1
|
Zhong C, Li X, Guan D, Zhang B, Wang X, Qu L, Zhou H, Fang L, Sun C, Yang N. Age-dependent genetic architectures of chicken body weight explored by multidimensional GWAS and molQTL analyses. J Genet Genomics 2024:S1673-8527(24)00240-6. [PMID: 39306327 DOI: 10.1016/j.jgg.2024.09.003] [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: 04/24/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 11/11/2024]
Abstract
Chicken body weight (BW) is a critical trait in breeding. Although genetic variants associated with BW have been investigated by genome-wide association studies (GWAS), the contributions of causal variants and their molecular mechanisms remain largely unclear in chickens. In this study, we construct a comprehensive genetic atlas of chicken BW by integrative analysis of 30 age points and 5 quantitative trait loci (QTL) across 27 tissues. We find that chicken growth is a cumulative non-linear process, which can be divided into three distinct stages. Our GWAS analysis reveals that BW-related genetic variations show ordered patterns in these three stages. Genetic variations in chromosome 1 may regulate the overall growth process, likely by modulating the hypothalamus-specific expression of SLC25A30 and retina-specific expression of NEK3. Moreover, genetic variations in chromosome 4 and chromosome 27 may play dominant roles in regulating BW during Stage Ⅱ (8-22 weeks) and Stage Ⅲ (23-72 weeks), respectively. In summary, our study presents a comprehensive genetic atlas regulating developmental stage-specific changes in chicken BW, thus providing important resources for genomic selection in breeding programs.
Collapse
Affiliation(s)
- Conghao Zhong
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China
| | - Xiaochang Li
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China
| | - Dailu Guan
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Boxuan Zhang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China
| | - Xiqiong Wang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China
| | - Liang Qu
- Jiangsu Institute of Poultry Science, Yangzhou, Jiangsu 225125, China
| | - Huaijun Zhou
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Lingzhao Fang
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus, 8000, Denmark
| | - Congjiao Sun
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China.
| | - Ning Yang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
2
|
Hull HR, Brown A, Gajewski B, Sullivan DK, Carlson SE. The Effect of Prenatal Docosahexaenoic Acid Supplementation on Offspring Fat Mass and Distribution at 24 Months Old. Curr Dev Nutr 2024; 8:103771. [PMID: 38948108 PMCID: PMC11214179 DOI: 10.1016/j.cdnut.2024.103771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 07/02/2024] Open
Abstract
Background Excessive gestational weight gain (GWG) is related to increased offspring fat accrual, and increased fat mass (FM) is related to obesity development. Prenatal DHA supplementation has been linked to lower levels of offspring FM; however, conflicting data exist. Objectives This study aimed to determine if there is a protective effect of prenatal DHA supplementation on offspring fat accrual and adipose tissue deposition at 24 mo in offspring born to females who gain excessive weight compared with nonexcessive weight during pregnancy. We also explored if the effect of DHA dose on FM differed by offspring sex. Methods Infants born to females who participated in the Assessment of DHA on Reducing Early Preterm Birth randomized controlled trial (ADORE) were recruited. In ADORE, females were randomly assigned to either a high or low prenatal DHA supplement. Offspring body composition and adipose tissue distribution were measured using dual-energy x-ray absorptiometry (DXA). GWG was categorized as excessive or not excessive based on clinical guidelines. Results For total FM, there was a significant main effect for the DHA dose (P = 0.03); however, the dose by GWG status was nonsignificant (P = 0.44). Therefore, a higher prenatal DHA dose was related to greater offspring FM (622.9 g greater) and unrelated to GWG status. When investigating a DHA dose by sex effect, a significant main effect for DHA dose (P = 0.01) was detected for central FM. However, no interaction was detected (P = 0.98), meaning that both boys and girls had greater central FM if their mother was assigned to the higher DHA dose. Conclusions Greater prenatal DHA supplementation was associated with greater offspring FM and adipose tissue distribution at 24 mo. It will be important to understand if these effects persist into childhood.This trial was registered at clinicaltrials.gov as NCT03310983.
Collapse
Affiliation(s)
- Holly R Hull
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
| | - Alexandra Brown
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Byron Gajewski
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Debra K Sullivan
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
| | - Susan E Carlson
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
3
|
Hashemi Javaheri FS, Karbin K, Senobari MA, Hakim HG, Hashemi M. The association between maternal body mass index and breast milk composition: a systematic review. Nutr Rev 2024:nuad174. [PMID: 38273741 DOI: 10.1093/nutrit/nuad174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024] Open
Abstract
CONTEXT Breast milk composition is influenced by many factors, ranging from maternal nutritional status to infant sex. Previous studies have explored the relationship between maternal body mass index (BMI) and breast milk composition; however, the findings have been inconsistent and controversial. OBJECTIVE To systematically review the evidence on the association of maternal weight and BMI with breast milk composition. DATA SOURCES The PubMed and Scopus databases were searched up to May 3, 2023, using the following search strategy: ("maternal weight" OR "maternal BMI" OR "mother's weight" OR "mother's BMI") AND ("maternal milk" OR "human milk" OR "breast milk"). DATA EXTRACTION A total of 83 publications, involving data from more than 11 310 lactating women, were identified. All extracted data were compiled, compared, and critically analyzed. DATA ANALYSIS Overall, maternal BMI was associated with higher levels of leptin and insulin, and the ratio of omega-6 to omega-3 polyunsaturated fatty acids in breast milk. However, no conclusive associations were found between maternal BMI and the levels of energy, macronutrients, micronutrients, and other components of breast milk. CONCLUSIONS This systematic review provides robust evidence supporting a positive correlation between maternal BMI and breast milk concentrations of leptin, insulin, and the omega-6 to omega-3 polyunsaturated fatty acid ratio. Nevertheless, disparities in findings are noticeable for other constituents of breast milk. To comprehensively grasp the influence of maternal weight and BMI on breast milk composition, further research endeavors are imperative. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42023458667.
Collapse
Affiliation(s)
- Fatemeh Sadat Hashemi Javaheri
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Karbin
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Mohammad Amin Senobari
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hakime Ghadiri Hakim
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Hashemi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Hou M, Qiu C. Ultra-Processed Food as Mediator of the Association between Birthweight and Childhood Body Weight Outcomes: A Retrospective Cohort Study. Nutrients 2023; 15:4178. [PMID: 37836460 PMCID: PMC10574691 DOI: 10.3390/nu15194178] [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: 08/25/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 10/15/2023] Open
Abstract
Previous studies have shown conflicting findings regarding the association between birthweight and childhood adiposity. We aimed to explore the interaction between ultra-processed food (UPF) and birthweight and its associations with bodyweight markers. The retrospective analysis of data from a Multicity Cohort Study across eastern China was conducted. UPF was computed as percentage of the energy intake and categorized into quartiles. Birthweight was categorized into low (LBW), normal (NBW) and high (HBW). The BMI z-score was calculated using the lambda-mu-sigma method. The sex- and age-specific BMI cutoff points were used to define weight status. Generalized linear models were used to examine modification effects and were performed after adjustment for covariates. The mean percentage of energy intake from UPF was 27.7% among 1370 children. Of all children, 2.3% and 21.4% were born with LBW and HBW, respectively. HBW was a permanent risk for high BMI measures, while LBW was associated with increased BMI measurements only by the addition of the interaction term. The subgroup analysis revealed that HBW and LBW were positively associated with BMI measurements in the lowest UPF intake (Q1), while HBW was related to high BMI measures in Q4. Our findings support efforts to recommend limiting UPF intake, especially for LBW children.
Collapse
Affiliation(s)
- Min Hou
- School of Public Health, College of Medicine, Shanghai Jiao Tong University, 227 Chongqing South Road, Shanghai 200025, China
| | - Chao Qiu
- College of Humanities, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| |
Collapse
|
5
|
Hull HR, Gajewski BJ, Sullivan DK, Carson SE. Growth and adiposity in newborns study (GAINS): The influence of prenatal DHA supplementation protocol. Contemp Clin Trials 2023; 132:107279. [PMID: 37406769 PMCID: PMC10852997 DOI: 10.1016/j.cct.2023.107279] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/22/2023] [Accepted: 06/30/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Obesity and central fat mass (FM) accrual drive disease development and are related to greater morbidity and mortality. Excessive gestational weight gain (GWG) increases fetal fat accretion resulting in greater offspring FM across the lifespan. Studies associate greater maternal docosahexaenoic acid (DHA) levels with lower offspring FM and lower visceral adipose tissue during childhood, however, most U.S. pregnant women do not consume an adequate amount of DHA. We will determine if prenatal DHA supplementation is protective for body composition changes during infancy and toddlerhood in offspring exposed to excessive GWG. METHODS AND DESIGN Infants born to women who participated in the Assessment of DHA on Reducing Early Preterm Birth randomized controlled trial (ADORE; NCT02626299) will be invited to participate. Women were randomized to either a high 1000 mg or low 200 mg daily prenatal DHA supplement starting in the first trimester of pregnancy. Offspring body composition and adipose tissue distribution will be measured at 2 weeks, 6 months, 12 months, and 24 months using dual energy x-ray absorptiometry. Maternal GWG will be categorized as excessive or not excessive based on clinical guidelines. DISCUSSION Effective strategies to prevent obesity development are lacking. Exposures during the prenatal period are important in the establishment of the offspring phenotype. However, it is largely unknown which exposures can be successfully targeted to have a meaningful impact. This study will determine if prenatal DHA supplementation modifies the relationship between maternal weight gain and offspring FM and FM distribution at 24 months of age. ETHICS AND DISSEMINATION The University of Kansas Medical Center Institutional Review Board (IRB) approved the study protocol (STUDY00140895). The results of the trial will be disseminated at conferences and in peer reviewed publications. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT03310983.
Collapse
Affiliation(s)
- Holly R Hull
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States of America.
| | - Byron J Gajewski
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Debra K Sullivan
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Susan E Carson
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States of America
| |
Collapse
|
6
|
Abbas MA, Al-Saigh NN, Saqallah FG. Regulation of adipogenesis by exosomal milk miRNA. Rev Endocr Metab Disord 2023; 24:297-316. [PMID: 36692804 DOI: 10.1007/s11154-023-09788-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/12/2023] [Indexed: 01/25/2023]
Abstract
Milk is a rich source of miRNA packaged in exosomes. Evidence for the systemic uptake and tissue distribution of milk exosomes was reported in newborn and adult humans and animals. Breastfeeding in infants was associated with a reduced risk of obesity. Numerous adipogenesis-related miRNAs have been detected in human milk exosomes. It has been demonstrated that ingested exosomal milk miRNAs may alter gene expression in offspring to regulate their metabolism and growth. In humans, consumption of other species' milk, such as cows and goats, is continued through adulthood. Since miRNAs are conserved, the concern of cross-species transfer of adipogenic miRNA has been raised in recent years, and the increase in obesity worldwide was attributed partially to dairy milk consumption by humans. However, evidence is still weak. Research emphasizes the need for an adequate number of exosomal milk's miRNAs to reach the target cell for biological action to be achieved. It was reported that obese women's milk had less miRNA-148a and miRNA-30b, which may affect the fat acquisition of their babies. Some exosomal milk miRNAs, such as miRNA-29, miRNA-148, miRNA-30b and miRNA-125b, may have epigenetic effects on milk recipients. Moreover, the ability of milk exosomes to cross the gastrointestinal barrier makes them a promising oral drug delivery tool. Yet, exosomes may also be tagged with specific ligands which target certain tissues. Thus, milk exosomes can be engineered and loaded with certain miRNAs responsible for adipocyte differentiation, conversion, or browning. Modifications in the miRNA cargo of exosomes can benefit human health and be an alternative to traditional drugs.
Collapse
Affiliation(s)
- Manal A Abbas
- Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, 19328, Jordan.
- Pharmacological and Diagnostic Research Center, Al-Ahliyya Amman University, Amman, 19328, Jordan.
| | - Noor Nadhim Al-Saigh
- Department of Basic Medical Sciences, Faculty of Medicine, Ibn Sina University for Medical Siences, Amman, 11104, Jordan
| | - Fadi G Saqallah
- Discipline of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
| |
Collapse
|
7
|
Tain YL, Hsu CN. Metabolic Syndrome Programming and Reprogramming: Mechanistic Aspects of Oxidative Stress. Antioxidants (Basel) 2022; 11:2108. [PMID: 36358480 PMCID: PMC9686950 DOI: 10.3390/antiox11112108] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/06/2022] [Accepted: 10/21/2022] [Indexed: 11/22/2023] Open
Abstract
Metabolic syndrome (MetS) is a worldwide public health issue characterized by a set of risk factors for cardiovascular disease. MetS can originate in early life by developmental programming. Increasing evidence suggests that oxidative stress, which is characterized as an imbalance between reactive oxygen species (ROS), nitric oxide (NO), and antioxidant systems, plays a decisive role in MetS programming. Results from human and animal studies indicate that maternal-derived insults induce MetS later in life, accompanied by oxidative stress programming of various organ systems. On the contrary, perinatal use of antioxidants can offset oxidative stress and thereby prevent MetS traits in adult offspring. This review provides an overview of current knowledge about the core mechanisms behind MetS programming, with particular focus on the occurrence of oxidative-stress-related pathogenesis as well as the use of potential oxidative-stress-targeted interventions as a reprogramming strategy to avert MetS of developmental origins. Future clinical studies should provide important proof of concept for the effectiveness of these reprogramming interventions to prevent a MetS epidemic.
