Hospital, Seoul, Republic of Korea
| | - Jung Sub Lim
- Department of Pediatrics, Korea Cancer Center Hospital, 215 Gongneungdong, Nowon-gu, 01812, Seoul, Republic of Korea
| |
Collapse
|
42
|
Ong KK, Cheng TS, Olga L, Prentice PM, Petry CJ, Hughes IA, Dunger DB. Which infancy growth parameters are associated with later adiposity? The Cambridge Baby Growth Study. Ann Hum Biol 2020; 47:142-149. [PMID: 32429763 PMCID: PMC7261401 DOI: 10.1080/03014460.2020.1745887] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/28/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022]
Abstract
Background: Highly consistent positive associations are reported between infancy growth and later obesity risk. However, it is unclear whether infancy growth parameters beyond body weight add to the prediction of later obesity risk.Aim: To assess whether infancy length and skinfold thicknesses add to infancy weight in the prediction of childhood adiposity.Subjects and methods: This analysis included 254 children with available data on infant growth from birth to 24 months and childhood adiposity at age 6-11 years measured by DXA. Multilevel linear regression was used to examine the predictors of childhood percent body fat (%BF), with adjustment for sex and age at follow-up visit.Results: Birth weight and weight gain (modelled as changes in z-score) between 0-3 months and 3-24 months showed independent positive relationships with childhood %BF. The addition of gains in infant length and skinfolds between 0-3 months, but not 3-24 months, improved overall model prediction, from 18.7% to 20.7% of the variance in childhood %BF (likelihood ratio test, p < 0.0001), although their independent effect estimates were small (infant length gain: negative trend, partial R-square 0.6%, p = 0.2; skinfolds: positive trend, 1.3%, p = 0.09).Conclusion: Infancy length and skinfolds contribute significantly, but only modestly, to the prediction of childhood adiposity.
Collapse
Affiliation(s)
- Ken K. Ong
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Tuck Seng Cheng
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - L. Olga
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - P. M. Prentice
- Department of Paediatrics, University of Cambridge, Cambridge, UK
- North Middlesex University Hospital NHS Trust, London, UK
| | - C. J. Petry
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - I. A. Hughes
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - D. B. Dunger
- Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| |
Collapse
|
43
|
English S, Wright I, Ashburn V, Ford G, Caramaschi D. Prenatal anxiety, breastfeeding and child growth and puberty: linking evolutionary models with human cohort studies. Ann Hum Biol 2020; 47:106-115. [PMID: 32429755 PMCID: PMC7261397 DOI: 10.1080/03014460.2020.1751286] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/25/2020] [Accepted: 03/31/2020] [Indexed: 11/23/2022]
Abstract
Background: Stress experienced by mothers during pregnancy can have both immediate and long-term effects on child development, potentially mediated by breastfeeding.Aim: Using a UK birth cohort study, we asked how maternal stress relates to breastfeeding and consequences for growth and puberty onset.Subjects and methods: We analysed data from the Avon Longitudinal Study of Parents and Children, collected via questionnaires and clinic visits (N: 698-8,506). We used reports of prenatal anxiety, breastfeeding, early growth and age at menarche or first voice change. Confounding by maternal age, parity, smoking, education and body mass index (BMI) was considered.Results: Mothers with higher levels of reported anxiety were less likely to breastfeed (Odds ratio (OR): 0.83, 95% confidence interval (CI): 0.71, 0.97). Breastfed infants had slower growth before weaning, although growth differences were unclear thereafter. Being breastfed for more than six months was associated with later puberty onset in females (2.76 months later than non-breastfed; CI: 0.9, 4.63), although the association was attenuated by confounders and BMI (1.51 months, CI: -0.38, 3.40). No association between breastfeeding and puberty onset in males was found.Conclusion: Our studies fit results shown previously, and we consider these in light of evolutionary life history theory while discussing key challenges in such an approach.