Collapse
Affiliation(s)
- You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
8
|
Kelly AC, J Rosario F, Chan J, Cox LA, Powell TL, Jansson T. Transcriptomic responses are sex-dependent in the skeletal muscle and liver in offspring of obese mice. Am J Physiol Endocrinol Metab 2022; 323:E336-E353. [PMID: 35858246 PMCID: PMC9529275 DOI: 10.1152/ajpendo.00263.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 11/22/2022]
Abstract
Infants born to obese mothers are more likely to develop metabolic disease, including glucose intolerance and hepatic steatosis, in adult life. We examined the effects of maternal obesity on the transcriptome of skeletal muscle and liver tissues of the near-term fetus and 3-mo-old offspring in mice born to dams fed a high-fat and -sugar diet. Previously, we have shown that male, but not female, offspring develop glucose intolerance, insulin resistance, and liver steatosis at 3 mo old. Female C57BL6/J mice were fed normal chow or an obesogenic high-calorie diet before mating and throughout pregnancy. RNAseq was performed on the liver and gastrocnemius muscle following collection from fetuses on embryonic day 18.5 (E18.5) as well as from 3-mo-old offspring from obese dams and control dams. Significant genes were generated for each sex, queried for enrichment, and modeled to canonical pathways. RNAseq was corroborated by protein quantification in offspring. The transcriptomic response to maternal obesity in the liver was more marked in males than females. However, in both male and female offspring of obese dams, we found significant enrichment for fatty acid metabolism, mitochondrial transport, and oxidative stress in the liver transcriptomes as well as decreased protein concentrations of electron transport chain members. In skeletal muscle, pathway analysis of gene expression revealed sexual dimorphic patterns, including metabolic processes of fatty acids and glucose, as well as PPAR, AMPK, and PI3K-Akt signaling pathways. Transcriptomic responses to maternal obesity in skeletal muscle were more marked in female offspring than males. Female offspring had greater expression of genes associated with glucose uptake, and protein abundance reflected greater activation of mTOR signaling. Skeletal muscle and livers in mice born to obese dams had sexually dimorphic transcriptomic responses that changed from the fetus to the adult offspring. These data provide insights into mechanisms underpinning metabolic programming in maternal obesity.NEW & NOTEWORTHY Transcriptomic data support that fetuses of obese mothers modulate metabolism in both muscle and liver. These changes were strikingly sexually dimorphic in agreement with published findings that male offspring of obese dams exhibit pronounced metabolic disease earlier. In both males and females, the transcriptomic responses in the fetus were different than those at 3 mo, implicating adaptive mechanisms throughout adulthood.
Collapse
Affiliation(s)
- Amy C Kelly
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Fredrick J Rosario
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jeannie Chan
- Section of Molecular Medicine, Department of Internal Medicine, Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Laura A Cox
- Section of Molecular Medicine, Department of Internal Medicine, Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Theresa L Powell
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
9
|
Seneviratne SN, Rajindrajith S. Fetal programming of obesity and type 2 diabetes. World J Diabetes 2022; 13:482-497. [PMID: 36051425 PMCID: PMC9329845 DOI: 10.4239/wjd.v13.i7.482] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/18/2021] [Accepted: 06/03/2022] [Indexed: 02/06/2023] Open
Abstract
The prevalence of obesity and type 2 diabetes mellitus has increased rapidly over the past few decades, and prevention efforts have not been successful. Fetal programming involves the earliest stage of obesity development, and provides a novel concept to complement other strategies for lifelong prevention of obesity and type 2 diabetes mellitus. The World Health Organization now advocates a life-course approach to prevent/control obesity, starting with pre-conceptional and antenatal maternal health. Maternal overnutrition, gestational diabetes mellitus and excessive gestational weight gain lead to fetal overgrowth, and “programs” the offspring with an increased risk of obesity and type 2 diabetes mellitus in childhood and adulthood. This review summarizes current data on fetal programming of obesity and type 2 diabetes mellitus including potential causative factors, mechanisms and interventions to reduce its impact.
Collapse
Affiliation(s)
| | - Shaman Rajindrajith
- Department of Paediatrics, Faculty of Medicine, University of Colombo, Colombo 08, Sri Lanka
| |
Collapse
|
10
|
Langley-Evans SC. Early life programming of health and disease: the long-term consequences of obesity in pregnancy: a narrative review. J Hum Nutr Diet 2022; 35:816-832. [PMID: 35475555 PMCID: PMC9540012 DOI: 10.1111/jhn.13023] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/06/2022] [Indexed: 11/28/2022]
Abstract
The prevalence of overweight and obesity is rising in all parts of the world and among young women it presents a very clear danger during pregnancy. Women who are overweight or who gain excessive weight during pregnancy are at greater risk of complications in pregnancy and labour, and are more likely to lose their child to stillbirth, or themselves die during pregnancy. This narrative review considers the evidence that in addition to increasing risk of poor pregnancy outcomes, obesity has the capacity to programme fetuses to be at greater risk of cardiometabolic disorders later in life. An extensive body of evidence from prospective and retrospective cohorts, and record linkage studies demonstrates associations of maternal obesity and/or gestational diabetes with cardiovascular disease, type-1 and type-2 diabetes. Studies in animals suggest that these associations are underpinned by adaptations that occur in fetal life, which remodel the structures of major organs including the brain, kidney and pancreas. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Simon C Langley-Evans
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD
| |
Collapse
|
11
|
Sanches APV, de Oliveira JL, Ferreira MS, Lima BDS, Miyamoto JÉ, Simino LADP, Torsoni MA, Torsoni AS, Milanski M, Ignácio-Souza LM. Obesity phenotype induced by high-fat diet leads to maternal-fetal constraint, placental inefficiency, and fetal growth restriction in mice. J Nutr Biochem 2022; 104:108977. [PMID: 35248701 DOI: 10.1016/j.jnutbio.2022.108977] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 11/20/2021] [Accepted: 02/09/2022] [Indexed: 11/28/2022]
Abstract
The aim of this study was to investigate certain parameters regarding the maternal-fetal outcomes in a diet-induced obesity model. Obese, glucose-intolerant females who were exposed to a high-fat diet prior to pregnancy had lower placental efficiency and lower birth weight pups compared to the controls. Simple linear regression analyses showed that maternal obesity disrupts the proportionality between maternal and fetal outcomes during pregnancy. Maternal obesity is correlated with fetal outcomes, perhaps because of problems with hormonal signaling and exacerbation of inflammation in the maternal metabolic environment. The maternal obese phenotype altered the thickness of the placental layer, the transport of fatty acids, and the expression of growth factors. For example, lower expression of epidermal growth factor receptor (EGFR) mRNA in the obesity-prone group may have contributed to the rupture of the placental layers, leading to adverse fetal outcomes. Furthermore, maintenance of maternal glucose homeostasis and overexpression of placental growth factor (PGF) in the obesity-resistant group likely protected the placenta and fetuses from morphological and functional damage.
Collapse
Affiliation(s)
- Ana Paula Varela Sanches
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Josilene Lopes de Oliveira
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Maíra Schuchter Ferreira
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Bruna de Souza Lima
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Josiane Érica Miyamoto
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Laís Angélica de Paula Simino
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Márcio Alberto Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Adriana Souza Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Marciane Milanski
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Letícia Martins Ignácio-Souza
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil.
| |
Collapse
|
12
|
Gätjens I, Fedde S, Schmidt SCE, Hasler M, Plachta-Danielzik S, Müller MJ, Bosy-Westphal A. Relationship between Birth Weight, Early Growth Rate, and Body Composition in 5- to 7-Year-Old Children. Obes Facts 2022; 15:519-527. [PMID: 35292608 PMCID: PMC9421709 DOI: 10.1159/000522509] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 02/03/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Programing of body composition during intrauterine growth may contribute to the higher risk for cardio-metabolic disease in individuals born small or large for gestational age (SGA, LGA). Compensations of intrauterine growth by catch-up or catch-down postnatal growth may lead to adverse consequences like a thin-fat phenotype. METHODS The impact of (i) birth weight as well as (ii) the interaction between birth weight and catch-up or catch-down growth during the first 2 years of life on fat-free mass index (FFMI) and fat mass index (FMI) in 3,204 5-7-year-old children were investigated using Hattori's body composition chart. Body composition results were compared to appropriate for gestational age (AGA) birth weight with the same body mass index (BMI). RESULTS In total, 299 children at age 5-7 years were categorized as SGA, 2,583 as AGA, and 322 as LGA. When compared to AGA-children, BMI at 5-7 years of age was higher in LGA-children (15.5 vs. 16.2 kg/m2; p < 0.001) but not different in SGA-children. Compared to AGA with the same BMI, LGA was associated with higher FMI and a lower FFMI in 5-7-year-old girls. This phenotype was also seen for both sexes with catch-down growth during the first 2 years of life whereas catch-up growth prevented the higher FMI and lower FFMI per BMI. By contrast, SGA was associated with a higher FFMI and lower FMI in 5-7-year-old boys compared to AGA boys with the same BMI. This phenotype was also seen with catch-down growth in both genders whereas catch-up growth in girls led to more gain in FMI per BMI. CONCLUSION LGA with a compensatory catch-down postnatal growth may be a risk factor for the development of disproportionate gain in fat over lean mass whereas SGA with a catch-down postnatal growth seems to favor the subsequent accretion of lean over fat mass. A higher propensity of lean mass accretion during postnatal growth in boys compared to girls explains sex differences in these phenotypes.
Collapse
Affiliation(s)
- Isabel Gätjens
- Institute of Human Nutrition and Food Science, Christian-Albrechts University Kiel, Kiel, Germany
| | - Svenja Fedde
- Institute of Human Nutrition and Food Science, Christian-Albrechts University Kiel, Kiel, Germany
| | | | - Mario Hasler
- Applied Statistics, Agricultural and Food Economics Faculty, Christian-Albrechts University, Kiel, Germany
| | - Sandra Plachta-Danielzik
- Institute of Human Nutrition and Food Science, Christian-Albrechts University Kiel, Kiel, Germany
- Kompetenznetz Darmerkrankungen e.V., Kiel, Germany
| | - Manfred James Müller
- Institute of Human Nutrition and Food Science, Christian-Albrechts University Kiel, Kiel, Germany
| | - Anja Bosy-Westphal
- Institute of Human Nutrition and Food Science, Christian-Albrechts University Kiel, Kiel, Germany
- *Anja Bosy-Westphal,
| |
Collapse
|
13
|
Al Ghali R, Smail L, Muqbel M, Haroun D. Maternal investment, life-history trajectory of the off-spring and cardiovascular disease risk in Emirati females in the United Arab Emirates. BMC Public Health 2021; 21:1237. [PMID: 34176485 PMCID: PMC8237435 DOI: 10.1186/s12889-021-11182-0] [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: 10/15/2020] [Accepted: 06/02/2021] [Indexed: 01/02/2023] Open
Abstract
Background Variations in cardiovascular disease risk (CVD) are suggested to be partly influenced by factors that affect prenatal growth patterns and outcomes, namely degree of maternal investment (proxied by birth weight and gestational age). Using the life history trajectory model, this study investigates whether maternal investment in early prenatal life associates with menarcheal age and whether maternal investment affects CVD risk in adulthood and predicts adult size and adiposity levels. Methods A cross-sectional study was conducted among 94 healthy Emirati females. Birth weight, gestational age and menarcheal age were obtained. Anthropometrical measurements, body composition analysis, and blood pressure values were collected. Regression analyses were conducted to establish associations. Results There was no association between birth weight standard deviation score (SDS) and age at menarche. When investigating the associations of birth weight SDS and age at menarche with growth indices, it was found that only birth weight was positively and significantly associated with both height (β = 1.342 cm, 95% CI (0.12, 2.57), p = 0.032) and leg length (β = 0.968 cm, 95% CI (0.08, 1.86), p = 0.034). Menarcheal age was significantly and inversely associated with fat mass index (FMI) (β = − 0.080 cm, 95% CI (− 0.13, − 0.03), p = 0.002), but not with waist circumference and fat free mass index (FFMI) (p > 0.05). Birth weight SDS was positively and significantly associated with waist circumference (β = 0.035 cm, 95% CI (0.01, 0.06), p = 0.009), FMI (β = 0.087 cm, 95% CI (0.01, 0.16), p = 0.027), and FFMI (β = 0.485 cm, 95% CI (0.17, 0.80), p = 0.003). Birth weight SDS was not significantly associated with either systolic blood pressure (SBP) or diastolic blood pressure (DBP) (p > 0.05). However, FMI, waist circumference, and FFMI were positively and significantly associated with SBP. Regarding DBP, the relationship was negatively and significantly associated with only FFMI (β = − 1.6111 kg/m2, 95% CI (− 2.63, − 0.60), p = 0.002). Conclusion Although the results do not fully support that Emirati females fast-life history is associated with increased chronic disease risk, the data does suggest a link between restricted fetal growth in response to low maternal investment and metabolic and reproductive health. Supplementary Information The online version contains supplementary material available at 10.1186/s12889-021-11182-0.