Collapse
Affiliation(s)
- Sinead English
- School of Biological Sciences, University of Bristol, Bristol, UK
| | - India Wright
- Reproduction and Development Programme, Bristol Medical School (Translational Health Sciences), University of Bristol, Bristol, UK
| | - Verity Ashburn
- Reproduction and Development Programme, Bristol Medical School (Translational Health Sciences), University of Bristol, Bristol, UK
| | - Gemma Ford
- Reproduction and Development Programme, Bristol Medical School (Translational Health Sciences), University of Bristol, Bristol, UK
| | - Doretta Caramaschi
- Medical Research Council Integrative Epidemiology Unit, Bristol Medical School (Population Health Sciences), University of Bristol, Bristol, UK
| |
Collapse
|
44
|
Pesch MH, Pont CM, Lumeng JC, McCaffery H, Tan CC. Mother and Infant Predictors of Rapid Infant Weight Gain. Clin Pediatr (Phila) 2019; 58:1515-1521. [PMID: 31556703 DOI: 10.1177/0009922819877875] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Objective. To examine characteristics of the infant and mother associated with rapid infant weight gain (RIWG). Methods. Electronic health records (N = 4626) of term infants born were reviewed. Multivariable logistic regression examined the presence of RIWG (vs not) using participant characteristics in the whole sample and in stratified groups. Results. The prevalence of RIWG was 18.7%. Predictors of RIWG were infant male sex, younger infant gestational age, firstborn (vs later born) status, maternal Black or Other (Asian, American Indian, etc), non-Hispanic race/ethnicity (vs White non-Hispanic), Medicaid (vs non-Medicaid insurance), and maternal cigarette smoking status (vs never smoker). The regression model explained between 7.0% and 11.4% of the variance in RIWG. There were few differences in predictors of RIWG in stratified samples. Conclusions. Early childhood obesity intervention efforts may target the modifiable risk factors for RIWG starting prenatally.
Collapse
Affiliation(s)
| | | | | | | | - Cin C Tan
- University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
45
|
Chen YC, Fan HY, Yang C, Hsieh RH, Pan WH, Lee YL. Assessing causality between childhood adiposity and early puberty: A bidirectional Mendelian randomization and longitudinal study. Metabolism 2019; 100:153961. [PMID: 31422054 DOI: 10.1016/j.metabol.2019.153961] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/31/2019] [Accepted: 08/11/2019] [Indexed: 01/27/2023]
Abstract
AIMS Obesity and early puberty have been reported to be mutually causative. We investigated the causal relationship between adiposity and early puberty by performing bidirectional Mendelian randomization (MR) and longitudinal data analyses. METHODS We used information from the Taiwan Children Health Study (3109 adolescents aged 11-12 years) with 17 body mass index (BMI)- and 10 puberty-related single-nucleotide polymorphisms (SNPs) to produce genetic instrumental variables (IVs). The two-stage least squares (2SLS) method, MR sensitivity analysis, and survival analysis were used to explore and confirm causality. RESULTS Regression estimates from IVs revealed that significantly increased association of BMI with early puberty was noted (coefficients: 0.13, 0.10, and 0.09; 95% CI: 0.07-0.19, 0.02-0.19, and 0.02-0.16 for all participants, male adolescents, and female adolescents, respectively). Genetic IVs for puberty were not associated with BMI. MR sensitivity and two-sample MR analyses produced similar results. Longitudinal analysis results revealed that prepubertal overweight and obesity could predict early onset of puberty. However, after excluding children with a history of overweight and obesity at the age of 7-12 years, early puberty was not found to trigger new-onset of overweight and obesity at the age of 18 years in either sex. CONCLUSIONS Higher adiposity may lead to early puberty. However, the causal effects of early puberty on adiposity accumulation were not supported by our data. Targeted interventions to reduce childhood obesity are strongly recommended to prevent obesity-related comorbidities, as well as early puberty onset.
Collapse
Affiliation(s)
- Yang-Ching Chen
- Department of Family Medicine, Taipei Medical University Hospital, Taipei, Taiwan; Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei, Taiwan
| | - Hsien-Yu Fan
- Department of Family Medicine, Taipei Medical University Hospital, Taipei, Taiwan; Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen Yang
- Department of Pediatrics, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Rong-Hong Hsieh
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Wen-Harn Pan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yungling L Lee
- Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
46
|
Barrios PL, Garcia-Feregrino R, Rivera JA, Barraza-Villarreal A, Hernández-Cadena L, Romieu I, Gonzalez-Casanova I, Ramakrishnan U, Hoffman DJ. Height Trajectory During Early Childhood Is Inversely Associated with Fat Mass in Later Childhood in Mexican Boys. J Nutr 2019; 149:2011-2019. [PMID: 31334762 PMCID: PMC6825831 DOI: 10.1093/jn/nxz157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/22/2019] [Accepted: 06/11/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Childhood obesity continues to be a global health problem. Previous research suggests that linear growth retardation or stunting during early childhood increases the risk of obesity, but others have reported that rapid linear growth poses a greater concern than early nutritional status. OBJECTIVE The objective of this study was to determine if growth trajectories are associated with body composition at age 8-10 y. METHODS Study participants consisted of 255 girls and 281 boys who participated in a follow-up of the Prenatal Omega-3 Fatty Acid Supplementation and Child Growth and Development (POSGRAD) Study. Sex-specific latent height class (LHC) trajectories were derived from 11 measures of height from birth to 5 y of age and used to calculate 3 distinct growth classes for boys (low, intermediate, and high) and 2 distinct classes for girls (low and high). Body composition at age 8-10 y was estimated using bioelectrical impedance analysis. Multivariable linear regression analysis was used to determine the relationship between growth trajectory classes and fat mass (FM) and fat-free mass (FFM) in late childhood, controlling for confounding factors. RESULTS In girls, there were no significant associations between LHC and FM or FFM. In boys, relative to the intermediate LHC, the low LHC had higher FM (β = 0.69 kg; 95% CI: 0.26-1.11 kg) and the high LHC had lower FM (β = -0.40 kg; 95% CI: -0.76 to -0.05 kg). Boys in the low LHC had significantly less FFM (β = -0.69 kg; 95% CI: -1.11 to -0.26 kg), and boys in the high LHC had more FFM (β = 0.40 kg; 95% CI: 0.05-0.76 kg) compared with the intermediate LHC. CONCLUSION Gain in height among boys, but not girls, in early childhood was associated with lower adiposity in late childhood compared with children with a slower rate of growth. Clinical trial registration number: NCT00646360.
Collapse
Affiliation(s)
- Pamela L Barrios
- Department of Nutritional Sciences, Program in International Nutrition, Center for Childhood Nutrition Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers, the State University of New Jersey, New Brunswick, NJ, USA
| | - Raquel Garcia-Feregrino
- Health and Nutrition Research Center, National Institute of Public Health, Cuernavaca, Mexico
| | - Juan A Rivera
- National Institute of Public Health, Cuernavaca, Mexico
| | | | | | - Isabel Romieu
- Population Health Research Center, National Institute of Public Health, Cuernavaca, Mexico
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Ines Gonzalez-Casanova
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Usha Ramakrishnan
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Daniel J Hoffman
- Department of Nutritional Sciences, Program in International Nutrition, Center for Childhood Nutrition Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers, the State University of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
47
|
Rotevatn TA, Overgaard C, Melendez-Torres GJ, Mortensen RN, Ullits LR, Høstgaard AMB, Torp-Pedersen C, Bøggild H. Infancy weight gain, parental socioeconomic position, and childhood overweight and obesity: a Danish register-based cohort study. BMC Public Health 2019; 19:1209. [PMID: 31477065 PMCID: PMC6720844 DOI: 10.1186/s12889-019-7537-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/22/2019] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Rapid infant weight gain (RIWG) is a very strong predictor of childhood overweight and obesity (COO). Socioeconomic position (SEP) is also related to the risk of COO and parents of different SEP may differ in their reaction to accelerated infant weight gain. Together this could lead to differences in how weight gain and COO risk relate across SEP. This study aimed to analyse possible interaction