Collapse
Affiliation(s)
- Rola Al Ghali
- Department of Health Sciences, Zayed University, College of Natural and Health Sciences, Dubai, United Arab Emirates
| | - Linda Smail
- Department of Mathematics and Statistics, Zayed University, College of Natural and Health Sciences, Dubai, United Arab Emirates
| | - Maryam Muqbel
- Department of Medicine, Imperial College, London, UK
| | - Dalia Haroun
- Department of Health Sciences, Zayed University, College of Natural and Health Sciences, Dubai, United Arab Emirates.
| |
Collapse
|
14
|
Kuiper-Makris C, Selle J, Nüsken E, Dötsch J, Alejandre Alcazar MA. Perinatal Nutritional and Metabolic Pathways: Early Origins of Chronic Lung Diseases. Front Med (Lausanne) 2021; 8:667315. [PMID: 34211985 PMCID: PMC8239134 DOI: 10.3389/fmed.2021.667315] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Lung development is not completed at birth, but expands beyond infancy, rendering the lung highly susceptible to injury. Exposure to various influences during a critical window of organ growth can interfere with the finely-tuned process of development and induce pathological processes with aberrant alveolarization and long-term structural and functional sequelae. This concept of developmental origins of chronic disease has been coined as perinatal programming. Some adverse perinatal factors, including prematurity along with respiratory support, are well-recognized to induce bronchopulmonary dysplasia (BPD), a neonatal chronic lung disease that is characterized by arrest of alveolar and microvascular formation as well as lung matrix remodeling. While the pathogenesis of various experimental models focus on oxygen toxicity, mechanical ventilation and inflammation, the role of nutrition before and after birth remain poorly investigated. There is accumulating clinical and experimental evidence that intrauterine growth restriction (IUGR) as a consequence of limited nutritive supply due to placental insufficiency or maternal malnutrition is a major risk factor for BPD and impaired lung function later in life. In contrast, a surplus of nutrition with perinatal maternal obesity, accelerated postnatal weight gain and early childhood obesity is associated with wheezing and adverse clinical course of chronic lung diseases, such as asthma. While the link between perinatal nutrition and lung health has been described, the underlying mechanisms remain poorly understood. There are initial data showing that inflammatory and nutrient sensing processes are involved in programming of alveolarization, pulmonary angiogenesis, and composition of extracellular matrix. Here, we provide a comprehensive overview of the current knowledge regarding the impact of perinatal metabolism and nutrition on the lung and beyond the cardiopulmonary system as well as possible mechanisms determining the individual susceptibility to CLD early in life. We aim to emphasize the importance of unraveling the mechanisms of perinatal metabolic programming to develop novel preventive and therapeutic avenues.
Collapse
Affiliation(s)
- Celien Kuiper-Makris
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jaco Selle
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Eva Nüsken
- Department of Pediatric and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatric and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Miguel A. Alejandre Alcazar
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Excellence Cluster on Stress Responses in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Member of the German Centre for Lung Research (DZL), Institute for Lung Health, University of Giessen and Marburg Lung Centre (UGMLC), Gießen, Germany
| |
Collapse
|
15
|
Desai M, Ross MG. Maternal-infant nutrition and development programming of offspring appetite and obesity. Nutr Rev 2021; 78:25-31. [PMID: 33196091 PMCID: PMC7667467 DOI: 10.1093/nutrit/nuaa121] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the United States and Mexico, the obesity epidemic represents a significant public health problem. Although obesity is often attributed to a Western-style, high-fat diet and decreased activity, there is now compelling evidence that this, in part, occurs because of the developmental programming effects resulting from exposure to maternal overnutrition. Human and animal studies demonstrate that maternal obesity and high-fat diet result in an increased risk for childhood and adult obesity. The potential programming effects of obesity have been partly attributed to hyperphagia, which occurs as a result of increased appetite with reduced satiety neuropeptides or neurons. However, depending on maternal nutritional status during the nursing period, the programmed hyperphagia and obesity can be exacerbated or prevented in offspring born to obese mothers. The underlying mechanism of this phenomenon likely involves the plasticity of the appetite regulatory center and thus presents an opportunity to modulate feeding and satiety regulation and break the obesity cycle.
Collapse
Affiliation(s)
- Mina Desai
- Department of Obstetrics and Gynecology, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA; and David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, USA
| | - Michael G Ross
- Department of Obstetrics and Gynecology, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA; and David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
16
|
Andriani H. Birth weight and childhood obesity: effect modification by residence and household wealth. Emerg Themes Epidemiol 2021; 18:6. [PMID: 33975611 PMCID: PMC8111737 DOI: 10.1186/s12982-021-00096-2] [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: 07/06/2019] [Accepted: 04/27/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There are both genetic and environmental factors which contribute to a child's chances of being obese. When low birth weight (LBW) has been specifically evaluated relative to its association with childhood obesity, the results have produced conflicting findings. This study aims to describe the relationship between birth weight and childhood obesity and investigate the influence that residence and household wealth has on this relationship. METHODS I performed a secondary analysis on the 2013 Riskesdas (or Basic Health Research), a cross-sectional, nationally representative survey of the Indonesian population. Height, weight, information regarding child's birth weight, and basic characteristics of the study population were collected from parents with children aged 0 to 5 years (n = 63,237) in 2013. The exposure was child's birth weight and the outcomes were child's current weight, BMI z-score, and obesity. Data were analyzed by using multiple linear regression and multiple logistic regression. RESULTS I found a significant increase in the weight, BMI z-score, and risk of childhood obesity to be associated with LBW. LBW children in rural area were associated with higher BMI z-score (mean ± standard error: 1.44 ± 0.02) and higher odds (odds ratio (95% confidence interval): 7.46 (6.77-8.23)) of obesity than those in urban area. LBW children from low class families were associated with higher BMI z-score (1.79 ± 0.04) and had higher odds (14.79 (12.47-17.54)) of obesity than those from middle class and wealthy families. CONCLUSIONS Effective prevention and intervention to childhood obesity as early as possible are imperative. As far as this study was concerned, efforts, policies, and targets are required to reduce the prevalence of LBW. Children born of LBW, who live in a rural area and from low income families, should be emphatically intervened as early as possible.
Collapse
Affiliation(s)
- Helen Andriani
- Department of Health Policy and Administration, Faculty of Public Health, Universitas Indonesia, Lingkar Kampus Raya Universitas Indonesia Street, Depok, 16424, Indonesia.
| |
Collapse
|
17
|
Differences and Correlation Analysis of Birth Weight and Overweight/Obesity in Shanghai Twin Cohort. Twin Res Hum Genet 2021; 24:29-36. [PMID: 33645497 DOI: 10.1017/thg.2021.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The objective of this study was to analyze differences in birth weight and overweight/obesity in a Shanghai twin cohort. We also wanted to study their association and explore possible risk factors for the discordance of overweight/obesity within twins. This was an internal case-control study designed for twins. The 2012 Shanghai Twin Registration System baseline survey data of a total of 3417 twin pairs were statistically analyzed using SPSS22 software. Results show that the body mass index (BMI) of the Shanghai twin population increased with age. Twins with a high birth weight had a higher BMI and a higher rate of overweight and obesity; 0- to 6-year-old twins, male twins and dizygotic (DZ) twins had higher rates of overweight/obesity than other groups. The greater the discordant birth weight rate of twins, the more obvious the difference in BMI (p < .05). There was a significant difference in overweight/obesity between twins with a relative difference of birth weight ≥15% in DZ twins (p < .05). DZ twins, male twins and 0- to 6-year-old twins were more likely to be discordant in overweight/obese than others. The discordant birth weight within twins was not a risk factor for discordant overweight/obesity. However, attention should be paid to childhood obesity, and appropriate interventions should be made at the appropriate time. Genetics may play an important role in the occurrence and development of overweight/obesity. In conclusion, discordant growth and development in the uterus early in life may not lead to discordant weight development in the future.
Collapse
|
18
|
Ni Y, Beckmann J, Hurst JR, Morris JK, Marlow N. Size at birth, growth trajectory in early life, and cardiovascular and metabolic risks in early adulthood: EPICure study. Arch Dis Child Fetal Neonatal Ed 2021; 106:149-155. [PMID: 32796060 PMCID: PMC7116791 DOI: 10.1136/archdischild-2020-319328] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To investigate whether size at birth and growth trajectories in infancy and childhood are associated with determinants of cardiovascular and metabolic risks in young adults born extremely preterm (EP, <26 weeks of gestation). METHODS We used longitudinal data from the EPICure study of 129 EP survivors up to 19 years in the UK and Ireland in 1995. Determinants of cardiovascular and metabolic risks at 19 years included the presence of metabolic syndrome, body mass index (BMI) and systolic blood pressure (SBP). Predictors were birth weight for gestation and gain in weight z-scores in the following periods: birth-postmenstrual age of 40 weeks (term), infancy (term-2.5 years), early childhood (2.5-6.0 years) and late childhood (6-11 years). RESULTS Metabolic syndrome was present in 8.7% of EP participants at 19 years. Compared with subjects without metabolic syndrome, those with metabolic syndrome tended to have a smaller size at birth (difference in means: -0.55 SD, 95% CI -1.10 to 0.01, p=0.053) and a greater increase in weight z-scores from term to 2.5 years (difference in means: 1.00 SD, 95% CI -0.17 to 2.17, p=0.094). BMI at 19 years was positively related to growth from 2.5 to 6.0 years ( β : 1.03, 95% CI 0.31 to 1.75, p=0.006); an inverse association with birthweight z-scores was found in the lower socioeconomic status group ( β : -1.79, 95% CI -3.41 to -0.17, p=0.031). Central SBP was positively related to growth from 2.5 to 6.0 years ( β : 1.75, 95% CI 0.48 to 3.02, p=0.007). CONCLUSION Size at EP birth and increased catch-up in weight from 2.5 to 6.0 years were associated with BMI and central SBP in early adulthood.
Collapse
Affiliation(s)
- Yanyan Ni
- UCL EGA Institute for Women's Health, University College London, London, UK
| | - Joanne Beckmann
- Institute for Women’s Health, University College London, London, UK
| | - John R Hurst
- UCL Respiratory, University College London, London, UK
| | - Joan K Morris
- Population Health Research Institute, St George’s, University of London, UK
| | - Neil Marlow
- Institute for Women’s Health, University College London, London, UK
| |
Collapse
|
19
|
Jasper EA, Cho H, Breheny PJ, Bao W, Dagle JM, Ryckman KK. Perinatal determinants of growth trajectories in children born preterm. PLoS One 2021; 16:e0245387. [PMID: 33507964 PMCID: PMC7842887 DOI: 10.1371/journal.pone.0245387] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/29/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND A growing amount of evidence indicates in utero and early life growth has profound, long-term consequences for an individual's health throughout the life course; however, there is limited data in preterm infants, a vulnerable population at risk for growth abnormalities. OBJECTIVE To address the gap in knowledge concerning early growth and its determinants in preterm infants. METHODS A retrospective cohort study was performed using a population of preterm (< 37 weeks gestation) infants obtained from an electronic medical record database. Weight z-scores were acquired from discharge until roughly two years corrected age. Linear mixed effects modeling, with random slopes and intercepts, was employed to estimate growth trajectories. RESULTS Thirteen variables, including maternal race, hypertension during pregnancy, preeclampsia, first trimester body mass index, multiple status, gestational age, birth weight, birth length, head circumference, year of birth, length of birth hospitalization stay, total parenteral nutrition, and dextrose treatment, were significantly associated with growth rates of preterm infants in univariate analyses. A small percentage (1.32% - 2.07%) of the variation in the growth of preterm infants can be explained in a joint model of these perinatal factors. In extremely preterm infants, additional variation in growth trajectories can be explained by conditions whose risk differs by degree of prematurity. Specifically, infants with periventricular leukomalacia or retinopathy of prematurity experienced decelerated rates of growth compared to infants without such conditions. CONCLUSIONS Factors found to influence growth over time in children born at term also affect growth of preterm infants. The strength of association and the magnitude of the effect varied by gestational age, revealing that significant heterogeneity in growth and its determinants exists within the preterm population.