of SEP and RIWG on COO risk. METHODS A register-based longitudinal cohort study followed 19,894 healthy, term infants, born in Denmark between December 2011 and May 2015. Logistic regression models were used to estimate odds ratios (OR) of COO risk at 2 years (22-26 months) of age with 95% confidence intervals (95% CI) for categories of infancy weight gain based on changes in weight-for-age z-scores between 0 and 8-10 months of age (slow (<- 0.67), mean (- 0.67-0.67), rapid (> 0.67-1.34) and very rapid (> 1.34)). Possible multiplicative and additive interaction of SEP (based on household income and maternal education) on the relationship between infancy weight gain and COO were analysed. RESULTS In total, 19.1 and 15.1% experienced rapid or very rapid weight gain, respectively, and 1497 (7.5%) children were classified with COO at follow-up. These prevalences were higher in those with lower levels of SEP. Adjusted OR for COO were 3.09 (95% CI [2.66-3.59]) and 7.58 (95% CI [6.51-8.83]) for rapid and very rapid weight gain, respectively, when household income was included in the model. Results were similar in the model including maternal education. No signs of interactions were detected on a multiplicative scale. Weak signs of additive interaction were present, but these values did not reach significance. CONCLUSION Both rapid and very rapid weight gain were associated with substantially higher risks of COO but these associations were not modified by SEP. This indicates that promotion of healthy weight gain should take place in all population groups irrespective of their SEP.
Collapse
Affiliation(s)
- Torill Alise Rotevatn
- Public Health and Epidemiology Group, Department of Health Science and Technology, Aalborg University, Niels Jernes Vej 14, 9220, Aalborg East, Denmark.
| | - Charlotte Overgaard
- Public Health and Epidemiology Group, Department of Health Science and Technology, Aalborg University, Niels Jernes Vej 14, 9220, Aalborg East, Denmark
| | - G J Melendez-Torres
- DECIPHer, Cardiff School of Social Sciences, Cardiff University, 1-3 Museum Place, Cardiff, CF10 3BD, Wales, UK
| | - Rikke Nørmark Mortensen
- Unit of Epidemiology and Biostatistics, Aalborg University Hospital, Søndre Skovvej 15, 9000, Aalborg, Denmark
| | - Line Rosenkilde Ullits
- Public Health and Epidemiology Group, Department of Health Science and Technology, Aalborg University, Niels Jernes Vej 14, 9220, Aalborg East, Denmark
| | - Anna Marie Balling Høstgaard
- Public Health and Epidemiology Group, Department of Health Science and Technology, Aalborg University, Niels Jernes Vej 14, 9220, Aalborg East, Denmark
| | - Christian Torp-Pedersen
- Public Health and Epidemiology Group, Department of Health Science and Technology, Aalborg University, Niels Jernes Vej 14, 9220, Aalborg East, Denmark.,Unit of Epidemiology and Biostatistics, Aalborg University Hospital, Søndre Skovvej 15, 9000, Aalborg, Denmark
| | - Henrik Bøggild
- Public Health and Epidemiology Group, Department of Health Science and Technology, Aalborg University, Niels Jernes Vej 14, 9220, Aalborg East, Denmark.,Unit of Epidemiology and Biostatistics, Aalborg University Hospital, Søndre Skovvej 15, 9000, Aalborg, Denmark
| |
Collapse
|
48
|
Cohn BA, La Merrill MA, Krigbaum NY, Wang M, Park JS, Petreas M, Yeh G, Hovey RC, Zimmermann L, Cirillo PM. In utero exposure to poly- and perfluoroalkyl substances (PFASs) and subsequent breast cancer. Reprod Toxicol 2019; 92:112-119. [PMID: 31323350 DOI: 10.1016/j.reprotox.2019.06.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 05/08/2019] [Accepted: 06/28/2019] [Indexed: 01/09/2023]
Abstract
We tested the hypothesis that maternal perinatal serum levels of poly and perfluoroalkyl substances (PFASs) predict risk for breast cancer in daughters in a 54-year follow-up of 9300 daughters born 1959-1967 in the Child Health and Development Studies pregnancy cohort. Total cholesterol and PFASs were measured in archived maternal perinatal serum for 102 daughter breast cancer cases diagnosed by age 52, and 310 controls matched on birth year and blood draw trimester. High maternal N-ethyl-perfluorooctane sulfonamido acetic acid (EtFOSAA), a precursor of perfluorooctane sulfonic acid (PFOS), in combination with high maternal total cholesterol predicted a 3.6-fold increased risk of breast cancer (pinteraction<0.05). Conversely, maternal PFOS was associated with decreased daughters' breast cancer risk. Predictions were robust to alternative modeling and independent of other maternal factors. Future generations continue to be exposed to ubiquitous, persistent PFASs. These findings are relevant to breast cancer prevention if confirmed experimentally and where possible, in additional epidemiology studies of internal doses of PFASs and other chemical mixtures especially during vulnerable windows in early life.