Collapse
Affiliation(s)
- Elizabeth A. Jasper
- Department of Epidemiology, University of Iowa, Iowa City, IA, United States of America
- * E-mail:
| | - Hyunkeun Cho
- Department of Biostatistics, University of Iowa, Iowa City, IA, United States of America
| | - Patrick J. Breheny
- Department of Biostatistics, University of Iowa, Iowa City, IA, United States of America
| | - Wei Bao
- Department of Epidemiology, University of Iowa, Iowa City, IA, United States of America
| | - John M. Dagle
- Department of Pediatrics, University of Iowa, Iowa City, IA, United States of America
| | - Kelli K. Ryckman
- Department of Epidemiology, University of Iowa, Iowa City, IA, United States of America
| |
Collapse
|
20
|
Mangwiro YT, Cuffe JS, Vickers MH, Reynolds CM, Mahizir D, Anevska K, Gravina S, Romano T, Moritz KM, Briffa JF, Wlodek ME. Maternal exercise alters rat fetoplacental stress response: Minimal effects of maternal growth restriction and high-fat feeding. Placenta 2020; 104:57-70. [PMID: 33276236 DOI: 10.1016/j.placenta.2020.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 11/09/2020] [Accepted: 11/15/2020] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Fetal growth restriction complicates 10% of pregnancies and increases offspring (F1) risk of metabolic disorders, including obesity and gestational diabetes mellitus (GDM). This disease predisposition can be passed onto the next generation (F2). Importantly, the risk of pregnancy complications in obese women can be exacerbated by a stressful pregnancy. Exercise can reduce adiposity and improve health outcomes in obese women and those with GDM. This study investigated the impacts of maternal growth restriction, obesity, exercise, and stress on fetal and placental endocrine function. METHODS Uteroplacental insufficiency (Restricted) or sham (Control) surgery was induced on embryonic day (E) 18 in F0 Wistar-Kyoto rats. F1 offspring were fed a Chow or High-fat (HFD) diet from weaning and, at 16 weeks, were randomly allocated an exercise protocol; Sedentary, Exercised prior to and during pregnancy (Exercise), or Exercised only during pregnancy (PregEx). Females were mated and further randomly allocated to either undergo (Stress), or not undergo (Unstressed), physiological measurements during pregnancy. On E20, F2 fetal plasma (steroid hormones), tissues (brain, liver), and placentae (morphology, stress genes) were collected. RESULTS Maternal growth restriction and high-fat feeding had minimal impact on fetoplacental endocrine function. PregEx and Exercise increased cross-sectional labyrinth and junctional zone areas. PregEx, but not Exercise, increased fetal deoxycorticosterone concentrations and reduced placental Hsd11b2 and Nr3c2 gene abundance. Maternal stress increased fetal corticosterone concentrations in Sedentary HFD dams and increased placental cross-sectional areas in PregEx mothers. DISCUSSION PregEx and Stress independently dysregulates the endocrine status of the developing fetus, which may program future disease.
Collapse
Affiliation(s)
- Yeukai Tm Mangwiro
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, 3083, Australia; Department of Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - James Sm Cuffe
- School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Mark H Vickers
- Liggins Institute, University of Auckland, Grafton, Auckland, 1142, New Zealand
| | - Clare M Reynolds
- Liggins Institute, University of Auckland, Grafton, Auckland, 1142, New Zealand
| | - Dayana Mahizir
- Department of Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Kristina Anevska
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, 3083, Australia; Department of Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Sogand Gravina
- Department of Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Tania Romano
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, 3083, Australia
| | - Karen M Moritz
- School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD, 4072, Australia; Child Health Research Centre, The University of Queensland, South Brisbane, QLD, 4101, Australia
| | - Jessica F Briffa
- Department of Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Mary E Wlodek
- Department of Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
21
|
Ni Y, Beckmann J, Gandhi R, Hurst JR, Morris JK, Marlow N. Growth to early adulthood following extremely preterm birth: the EPICure study. Arch Dis Child Fetal Neonatal Ed 2020; 105:496-503. [PMID: 31907276 PMCID: PMC7115967 DOI: 10.1136/archdischild-2019-318192] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/21/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To investigate growth trajectories from age 2.5 to 19 years in individuals born before 26 weeks of gestation (extremely preterm; EP) compared with term-born controls. METHODS Multilevel modelling of growth data from the EPICure study, a prospective 1995 birth cohort of 315 EP participants born in the UK and Ireland and 160 term-born controls recruited at school age. Height, weight, head circumference and body mass index (BMI) z-scores were derived from UK standards at ages 2.5, 6, 11 and 19 years. RESULTS 129 (42%) EP children were assessed at 19 years. EP individuals were on average 4.0 cm shorter and 6.8 kg lighter with a 1.5 cm smaller head circumference relative to controls at 19 years. Relative to controls, EP participants grew faster in weight by 0.06 SD per year (95% CI 0.05 to 0.07), in head circumference by 0.04 SD (95% CI 0.03 to 0.05), but with no catch-up in height. For the EP group, because of weight catch-up between 6 and 19 years, BMI was significantly elevated at 19 years to +0.32 SD; 23.4% had BMI >25 kg/m2 and 6.3% >30 kg/m2 but these proportions were similar to those in control subjects. EP and control participants showed similar pubertal development in early adolescence, which was not associated with height at 19 years in either study group. Growth through childhood was related to birth characteristics and to neonatal feeding practices. CONCLUSIONS EP participants remained shorter and lighter and had smaller head circumferences than reference data or controls in adulthood but had elevated BMI.
Collapse
Affiliation(s)
- Yanyan Ni
- Institute for Women's Health, University College London, London, UK
| | - Joanne Beckmann
- Institute for Women's Health, University College London, London, UK
| | | | - John R Hurst
- UCL Respiratory, University College London, London, UK
| | - Joan K Morris
- Population Health Research Institute, St George's University of London, London, UK
| | - Neil Marlow
- Institute for Women's Health, University College London, London, UK
| |
Collapse
|
22
|
Placental function in maternal obesity. Clin Sci (Lond) 2020; 134:961-984. [PMID: 32313958 DOI: 10.1042/cs20190266] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023]
Abstract
Maternal obesity is associated with pregnancy complications and increases the risk for the infant to develop obesity, diabetes and cardiovascular disease later in life. However, the mechanisms linking the maternal obesogenic environment to adverse short- and long-term outcomes remain poorly understood. As compared with pregnant women with normal BMI, women entering pregnancy obese have more pronounced insulin resistance, higher circulating plasma insulin, leptin, IGF-1, lipids and possibly proinflammatory cytokines and lower plasma adiponectin. Importantly, the changes in maternal levels of nutrients, growth factors and hormones in maternal obesity modulate placental function. For example, high insulin, leptin, IGF-1 and low adiponectin in obese pregnant women activate mTOR signaling in the placenta, promoting protein synthesis, mitochondrial function and nutrient transport. These changes are believed to increase fetal nutrient supply and contribute to fetal overgrowth and/or adiposity in offspring, which increases the risk to develop disease later in life. However, the majority of obese women give birth to normal weight infants and these pregnancies are also associated with activation of inflammatory signaling pathways, oxidative stress, decreased oxidative phosphorylation and lipid accumulation in the placenta. Recent bioinformatics approaches have expanded our understanding of how maternal obesity affects the placenta; however, the link between changes in placental function and adverse outcomes in obese women giving birth to normal sized infants is unclear. Interventions that specifically target placental function, such as activation of placental adiponectin receptors, may prevent the transmission of metabolic disease from obese women to the next generation.
Collapse
|
23
|
Eitmann S, Németh D, Hegyi P, Szakács Z, Garami A, Balaskó M, Solymár M, Erőss B, Kovács E, Pétervári E. Maternal overnutrition impairs offspring's insulin sensitivity: A systematic review and meta-analysis. MATERNAL AND CHILD NUTRITION 2020; 16:e13031. [PMID: 32567808 PMCID: PMC7503101 DOI: 10.1111/mcn.13031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 04/17/2020] [Accepted: 04/27/2020] [Indexed: 12/31/2022]
Abstract
This systematic review and meta‐analysis aimed to investigate the association between maternal overnutrition and offspring's insulin sensitivity—following the Preferred Reporting Items for Systematic Reviews and Meta‐analyses statement. Studies published in English before April 22, 2019, were identified through searches of four medical databases. After selection, 15 studies aiming to explore the association between prepregnancy body mass index (ppBMI) or gestational weight gain (GWG) of non‐diabetic mothers and their offspring's insulin sensitivity (fasting insulin or glucose level and Homeostatic Measurement Assessment for Insulin Resistance [HOMA‐IR]) were included in the meta‐analysis. Associations of ppBMI and GWG with offspring's insulin sensitivity were analysed by pooling regression coefficients or standardized differences in means with 95% confidence intervals (CIs). Maternal ppBMI showed significant positive correlations with the level of both fasting insulin and HOMA‐IR in offspring (standardized regression coefficient for fasting insulin: 0.107, CI [0.053, 0.160], p < 0.001 and that for HOMA‐IR: 0.063, CI [0.006, 0.121], p = 0.031). However, the result of the analysis on coefficients adjusted for offspring's actual anthropometry (BMI and adiposity) was not significant. Independent from ppBMI, GWG tended to show a positive correlation with insulin level, but not after adjustment for offspring's anthropometry. Offspring of mothers with excessive GWG showed significantly higher HOMA‐IR than those of mothers with optimal GWG (p = 0.004). Our results demonstrate that both higher ppBMI and GWG increase the risk of offspring's insulin resistance, but the effect of ppBMI on insulin sensitivity in offspring may develop as consequence of their adiposity.
Collapse
Affiliation(s)
- Szimonetta Eitmann
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Dávid Németh
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Péter Hegyi
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Zsolt Szakács
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary.,János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - András Garami
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Márta Balaskó
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Margit Solymár
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Bálint Erőss
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary.,Division of Gastroenterology, First Department of Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Enikő Kovács
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Erika Pétervári
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
24
|
Lima RS, Assis Silva Gomes J, Moreira PR. An overview about DNA methylation in childhood obesity: Characteristics of the studies and main findings. J Cell Biochem 2020; 121:3042-3057. [DOI: 10.1002/jcb.29544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 10/10/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Rafael Silva Lima
- Laboratory of Cell‐Cell Interactions, Department of Morphology, Institute of Biological SciencesFederal University of Minas Gerais Minas Gerais Brazil
| | - Juliana Assis Silva Gomes
- Laboratory of Cell‐Cell Interactions, Department of Morphology, Institute of Biological SciencesFederal University of Minas Gerais Minas Gerais Brazil
| | - Paula Rocha Moreira
- Laboratory of Cell‐Cell Interactions, Department of Morphology, Institute of Biological SciencesFederal University of Minas Gerais Minas Gerais Brazil
| |
Collapse
|
25
|
Trasande L, Ghassabian A, Kahn LG, Jacobson MH, Afanasyeva Y, Liu M, Chen Y, Naidu M, Alcedo G, Gilbert J, Koshy TT. The NYU Children's Health and Environment Study. Eur J Epidemiol 2020; 35:305-320. [PMID: 32212050 PMCID: PMC7154015 DOI: 10.1007/s10654-020-00623-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 03/11/2020] [Indexed: 10/31/2022]
Abstract
The aims of the NYU Children's Health and Environment Study (CHES) are to evaluate influences of prenatal non-persistent chemical exposures on fetal and postnatal growth and pool our data with the US National Institutes of Health Environmental influences on Child Health Outcomes (ECHO) Program to answer collaborative research questions on the impact of the preconceptual, prenatal, and postnatal environment on childhood obesity, neurodevelopment, pre/peri/postnatal outcomes, upper and lower airway outcomes, and positive health. Eligible women were ≥ 18 years old, < 18 weeks pregnant, had a pregnancy that is not medically threatened, and planned to deliver at NYU Langone Hospital-Manhattan, Bellevue Hospital, or NYU Langone Hospital-Brooklyn. Between March 22, 2016 and April 15, 2019, we recruited 2469 pregnant women, from whom 2193 completed an initial questionnaire and continued into NYU CHES. Of the 2193, 88 miscarried, 28 terminated, and 20 experienced stillbirth, while 57 were lost to follow up. We report here demographic and other characteristics of the 2000 live deliveries (2037 children), from whom 1624 (80%) consented to postnatal follow-up. Data collection in pregnancy was nested in clinical care, with questionnaire and specimen collection conducted during routine prenatal visits at < 18, 18-25, and > 25 weeks gestation. These have been followed by questionnaire and specimen collection at birth and regular postpartum intervals.
Collapse
Affiliation(s)
- Leonardo Trasande
- Department of Pediatrics, New York University School of Medicine, New York, NY, USA. .,Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA. .,Department of Population Health, New York University School of Medicine, New York, NY, USA. .,NYU Wagner School of Public Service, New York, NY, USA. .,NYU College of Global Public Health, New York, NY, USA.
| | - Akhgar Ghassabian
- Department of Pediatrics, New York University School of Medicine, New York, NY, USA.,Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA.,Department of Population Health, New York University School of Medicine, New York, NY, USA
| | - Linda G Kahn
- Department of Pediatrics, New York University School of Medicine, New York, NY, USA
| | - Melanie H Jacobson
- Department of Pediatrics, New York University School of Medicine, New York, NY, USA
| | - Yelena Afanasyeva
- Department of Pediatrics, New York University School of Medicine, New York, NY, USA.,Department of Population Health, New York University School of Medicine, New York, NY, USA
| | - Mengling Liu
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA.,Department of Population Health, New York University School of Medicine, New York, NY, USA
| | - Yu Chen
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA.,Department of Population Health, New York University School of Medicine, New York, NY, USA
| | - Mrudula Naidu
- Department of Pediatrics, New York University School of Medicine, New York, NY, USA.,Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
| | - Garry Alcedo
- Department of Pediatrics, New York University School of Medicine, New York, NY, USA
| | - Joseph Gilbert
- Department of Pediatrics, New York University School of Medicine, New York, NY, USA
| | - Tony T Koshy
- Department of Pediatrics, New York University School of Medicine, New York, NY, USA
| | | |
Collapse
|
26
|
A heretical view: rather than a solely placental protective function, placental 11β hydroxysteroid dehydrogenase 2 also provides substrate for fetal peripheral cortisol synthesis in obese pregnant ewes. J Dev Orig Health Dis 2020; 12:94-100. [PMID: 32151296 DOI: 10.1017/s2040174420000112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Exposure to glucocorticoid levels higher than appropriate for current developmental stages induces offspring metabolic dysfunction. Overfed/obese (OB) ewes and their fetuses display elevated blood cortisol, while fetal Adrenocorticotropic hormone (ACTH) remains unchanged. We hypothesized that OB pregnancies would show increased placental 11β hydroxysteroid dehydrogenase 2 (11β-HSD2) that converts maternal cortisol to fetal cortisone as it crosses the placenta and increased 11β-HSD system components responsible for peripheral tissue cortisol production, providing a mechanism for ACTH-independent increase in circulating fetal cortisol. Control ewes ate 100% National Research Council recommendations (CON) and OB ewes ate 150% CON diet from 60 days before conception until necropsy at day 135 gestation. At necropsy, maternal jugular and umbilical venous blood, fetal liver, perirenal fat, and cotyledonary tissues were harvested. Maternal plasma cortisol and fetal cortisol and cortisone were measured. Fetal liver, perirenal fat, cotyledonary 11β-HSD1, hexose-6-phosphate dehydrogenase (H6PD), and 11β-HSD2 protein abundance were determined by Western blot. Maternal plasma cortisol, fetal plasma cortisol, and cortisone were higher in OB vs. CON (p < 0.01). 11β-HSD2 protein was greater (p < 0.05) in OB cotyledonary tissue than CON. 11β-HSD1 abundance increased (p < 0.05) in OB vs. CON fetal liver and perirenal fat. Fetal H6PD, an 11β-HSD1 cofactor, also increased (p < 0.05) in OB vs. CON perirenal fat and tended to be elevated in OB liver (p < 0.10). Our data provide evidence for increased 11β-HSD system components responsible for peripheral tissue cortisol production in fetal liver and adipose tissue, thereby providing a mechanism for an ACTH-independent increase in circulating fetal cortisol in OB fetuses.
Collapse
|
27
|
Prenatal Ambient Particulate Matter Exposure and Longitudinal Weight Growth Trajectories in Early Childhood. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17041444. [PMID: 32102302 PMCID: PMC7068568 DOI: 10.3390/ijerph17041444] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/12/2022]
Abstract
Air pollution exposure during pregnancy has been associated with impaired fetal growth and postnatal weight gain, but few studies have examined the effect on weight growth trajectories. We examine the association between validated 1 km2 resolution particulate matter (PM2.5) concentrations, averaged over pregnancy, and sex-specific growth trajectories from birth to age six of participants in the Boston-based Children's HealthWatch cohort (4797 participants, 84,283 measures). We compared weight trajectories, predicted using polynomial splines in mixed models, between prenatal PM2.5 above or below the median (9.5 µg/m3), and examined birth weight as an effect modifier. Females exposed to average prenatal PM2.5 ≥ 9.5 µg/m3 had higher weights compared to females exposed to < 9.5 µg/m3 throughout the study period (0.16 kg at 24 months, 0.61 kg at 60 months). In males, higher prenatal PM2.5 exposure was associated with significantly lower weights after 24 months of age, with differences increasing with time (-0.17 at 24 months, -0.72 kg at 60 months). Associations were more pronounced among low birth weight (<2500 g) females, but did not differ by birth weight status in males. Our findings demonstrate the complex association between air pollution exposures and childhood weight trajectories and emphasize the importance of sex-stratified analyses.
Collapse
|
28
|
Javadi M, Rafiei S, Zahedifar F, Barikani A. Relationships between maternal characteristics and infant birth weight. Int J Health Care Qual Assur 2019; 32:688-697. [PMID: 31111782 DOI: 10.1108/ijhcqa-05-2017-0081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE Nowadays, the importance of infant birth weight (IBW) as a key factor in determining the future of physical and mental development of children is a growing concern. The purpose of this paper is to investigate the relationship between maternal characteristics and IBW among pregnant women who were referred to health centers in Qazvin city in the year 2016. DESIGN/METHODOLOGY/APPROACH A descriptive-analytical study was conducted among pregnant women in 28-36 weeks of gestation who referred to healthcare centers and facilities affiliated by the Qazvin University of Medical Sciences in April-June 2016. The associations between maternal physical activity, mothers' socioeconomic status and birth weight were examined by SPSS Software Package version 16 through linear and logistic regression tests. FINDINGS Linear regression modeling suggested that maternal weight (p=0.001), income (p=0.04), gestational age of delivery (p=0.00) and pre-pregnancy BMI (p=0.02) were positively associated with birth weight, while occupational and heavy physical activity (p=0.003 and 0.008, respectively) were negatively associated with IBW. In this study, low birth weight infants are compared to those with normal weight belonged to mothers who have spent more time in doing heavy physical activities (OR=1.11, 95% CI 1.01-1.23). Also infants with low birth weight compared to others in the normal weight category were born from mothers with lower pre-pregnancy BMI (OR=0.65, 95% CI 0.62-0.78), gestational age of delivery (OR=0.82, 95% CI 0.79-0.86), maternal weight (OR=0.86, 95% CI 0.84-0.88) and income (OR=0.79, 95% CI 0.69-0.83). PRACTICAL IMPLICATIONS The study findings revealed that certain maternal characteristics could play a significant role in IBW. Despite the importance, in most of developing countries (particularly Iran), future mothers are not advised about an appropriate weight gain during pregnancy or the optimal level of physical activity in such a period of time. Therefore, counseling pregnant women and giving them proper information on appropriate perinatal care would be helpful in order to have pregnancies with optimal outcomes. ORIGINALITY/VALUE The authors applied several statistical methods to analyze IBW among mothers with different maternal characteristics and predict birth weight based on contributing factors.
Collapse
Affiliation(s)
- Maryam Javadi
- Department of Nutrition, Children Growth Research Center, Qazvin University of Medical Sciences , Qazvin, Islamic Republic of Iran
| | - Sima Rafiei
- Department of Healthcare Management, School of Health, Qazvin University of Medical Sciences , Qazvin, Islamic Republic of Iran
| | - Fariba Zahedifar
- Department of Health Education, School of Health, Qazvin University of Medical Sciences , Qazvin, Islamic Republic of Iran
| | - Ameneh Barikani
- Department of Social Medicine, Children Growth Research Center, Qazvin University of Medical Sciences , Qazvin, Islamic Republic of Iran
| |
Collapse
|
29
|
Breast Milk Supply of MicroRNA Associated with Leptin and Adiponectin Is Affected by Maternal Overweight/Obesity and Influences Infancy BMI. Nutrients 2019; 11:nu11112589. [PMID: 31661820 PMCID: PMC6893542 DOI: 10.3390/nu11112589] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/04/2019] [Accepted: 10/07/2019] [Indexed: 12/17/2022] Open
Abstract
Breast milk constitutes a dietary source of leptin, adiponectin and microRNAs (miRNAs) for newborns. Expression of miRNAs previously associated with maternal obesity, leptin or adiponectin function were assessed and their impact on infant weight analyzed. Milk samples were collected (at month 1, 2, and 3) from a cohort of 59 healthy lactating mothers (38 normal-weight and 21 overweight/obese (BMI ≥ 25)), and infant growth was followed up to 2 years of age. Thirteen miRNAs, leptin and adiponectin were determined in milk. Leptin, adiponectin and miRNA showed a decrease over time of lactation in normal-weight mothers that was altered in overweight/obesity. Furthermore, negative correlations were observed in normal-weight mothers between the expression of miRNAs in milk and the concentration of leptin or adiponectin, but were absent in overweight/obesity. Moreover, miRNAs negatively correlated with infant BMI only in normal-weight mothers (miR-103, miR-17, miR-181a, miR-222, miR-let7c and miR-146b). Interestingly, target genes of milk miRNAs differently regulated in overweight/obesity could be related to neurodevelopmental processes. In conclusion, a set of miRNAs present in breast milk, in close conjunction with leptin and adiponectin, are natural bioactive compounds with the potential to modulate infant growth and brain development, an interplay that is disturbed in the case of maternal overweight/obesity.
Collapse
|
30
|
Law JM, Morris DE, Astle V, Finn E, Muros JJ, Robinson LJ, Randell T, Denvir L, Symonds ME, Budge H. Brown Adipose Tissue Response to Cold Stimulation Is Reduced in Girls With Autoimmune Hypothyroidism. J Endocr Soc 2019; 3:2411-2426. [PMID: 31777769 PMCID: PMC6872489 DOI: 10.1210/js.2019-00342] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 10/04/2019] [Indexed: 01/15/2023] Open
Abstract
Objective The interaction between thyroid status and brown adipose tissue (BAT) activation is complex. We assessed the effect of autoimmune hypothyroidism (ATD) in female children on BAT activation, measured using infrared thermography. Design Twenty-six female participants (14 with ATD and 12 healthy controls) between 5 and 17 years of age attended a single study session. Thermal images were taken of the supraclavicular region before, and after, the introduction of a cool stimulus. Results Participants with ATD had lower resting (hypothyroid, 34.9 ± 0.7°C; control, 35.4 ± 0.5°C; P = 0.03) and stimulated (hypothyroid, 35.0 ± 0.6°C; control, 35.5 ± 0.5°C; P = 0.04) supraclavicular temperatures compared with controls, but there was no difference between groups in the temperature increase with stimulation. BAT activation, calculated as the relative temperature change comparing the supraclavicular temperature to a sternal reference region, was reduced in participants with ATD (hypothyroid, 0.1 ± 0.1°C; control, 0.2 ± 0.2°C; P = 0.04). Children with ATD were frequently biochemically euthyroid due to replacement therapy, but, despite this, increased relative supraclavicular temperature was closely associated with increased TSH (r = 0.7, P = 0.01) concentrations. Conclusions Girls with ATD had an attenuated thermogenic response to cold stimulation compared with healthy controls, but, contrary to expectation, those with suboptimal biochemical control (with higher TSH) showed increased BAT activation. This suggests that the underlying disease process may have a negative effect on BAT response, but high levels of TSH can mitigate, and even stimulate, BAT activity. In summary, thyroid status is a complex determinant of BAT activity in girls with ATD.