Collapse
Affiliation(s)
- Barbara A Cohn
- Child Health and Development Studies, Public Health Institute, Berkeley, CA, USA.
| | - Michele A La Merrill
- Department of Environmental Toxicology, Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Nickilou Y Krigbaum
- Child Health and Development Studies, Public Health Institute, Berkeley, CA, USA
| | - Miaomiao Wang
- Environmental Chemistry Laboratory, California Department of Toxic Substances Control, Berkeley, California, USA
| | - June-Soo Park
- Environmental Chemistry Laboratory, California Department of Toxic Substances Control, Berkeley, California, USA
| | - Myrto Petreas
- Environmental Chemistry Laboratory, California Department of Toxic Substances Control, Berkeley, California, USA
| | - Gregory Yeh
- Environmental Chemistry Laboratory, California Department of Toxic Substances Control, Berkeley, California, USA
| | - Russell C Hovey
- Department of Animal Science, University of California, Davis, CA, USA
| | - Lauren Zimmermann
- Child Health and Development Studies, Public Health Institute, Berkeley, CA, USA
| | - Piera M Cirillo
- Child Health and Development Studies, Public Health Institute, Berkeley, CA, USA
| |
Collapse
|
49
|
Juul A, Skakkebæk NE. Why Do Normal Children Have Acromegalic Levels of IGF-I During Puberty? J Clin Endocrinol Metab 2019; 104:2770-2776. [PMID: 30840065 DOI: 10.1210/jc.2018-02099] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/21/2018] [Indexed: 02/13/2023]
Abstract
CONTEXT The rapid pubertal height growth is unique to humans, but why do we have it? Although the spurt contributes 13% to 15% to the final adult height, we hypothesized that the biological significance of the high acromegalic levels of GH and IGF-I, which are behind the pubertal growth spurt, might primarily occur to stimulate the reproductive organs. EVIDENCE SYNTHESIS Animal data have demonstrated that adult Igf1 and Igf2 gene knockout mice that survive show a dramatic reduction in the size of the reproductive organs and are infertile. In humans, case reports of mutations in the genes affecting the GH-IGF axis and growth (GH, GHRH, GH-R, STAT5b, IGF-I, IGF-II, IGF-1R, PAPPA2) are also characterized by delayed pubertal onset and micropenis. Furthermore, GH treatment will tend to normalize the penile size in patients with GH deficiency. Thus, the endocrine effects of high IGF-I levels might be needed for the transition of the sexual organs, including the secondary sex characteristics, from the "dormant" stages of childhood into fully functioning reproductive systems. The peak IGF-I levels, on average, occur 2 years after the peak height growth velocity, suggesting reasons other than longitudinal growth for the high IGF-I levels, and remain high in the years after the height spurt, when the reproductive systems become fully functional. CONCLUSION We suggest that the serum levels of IGF-I should be monitored in children with poor development of sexual organs, although it remains to be investigated whether GH should be added to sex steroids in the management of hypogonadism for some pubertal children (e.g., boys with micropenis).
Collapse
Affiliation(s)
- Anders Juul
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen Ø, Denmark
- The International Research Centre in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Copenhagen Ø, Denmark
| | - Niels E Skakkebæk
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen Ø, Denmark
- The International Research Centre in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Copenhagen Ø, Denmark
| |
Collapse
|
50
|
Prentice PM, Schoemaker MH, Vervoort J, Hettinga K, Lambers TT, van Tol EAF, Acerini CL, Olga L, Petry CJ, Hughes IA, Koulman A, Ong KK, Dunger DB. Human Milk Short-Chain Fatty Acid Composition is Associated with Adiposity Outcomes in Infants. J Nutr 2019; 149:716-722. [PMID: 31050748 DOI: 10.1093/jn/nxy320] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/25/2018] [Accepted: 12/18/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Presumed benefits of human milk (HM) in avoiding rapid infancy weight gain and later obesity could relate to its nutrient composition. However, data on breast milk composition and its relation with growth are sparse. OBJECTIVE We investigated whether short-chain fatty acids (SCFAs), known to be present in HM and linked to energy metabolism, are associated with infancy anthropometrics. METHODS In a prospective birth cohort, HM hindmilk samples were collected from 619 lactating mothers at 4-8 wk postnatally [median (IQR) age: 33.9 (31.3-36.5) y, body mass index (BMI) (kg/m2): 22.8 (20.9-25.2)]. Their offspring, born at 40.1 (39.1-41.0) wk gestation with weight 3.56 (3.22-3.87) kg and 51% male, were assessed with measurement of weight, length, and skinfold thickness at ages 3, 12, and 24 mo, and transformed to age- and sex-adjusted z scores. HM SCFAs were measured by 1H-nuclear magnetic resonance spectroscopy (NMR) and GC-MS. Multivariable linear regression models were conducted to analyze the relations between NMR HM SCFAs and infancy growth parameters with adjustment for potential confounders. RESULTS NMR peaks for HM butyrate, acetate, and formic acid, but not propionate, were detected. Butyrate peaks were 17.8% higher in HM from exclusively breastfeeding mothers than mixed-feeding mothers (P = 0.003). HM butyrate peak values were negatively associated with changes in infant weight (standardized B = -0.10, P = 0.019) and BMI (B = -0.10, P = 0.018) between 3 and 12 mo, and negatively associated with BMI (B = -0.10, P = 0.018) and mean skinfold thickness (B = -0.10, P = 0.049) at age 12 mo. HM formic acid peak values showed a consistent negative association with infant BMI at all time points (B < = -0.10, P < = 0.014), whereas HM acetate was negatively associated with skinfold thickness at 3 mo (B = -0.10, P = 0.028) and 24 mo (B = -0.10, P = 0.036). CONCLUSIONS These results suggest that HM SCFAs play a beneficial role in weight gain and adiposity during infancy. Further knowledge of HM SCFA function may inform future strategies to support healthy growth.
Collapse
Affiliation(s)
- Philippa M Prentice
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | | | - Jacques Vervoort
- Department of Agrotechnology and Food Sciences, Wageningen University, the Netherlands
| | - Kasper Hettinga
- Department of Agrotechnology and Food Sciences, Wageningen University, the Netherlands
| | - Tim T Lambers
- Mead Johnson Pediatric Nutrition Institute, Nijmegen, the Netherlands
| | - Eric A F van Tol
- Mead Johnson Pediatric Nutrition Institute, Nijmegen, the Netherlands
| | - Carlo L Acerini
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - Laurentya Olga
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - Clive J Petry
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - Ieuan A Hughes
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - Albert Koulman
- MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, NIHR Cambridge Comprehensive Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Ken K Ong
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom.,Wellcome Trust-MRC Institute of Metabolic Science, NIHR Cambridge Comprehensive Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom.,MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, NIHR Cambridge Comprehensive Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - David B Dunger
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom.,Wellcome Trust-MRC Institute of Metabolic Science, NIHR Cambridge Comprehensive Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|