Collapse
Affiliation(s)
- James M Law
- Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, University of Nottingham, Nottingham, United Kingdom
| | - David E Morris
- Bioengineering Research Group, Faculty of Engineering, University of Nottingham, Nottingham, United Kingdom
| | - Valerie Astle
- Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, University of Nottingham, Nottingham, United Kingdom
| | - Ellie Finn
- School of Medicine, Monash University, Melbourne, Victoria, Australia
| | - José Joaquín Muros
- Department of Food Science, School of Pharmacy, University of Granada, Granada, Spain
| | - Lindsay J Robinson
- Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, University of Nottingham, Nottingham, United Kingdom
| | - Tabitha Randell
- Nottingham Children's Hospital, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Louise Denvir
- Nottingham Children's Hospital, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Michael E Symonds
- Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, University of Nottingham, Nottingham, United Kingdom.,Nottingham Digestive Disease Centre and Biomedical Research Centre, University of Nottingham, Nottingham, United Kingdom
| | - Helen Budge
- Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
31
|
Cheng H, Furnham A. Childhood locus of control and self-esteem, education, psychological distress and physical exercise as predictors of adult obesity. J Public Health (Oxf) 2019; 41:439-446. [PMID: 30239855 DOI: 10.1093/pubmed/fdy125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/26/2018] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To investigate to what extent locus of control, self-esteem, psychological distress, physical exercise, as well as socio-demographic factors are associated with obesity in 42-year-old adults in a longitudinal birth cohort study. METHOD The sample consisted of 5645 participants born in Great Britain in 1970 and followed up at 10, 34 and 42 years with data on body mass index measured at 34 and 42 years. RESULTS There was an increase of adult obesity from 15.5% at age 34 to 21.2% at 42 years. Locus of control and self-esteem measured at age 10 years, psychological distress and educational qualifications assessed at age 34, and current occupational levels and physical exercise were all significantly associated with adult obesity at age 42. The associations remained significant after controlling for birth weight and gestation, maternal and paternal BMI, childhood BMI, and intelligence. CONCLUSION Childhood locus of control and self-esteem, educational qualifications, psychological distress and physical exercise were all significantly and independently associated with adult obesity.
Collapse
Affiliation(s)
- Helen Cheng
- Department of Psychology, University College London, 26 Bedford Way, London WC1E 6BT, UK.,ESRC Centre for Learning and Life Chances in Knowledge Economies and Societies, Institute of Education, University College London, London WC1H 0AL, UK
| | - Adrian Furnham
- BI Norwegian Business School, Nydalsveien 37, 0484 Oslo, Norway
| |
Collapse
|
32
|
Badran M, Yassin BA, Lin DTS, Kobor MS, Ayas N, Laher I. Gestational intermittent hypoxia induces endothelial dysfunction, reduces perivascular adiponectin and causes epigenetic changes in adult male offspring. J Physiol 2019; 597:5349-5364. [PMID: 31441069 DOI: 10.1113/jp277936] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS Obstructive sleep apnoea (OSA) is characterized by intermittent hypoxia, which causes oxidative stress and inflammation and increases the risk of cardiovascular disease. OSA during pregnancy causes adverse maternal and fetal outcomes. The effects of pre-existing OSA in pregnant women on cardiometabolic outcomes in the offspring are unknown. We evaluated basic metabolic parameters, as well as aortic vascular and perivascular adipose tissue (PVAT) function in response to adiponectin, and examined DNA methylation of adiponectin gene promoter in PVAT in 16-week-old adult offspring exposed to gestational intermittent hypoxia (GIH). GIH decreased body weights at week 1 in both male and female offspring, and caused subsequent increases in body weight and food consumption in male offspring only. Adult female offspring had normal levels of lipids, glucose and insulin, with no endothelial dysfunction. Adult male offspring exhibited dyslipidaemia, insulin resistance and hyperleptinaemia. Decreased endothelial-dependent vasodilatation, loss of anti-contractile activity of PVAT and low circulating PVAT adiponectin levels, as well as increased pro-inflammatory gene expression and DNA methylation of adiponectin gene promoter, occurred in adult male offspring. Our results suggest that male offspring of women with OSA could be at risk of developing cardiometabolic disease during adulthood. ABSTRACT Perturbations during pregnancy can program the offspring to develop cardiometabolic diseases later in life. Obstructive sleep apnoea (OSA) is a chronic condition that frequently affects pregnancies and leads to adverse fetal outcomes. We assessed the offspring of female mice experiencing gestational intermittent hypoxia (GIH), a hallmark of OSA, for changes in metabolic profiles, aortic nitric oxide (NO)-dependent relaxations, perivascular adipose tissue (PVAT) anti-contractile activities and the responses to adiponectin, and DNA methylation of the adiponectin gene promoter in PVAT tissue. Pregnant mouse dams were exposed to intermittent hypoxic cycles ( F I O 2 21-12%) for 18 days. GIH resulted in lower body weights of pups at week 1, followed by significant weight gain by week 16 of age in male but not female offspring. Plasma lipids, leptin and insulin resistance were higher in GIH male adult offspring. Endothelium-dependent relaxation in response to ACh and the anti-contractile activity of PVAT in the abdominal aorta was reduced in GIH adult male offspring. Incubation of arteries from GIH adult male offspring with adiponectin restored the anti-contractile activity of PVAT. Both circulating and PVAT tissue homogenate levels of adiponectin, as well as gene expression of adiponectin in PVAT, were lower in GIH male offspring, along with an increased gene expression of inflammatory cytokines. Pyrosequencing of adiponectin gene promoter in PVAT showed increased DNA methylation in GIH male offspring. Our results indicate that GIH leads to vascular disease in adult male offspring through PVAT dysfunction, which was associated with low adiponectin levels and epigenetic modifications on the adiponectin gene promoter.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Bisher Abu Yassin
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - David Tse Shen Lin
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Najib Ayas
- Divisions of Critical Care and Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Sleep Disorders Program, UBC Hospital, Vancouver, BC, Canada.,Division of Critical Care Medicine, Providence Healthcare, Vancouver, BC, Canada
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
33
|
McLean C, Jun S, Kozyrskyj A. Impact of maternal smoking on the infant gut microbiota and its association with child overweight: a scoping review. World J Pediatr 2019; 15:341-349. [PMID: 31290060 DOI: 10.1007/s12519-019-00278-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/13/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND Childhood obesity is a growing public health concern with evidence demonstrating that while infant exposure to maternal smoking is linked to low birth weight at birth, there is a rapid catch up in weight and increased risk of obesity in later life. This scoping review aims to synthesize up-to-date evidence on the impact of maternal smoking on the infant gut microbiota and its association with child overweight. METHODS We conducted a PRISMA-compliant scoping review. Primary population-based cohort studies published between 1900 and April 2018 were included. Relevant publications were retrieved from seven databases: PubMed, Medline, Embase, Scopus, Biosis, Cochrane library, and Web of Science Core Collection. RESULTS A total of three prospective cohort studies were included which utilized high-throughput 16S rRNA gene sequencing to assess the gut microbiota and included a total of 1277 infant/neonatal participants. Neonates exposed to environmental smoke had a higher relative abundance of Ruminococcus and Akkermansia. Infants exposed to environmental smoke during pregnancy or postnatally were found to have increased gut bacterial richness, particularly Firmicutes at 3 months of age, while 6-month-old infants born to smoking mothers had an increased abundance of Bacteroides and Staphylococcus. Elevated Firmicutes richness at 3 months of age was associated with elevated odds of child overweight and obesity at 1 and 3 years of age. CONCLUSION The limited evidence to date warrants further large scale, longitudinal studies to explore the impact of maternal smoking and environmental tobacco smoke on the infant gut microbiome and its relation to child overweight.
Collapse
Affiliation(s)
- Cara McLean
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada
| | - Shelly Jun
- Department of Pediatrics, University of Alberta, 3-527 Edmonton Clinic Health Academy, 11405-87 Avenue, Edmonton, AB, T6G 1C9, Canada
| | - Anita Kozyrskyj
- Department of Pediatrics, University of Alberta, 3-527 Edmonton Clinic Health Academy, 11405-87 Avenue, Edmonton, AB, T6G 1C9, Canada. .,School of Public Health, University of Alberta, Edmonton, Canada.
| |
Collapse
|
34
|
Ferchaud-Roucher V, Barner K, Jansson T, Powell TL. Maternal obesity results in decreased syncytiotrophoblast synthesis of palmitoleic acid, a fatty acid with anti-inflammatory and insulin-sensitizing properties. FASEB J 2019; 33:6643-6654. [PMID: 30811959 PMCID: PMC6463919 DOI: 10.1096/fj.201802444r] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/28/2019] [Indexed: 12/16/2022]
Abstract
The fetus is dependent on delivery of fatty acids (FAs) by the syncytiotrophoblast, the transporting epithelium of the human placenta. Obese pregnant women have dyslipidemia; however, whether obesity impacts placental lipid transport and metabolism remains to be fully established. Palmitoleic acid (POA), an FA with anti-inflammatory and insulin-sensitizing properties, is synthesized from palmitic acid (PA) catalyzed by stearoyl-coenzyme A desaturase (SCD) activity. We hypothesized that the uptake and incorporation of FAs and POA synthesis are reduced in primary human trophoblasts (PHTs) isolated from pregnancies complicated by maternal obesity. Villous cytotrophoblasts were isolated from 7 placentas of obese [body mass index (BMI) = 37.5 ± 1.9] and 12 normal (BMI = 23.6 ± 0.6) mothers. FA uptake and incorporation were assessed using uniformly labeled (U[13C])-FA mixtures of PA, oleic acid (OA), linoleic acid, and docosahexaenoic acid. Cellular [13C] FAs were quantified both in total cellular lipids and in lipid classes by GC-MS. Uptake and incorporation of [13C] FAs in total cellular lipids were not different in PHTs isolated from obese mothers compared with normal mothers. Only the concentration of OA was increased in the triglyceride fraction (P < 0.05) if the mother was obese. We found an isotopic enrichment of POA after U[13C]-PA treatment, demonstrating SCD activity in PHT cells. Labeled POA content and the POA:PA ratio were significantly lower in PHTs isolated from placentas of obese mothers compared with normal, healthy controls. Decreased syncytiotrophoblast POA synthesis may contribute to insulin resistance and low-grade inflammation in the mother, placenta, or fetus (or a combination of the 3) in pregnancies complicated by obesity.-Ferchaud-Roucher, V., Barner, K., Jansson, T., Powell, T. L. Maternal obesity results in decreased syncytiotrophoblast synthesis of palmitoleic acid, a fatty acid with anti-inflammatory and insulin-sensitizing properties.
Collapse
Affiliation(s)
- Véronique Ferchaud-Roucher
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kelsey Barner
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Theresa L. Powell
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
35
|
Liberini CG, Lhamo R, Ghidewon M, Ling T, Juntereal N, Chen J, Cao A, Stein LM, Hayes MR. Liraglutide pharmacotherapy reduces body weight and improves glycaemic control in juvenile obese/hyperglycaemic male and female rats. Diabetes Obes Metab 2019; 21:866-875. [PMID: 30456866 PMCID: PMC7274726 DOI: 10.1111/dom.13591] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/05/2018] [Accepted: 11/15/2018] [Indexed: 12/16/2022]
Abstract
AIMS To examine whether the glucagon-like peptide-1 receptor agonist liraglutide could be used in juvenile male and female rats as an anti-obesity/diabetic pharmaceutical to prevent not only adolescent obesity/hyperglycaemia, but also early-adult onset obesity. MATERIAL AND METHODS Pregnant dams were fed either standard chow or a high-fat, high-sucrose diet (HFSD) from gestational day 2, throughout pregnancy and lactation. Offspring were weaned onto the respective maternal diet. Juveniles received daily subcutaneous injection of liraglutide (50 μg/kg, from postnatal day [PND]30 to PND40 and 200 μg/kg from PND40 to PND60) or vehicle. Food intake, body weight and glycaemic levels were evaluated across the experimental period. RESULTS Chronic liraglutide administration in juveniles prevented body weight gain in males and retained a normoglycaemic profile in both male and female rats. CONCLUSION These preclinical data suggest that maternal and early-life consumption of an HFSD increases caloric intake, body weight gain and hyperglycaemia, a collective set of unwanted metabolic effects that appear to be treatable in juveniles with liraglutide pharmacotherapy intervention.
Collapse
Affiliation(s)
- Claudia G. Liberini
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Rinzin Lhamo
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Misgana Ghidewon
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Tyler Ling
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Nina Juntereal
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Jack Chen
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Anh Cao
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Lauren M. Stein
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Matthew R. Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
36
|
Gupta MB, Jansson T. Novel roles of mechanistic target of rapamycin signaling in regulating fetal growth†. Biol Reprod 2019; 100:872-884. [PMID: 30476008 PMCID: PMC6698747 DOI: 10.1093/biolre/ioy249] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/08/2018] [Accepted: 11/19/2018] [Indexed: 12/18/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) signaling functions as a central regulator of cellular metabolism, growth, and survival in response to hormones, growth factors, nutrients, energy, and stress signals. Mechanistic TOR is therefore critical for the growth of most fetal organs, and global mTOR deletion is embryonic lethal. This review discusses emerging evidence suggesting that mTOR signaling also has a role as a critical hub in the overall homeostatic control of fetal growth, adjusting the fetal growth trajectory according to the ability of the maternal supply line to support fetal growth. In the fetus, liver mTOR governs the secretion and phosphorylation of insulin-like growth factor binding protein 1 (IGFBP-1) thereby controlling the bioavailability of insulin-like growth factors (IGF-I and IGF-II), which function as important growth hormones during fetal life. In the placenta, mTOR responds to a large number of growth-related signals, including amino acids, glucose, oxygen, folate, and growth factors, to regulate trophoblast mitochondrial respiration, nutrient transport, and protein synthesis, thereby influencing fetal growth. In the maternal compartment, mTOR is an integral part of a decidual nutrient sensor which links oxygen and nutrient availability to the phosphorylation of IGFBP-1 with preferential effects on the bioavailability of IGF-I in the maternal-fetal interface and in the maternal circulation. These new roles of mTOR signaling in the regulation fetal growth will help us better understand the molecular underpinnings of abnormal fetal growth, such as intrauterine growth restriction and fetal overgrowth, and may represent novel avenues for diagnostics and intervention in important pregnancy complications.
Collapse
Affiliation(s)
- Madhulika B Gupta
- Department of Pediatrics, University of Western Ontario, London, Ontario, Canada
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
- Children's Health Research Institute, London, Ontario, Canada
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, University of Colorado | Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
37
|
"The One Time You Have Control over What They Eat": A Qualitative Exploration of Mothers' Practices to Establish Healthy Eating Behaviours during Weaning. Nutrients 2019; 11:nu11030562. [PMID: 30845694 PMCID: PMC6471714 DOI: 10.3390/nu11030562] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 02/28/2019] [Accepted: 03/04/2019] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Weaning marks the transition from a milk-only diet to the consumption of solid foods. It is a time period where nutrition holds an undeniable importance and taste experiences have a long-lasting effect on food preferences. The factors and conditions that form parental feeding practices are yet to be fully understood; doing so can help target problematic behaviours and develop interventions aiming to modify them. OBJECTIVE This study used a qualitative methodology to gain a better understanding of parental experiences of weaning a child. Particular emphasis was placed on exploring the factors and conditions that favour the establishment of a healthy relationship with food in infancy and those that impede it. METHODS Thirty-seven mothers of healthy infants 3⁻14 months with no previous history of allergies or food-related disorders were recruited. Eight semi-structured focus group discussions were conducted, transcribed and analysed thematically. RESULTS Discussions revealed a number of opportunities to establish healthy eating habits during weaning, as well as relevant challenges. Important opportunities included: acting as a role model for healthy foods; giving multiple opportunities to try a food; food variety "so you don't have a fussy eater"; and without food variety "things aren't going to work properly". Additionally, some of the challenges identified were: misconceptions about the definition of food variety; and distractions occurring during feeding. CONCLUSIONS Mothers were mindful of the need to provide their children with appropriate nutritional stimuli during weaning. They were aware of their role in influencing their infants' likes and used strategies such as modelling and repeated food exposure. The importance of a diverse diet in infancy was acknowledged, although knowledge gaps exist in relation to its definition. Distractions were tactfully employed by mothers to assist feeding. Findings of this study have applications in developing interventions for nutritional education in the complementary feeding period.
Collapse
|
38
|
Mangwiro YTM, Cuffe JSM, Mahizir D, Anevska K, Gravina S, Romano T, Moritz KM, Briffa JF, Wlodek ME. Exercise initiated during pregnancy in rats born growth restricted alters placental mTOR and nutrient transporter expression. J Physiol 2019; 597:1905-1918. [PMID: 30734290 DOI: 10.1113/jp277227] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/24/2019] [Indexed: 02/06/2023] Open
Abstract
KEY POINTS Fetal growth is dependent on effective placental nutrient transportation, which is regulated by mammalian target of rapamycin (mTOR) complex 1 modulation of nutrient transporter expression. These transporters are dysregulated in pregnancies affected by uteroplacental insufficiency and maternal obesity. Nutrient transporters and mTOR were altered in placentae of mothers born growth restricted compared to normal birth weight dams, with maternal diet- and fetal sex-specific responses. Exercise initiated during pregnancy downregulated mTOR protein expression, despite an increase in mTOR activation in male associated placentae, and reduced nutrient transporter gene abundance, which was also dependent on maternal diet and fetal sex. Limited changes were characterized with exercise initiated before and continued throughout pregnancy in nutrient transporter and mTOR expression. Maternal exercise during pregnancy differentially regulated mTOR and nutrient transporters in a diet- and sex-specific manner, which likely aimed to improve late gestational placental growth and neonatal survival. ABSTRACT Adequate transplacental nutrient delivery is essential for fetoplacental development. Intrauterine growth restriction and maternal obesity independently alter placental nutrient transporter expression. Although exercise is beneficial for maternal health, limited studies have characterized how the timing of exercise initiation influences placental nutrient transport. Therefore, this study investigated the impact of maternal exercise on placental mechanistic target of rapamycin (mTOR) and nutrient transporter expression in growth restricted mothers and whether these outcomes were dependent on maternal diet or fetal sex. Uteroplacental insufficiency or sham surgery was induced on embryonic day (E) 18 in Wistar-Kyoto rats. F1 offspring were fed a chow or high-fat diet from weaning and at 16 weeks were randomly allocated to an exercise protocol: sedentary, exercised prior to and during pregnancy, or exercised during pregnancy only. Females were mated with normal males (20 weeks) and F2 placentae collected at E20. Exercise during pregnancy only, reduced mTOR protein expression in all groups and increased mTOR activation in male associated placentae. Exercise during pregnancy only, decreased the expression of amino acid transporters in a diet- and sex-specific manner. Maternal growth restriction altered mTOR and system A amino acid transporter expression in a sex- and diet-specific manner. These data highlight that maternal exercise initiated during pregnancy alters placental mTOR expression, which may directly regulate amino acid transporter expression, to a greater extent than exercise initiated prior to and continued during pregnancy, in a diet- and fetal sex-dependent manner. These findings highlight that the timing of exercise initiation is important for optimal placental function.
Collapse
Affiliation(s)
- Yeukai T M Mangwiro
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, 3083, Australia.,Department of Physiology, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - James S M Cuffe
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, 4072, Australia
| | - Dayana Mahizir
- Department of Physiology, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Kristina Anevska
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, 3083, Australia.,Department of Physiology, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Sogand Gravina
- Department of Physiology, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Tania Romano
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, 3083, Australia
| | - Karen M Moritz
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, 4072, Australia.,Child Health Research Centre, University of Queensland, South Brisbane, Queensland, 4101, Australia
| | - Jessica F Briffa
- Department of Physiology, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Mary E Wlodek
- Department of Physiology, University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
39
|
Draycott SAV, Liu G, Daniel ZC, Elmes MJ, Muhlhausler BS, Langley-Evans SC. Maternal dietary ratio of linoleic acid to alpha-linolenic acid during pregnancy has sex-specific effects on placental and fetal weights in the rat. Nutr Metab (Lond) 2019; 16:1. [PMID: 30622622 PMCID: PMC6318840 DOI: 10.1186/s12986-018-0330-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/20/2018] [Indexed: 01/30/2023] Open
Abstract
Background Increased consumption of linoleic acid (LA, omega-6) in Western diets coupled with the pro-inflammatory and adipogenic properties of its derivatives has led to suggestions that fetal exposure to this dietary pattern could be contributing to the intergenerational cycle of obesity. Method This study aimed to evaluate the effects of maternal consumption of a LA to alpha-linolenic acid (ALA) ratio similar to modern Western diets (9:1) compared to a lower ratio (1:1.5) on placental and fetal growth, and to determine any cumulative effects by feeding both diets at two total fat levels (18% vs 36% fat w/w). Female Wistar rats (n = 5–7/group) were assigned to one of the four experimental diets prior to mating until 20d of gestation. Results Fatty acid profiles of maternal and fetal blood and placental tissue at 20d gestation were different between dietary groups, and largely reflected dietary fatty acid composition. Female fetuses were heavier (2.98 ± 0.06 g vs 3.36 ± 0.07 g, P < 0.01) and male placental weight was increased (0.51 ± 0.02 g vs 0.58 ± 0.02 g, P < 0.05) in the low LA:ALA groups. Female fetuses of dams exposed to a 36% fat diet had a reduced relative liver weight irrespective of LA:ALA ratio (7.61 ± 0.22% vs 6.93 ± 0.19%, P < 0.05). These effects occurred in the absence of any effect of the dietary treatments on maternal bodyweight, fat deposition or expression of key lipogenic genes in maternal and fetal liver or maternal adipose tissue. Conclusion These findings suggest that both the total fat content as well as the LA:ALA ratio of the maternal diet have sex-specific implications for the growth of the developing fetus.
Collapse
Affiliation(s)
- Sally A V Draycott
- 1Food and Nutrition Research Group, Department of Food and Wine Science, School of Agriculture Food and Wine, University of Adelaide, Adelaide, Australia.,2School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Ge Liu
- 1Food and Nutrition Research Group, Department of Food and Wine Science, School of Agriculture Food and Wine, University of Adelaide, Adelaide, Australia.,3Healthy Mothers, Babies and Children's Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, 5001 Australia
| | - Zoe C Daniel
- 2School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Matthew J Elmes
- 2School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Beverly S Muhlhausler
- 1Food and Nutrition Research Group, Department of Food and Wine Science, School of Agriculture Food and Wine, University of Adelaide, Adelaide, Australia
| | - Simon C Langley-Evans
- 2School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| |
Collapse
|
40
|
Luan D, Mezuk B, Bauer KW. Remission of obesity among a nationally representative sample of US children. Pediatr Obes 2019; 14. [PMID: 30074306 DOI: 10.1111/ijpo.12457] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/25/2018] [Accepted: 06/15/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Little is known about the incidence and natural history of obesity remission among children outside of weight loss programmes. OBJECTIVES The objectives are to characterize and identify sociodemographic and early life predictors of obesity remission between kindergarten and eighth grade among a nationally representative sample of US children. METHODS The sample included children with obesity [age-specific and gender-specific body mass index percentile (BMI) ≥95] at the spring kindergarten assessment of the Early Childhood Longitudinal Study, Kindergarten Class of 1998-99. Weight categories across 8 years of follow-up were used to identify three transition patterns: persistent obesity remission, non-persistent obesity remission and non-remission. Weight, height and BMI changes between remission categories were examined and predictors of persistent remission were identified. RESULTS One-third of children with obesity in kindergarten experienced remission during follow-up and 21.6% of children experienced persistent remission through eighth grade. Female gender and high socio-economic status predicted persistent remission; these associations were attenuated after accounting for baseline BMI. Children experiencing persistent remission gained less weight across waves than those experiencing non-remission. CONCLUSIONS A meaningful proportion of young children with obesity experience remission by eighth grade. Further study is needed to identify factors that support obesity remission among children outside of treatment contexts.
Collapse
Affiliation(s)
- D Luan
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - B Mezuk
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA.,Institute for Social Research, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - K W Bauer
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| |
Collapse
|
41
|
Hollensted M, Ekstrøm CT, Pedersen O, Eiberg H, Hansen T, Gjesing AP. Genetic insights into fetal growth and measures of glycaemic regulation and adiposity in adulthood: a family-based study. BMC MEDICAL GENETICS 2018; 19:207. [PMID: 30514227 PMCID: PMC6278142 DOI: 10.1186/s12881-018-0718-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 11/14/2018] [Indexed: 01/26/2023]
Abstract
Background The genetics of fetal insulin release and/or action have been suggested to affect fetal growth, adult insulin resistance and adult body composition. The genetic correlation between body composition at birth versus glycaemic regulation and body composition in adulthood have, however, not been well studied. We therefore aimed to investigate these genetic correlations in a family-based cohort. Methods A Danish family cohort of 434 individuals underwent an oral glucose tolerance test with subsequent calculation of surrogate measures of serum insulin response and insulin sensitivity. Measures of fetal growth were retrieved from midwife journals. Heritability and genetic correlations were estimated using a variance component model. Results A high heritability of 0.80 was found for birth weight, whereas ponderal index had a heritability of 0.46. Adult insulin sensitivity measured as Matsuda index was genetically correlated with both birth weight and ponderal index (ρG = 0.36 (95% CI: 0.03; 0.69) and ρG = 0.52 (95% CI, 0.15; 0.89), respectively). Only birth weight showed a significant genetic correlation with adult weight (ρG = 0.38 (95% CI: 0.09; 0.67)) whereas only ponderal index was genetically inversely correlated with fasting insulin (ρG = - 0.47 (95% CI: - 0.86; - 0.08) and area under the curve for insulin release during the oral glucose tolerance test (ρG = - 0.66 (95% CI: - 1.13; - 0.19)). Individual as well as combined adjustment for 45 selected birth weight, obesity and type 2 diabetes susceptibility gene variants did not affect the correlations. Conclusions The genetics of both birth weight and ponderal index appear to be under the same genetic influence as adult insulin resistance. Furthermore, ponderal index and adult insulin release seem to be partly shared, as well as the genetics of birth weight and adult weight. Word count abstract: 281. Electronic supplementary material The online version of this article (10.1186/s12881-018-0718-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mette Hollensted
- Section of Metabolic Genetics, The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen, Denmark. .,The Danish Diabetes Academy, Odense, Denmark.
| | - Claus T Ekstrøm
- Section of Biostatistics, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- Section of Metabolic Genetics, The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen, Denmark
| | - Hans Eiberg
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Section of Metabolic Genetics, The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen, Denmark.,Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Anette Prior Gjesing
- Section of Metabolic Genetics, The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen, Denmark
| |
Collapse
|
42
|
Shrestha D, Rahman ML, Workalemahu T, Zhu C, Tekola-Ayele F. Influence of Fetal and Maternal Genetic Susceptibility to Obesity on Birthweight in African Ancestry Populations. Front Genet 2018; 9:511. [PMID: 30450111 PMCID: PMC6224338 DOI: 10.3389/fgene.2018.00511] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/12/2018] [Indexed: 02/02/2023] Open
Abstract
Fetal and maternal genetic propensity to obesity can influence birthweight. We investigated the effects of fetal and maternal genetic risk of obesity on birthweight and evaluated whether these genetic influences modify the well-known association between maternal pre-pregnancy body mass index (BMI) and birthweight. In 950 mother-baby pairs of African ancestry, a genetic risk score for adulthood obesity was generated for mothers (mGRS) and their babies (bGRS) as the weighted sum of BMI-increasing alleles of 97 single nucleotide polymorphisms known to be associated with BMI. The median GRS value was used as a cut-off to define high or low bGRS and mGRS. High bGRS was significantly associated with 70 g lower birthweight (95% Confidence Interval [CI] = −127.4 to −12.4) compared to low bGRS. mGRS was positively correlated with birthweight but the association was not significant. mGRS modified the significant birthweight-increasing effect of maternal pre-pregnancy BMI (P-for-interaction = 0.03); among mothers with low mGRS, those who were overweight or obese had 127.7 g heavier babies (95% CI = 27.1 to 228.2) compared to those who had normal weight. In summary, fetal obesity genetic risk loci exert direct influence on birthweight, and maternal loci modify the effect of pre-pregnancy BMI on birthweight.
Collapse
Affiliation(s)
- Deepika Shrestha
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Mohammad L Rahman
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Tsegaselassie Workalemahu
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Chunming Zhu
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Fasil Tekola-Ayele
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
43
|
Corbin KD, Driscoll KA, Pratley RE, Smith SR, Maahs DM, Mayer-Davis EJ. Obesity in Type 1 Diabetes: Pathophysiology, Clinical Impact, and Mechanisms. Endocr Rev 2018; 39:629-663. [PMID: 30060120 DOI: 10.1210/er.2017-00191] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 06/21/2018] [Indexed: 02/07/2023]
Abstract
There has been an alarming increase in the prevalence of obesity in people with type 1 diabetes in recent years. Although obesity has long been recognized as a major risk factor for the development of type 2 diabetes and a catalyst for complications, much less is known about the role of obesity in the initiation and pathogenesis of type 1 diabetes. Emerging evidence suggests that obesity contributes to insulin resistance, dyslipidemia, and cardiometabolic complications in type 1 diabetes. Unique therapeutic strategies may be required to address these comorbidities within the context of intensive insulin therapy, which promotes weight gain. There is an urgent need for clinical guidelines for the prevention and management of obesity in type 1 diabetes. The development of these recommendations will require a transdisciplinary research strategy addressing metabolism, molecular mechanisms, lifestyle, neuropsychology, and novel therapeutics. In this review, the prevalence, clinical impact, energy balance physiology, and potential mechanisms of obesity in type 1 diabetes are described, with a special focus on the substantial gaps in knowledge in this field. Our goal is to provide a framework for the evidence base needed to develop type 1 diabetes-specific weight management recommendations that account for the competing outcomes of glycemic control and weight management.
Collapse
Affiliation(s)
- Karen D Corbin
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| | - Kimberly A Driscoll
- Department of Pediatrics, School of Medicine, University of Colorado Denver, Aurora, Colorado.,Barbara Davis Center for Diabetes, Aurora, Colorado
| | - Richard E Pratley
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| | - Steven R Smith
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| | - David M Maahs
- Division of Pediatric Endocrinology, Department of Pediatrics, Stanford University, Stanford, California
| | - Elizabeth J Mayer-Davis
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | |
Collapse
|
44
|
Sliwowska JH, Ziarniak K, Dudek M, Matuszewska J, Tena-Sempere M. Dangerous liaisons for pubertal maturation: the impact of alcohol consumption and obesity on the timing of puberty†. Biol Reprod 2018; 100:25-40. [DOI: 10.1093/biolre/ioy168] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/25/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- Joanna H Sliwowska
- Laboratory of Neurobiology, Institute of Zoology, Poznan University of Life Sciences, Poznan, Poland
| | - Kamil Ziarniak
- Laboratory of Neurobiology, Institute of Zoology, Poznan University of Life Sciences, Poznan, Poland
| | - Monika Dudek
- Laboratory of Neurobiology, Institute of Zoology, Poznan University of Life Sciences, Poznan, Poland
| | - Julia Matuszewska
- Laboratory of Neurobiology, Institute of Zoology, Poznan University of Life Sciences, Poznan, Poland
| | - Manuel Tena-Sempere
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, and Instituto Maimonides de Investigación Biomédica de Cordoba (IMIBIC), Cordoba, Spain
- CIBEROBN - Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| |
Collapse
|
45
|
Elevations of inflammatory proteins in neonatal blood are associated with obesity and overweight among 2-year-old children born extremely premature. Pediatr Res 2018; 83:1110-1119. [PMID: 29244802 PMCID: PMC6003823 DOI: 10.1038/pr.2017.313] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 11/22/2017] [Indexed: 01/16/2023]
Abstract
BackgroundChildhood obesity is associated with elevated blood concentrations of inflammation markers. It is not known to what extent inflammation precedes the development of obesity.MethodsIn a cohort of 882 infants born before 28 weeks of gestation, we examined relationships between concentrations of 25 inflammation-related proteins in blood obtained during the first two postnatal weeks and body mass index at 2 years of age.ResultsAmong children delivered for spontaneous indications (n=734), obesity was associated with elevated concentrations of four proteins (IL-1β, IL-6, TNF-R1, and MCP-1) on the first postnatal day; one protein (IL-6) on postnatal day 7; and two proteins (ICAM-3 and VEGF-R1) on postnatal day 14. Among children delivered for maternal or fetal indications (n=148), obesity was associated with elevated concentrations of seven proteins on the 14th postnatal day. In multivariable models in the spontaneous indications subsample, elevated IL-6 on day 1 predicted obesity (odds ratio: 2.9; 95% confidence limits: 1.2, 6.8), whereas elevated VCAM-1 on day 14 predicted overweight at 2 years of age (odds ratio: 2.3; 95% confidence limits: 1.2, 4.3).ConclusionsIn this cohort, neonatal systemic inflammation preceded the onset of obesity, suggesting that inflammation might contribute to the development of obesity.
Collapse
|
46
|
Harris JE, Baer LA, Stanford KI. Maternal Exercise Improves the Metabolic Health of Adult Offspring. Trends Endocrinol Metab 2018; 29:164-177. [PMID: 29402734 PMCID: PMC5826804 DOI: 10.1016/j.tem.2018.01.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/04/2018] [Accepted: 01/05/2018] [Indexed: 12/22/2022]
Abstract
The intrauterine environment can modulate the course of development and confer an enduring effect on offspring health. The effects of maternal diet to impair offspring metabolic health are well established, but the effects of maternal exercise on offspring metabolic health have been less defined. Because physical exercise is a treatment for obesity and type 2 diabetes (T2D), maternal exercise is an appealing intervention to positively influence the intrauterine environment and improve the metabolic health of offspring. Recent research has provided insights into the effects of maternal exercise on the metabolic health of adult offspring, which is the focus of this review.
Collapse
Affiliation(s)
- Johan E Harris
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Lisa A Baer
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Kristin I Stanford
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
47
|
Nakagawa Y, Nakanishi T, Satake E, Matsushita R, Saegusa H, Kubota A, Natsume H, Shibata Y, Fujisawa Y. Postnatal BMI changes in children with different birthweights: A trial study for detecting early predictive factors for pediatric obesity. Clin Pediatr Endocrinol 2018; 27:19-29. [PMID: 29403153 PMCID: PMC5792818 DOI: 10.1297/cpe.27.19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 10/04/2017] [Indexed: 01/16/2023] Open
Abstract
The purpose of this study was to clarify the degree of early postnatal growth by birthweight and detect early predictive factors for pediatric obesity. Body mass index (BMI) and degree of obesity were examined in children in the fourth year of elementary school and second year of junior high school. Their BMI at birth and three years of age were also examined. Based on birthweight, participants were divided into three groups: low (< 2500 g), middle (2500-3500 g), and high (> 3500 g). Furthermore, according to the degree of obesity, they were divided into two groups: obese (20% ≤) and non-obese (20% >). The change of BMI from birth to three years of age (ΔBMI) showed a strong inverse relationship with birthweight and was significantly different among the three birthweight groups (low > middle > high). The ΔBMI and BMI at three years of age were higher in obese than in non-obese children and showed significant positive correlations with the degree of obesity. Early postnatal growth might be determined by birthweight and was higher in obese than in non-obese children. The ΔBMI from birth to three years of age and BMI at age of three years could be predictive factors for pediatric obesity.
Collapse
Affiliation(s)
- Yuichi Nakagawa
- Department of Internal Medicine and Pediatrics, Shiraume Toyooka Hospital, Shizuoka, Japan.,Department of Pediatrics, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Toshiki Nakanishi
- Department of Pediatrics, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Eiichiro Satake
- Department of Pediatrics, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Rie Matsushita
- Department of Pediatrics, Hamamatsu University School of Medicine, Shizuoka, Japan.,Department of Pediatrics, Kikugawa General Hospital, Shizuoka, Japan
| | - Hirokazu Saegusa
- Department of Pediatrics, Enshu General Hospital, Shizuoka, Japan
| | - Akira Kubota
- Department of Pediatrics, Chutoen General Medical Center, Shizuoka, Japan
| | - Hiromune Natsume
- Department of Pediatrics, Kosai General Hospital, Shizuoka, Japan
| | - Yukinobu Shibata
- Department of Pediatrics, Hamamatsu Red Cross Hospital, Shizuoka, Japan
| | - Yasuko Fujisawa
- Department of Pediatrics, Hamamatsu University School of Medicine, Shizuoka, Japan
| |
Collapse
|
48
|
Crispi F, Miranda J, Gratacós E. Long-term cardiovascular consequences of fetal growth restriction: biology, clinical implications, and opportunities for prevention of adult disease. Am J Obstet Gynecol 2018; 218:S869-S879. [PMID: 29422215 DOI: 10.1016/j.ajog.2017.12.012] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 02/07/2023]
Abstract
In the modern world, cardiovascular disease is a leading cause of death for both men and women. Epidemiologic studies consistently have suggested an association between low birthweight and/or fetal growth restriction and increased rate of cardiovascular mortality in adulthood. Furthermore, experimental and clinical studies have demonstrated that sustained nutrient and oxygen restriction that are associated with fetal growth restriction activate adaptive cardiovascular changes that might explain this association. Fetal growth restriction results in metabolic programming that may increase the risk of metabolic syndrome and, consequently, of cardiovascular morbidity in the adult. In addition, fetal growth restriction is strongly associated with fetal cardiac and arterial remodeling and a subclinical state of cardiovascular dysfunction. The cardiovascular effects ocurring in fetal life, includes cardiac morphology changes, subclinical myocardial dysfunction, arterial remodeling, and impaired endothelial function, persist into childhood and adolescence. Importantly, these changes have been described in all clinical presentations of fetal growth restriction, from severe early- to milder late-onset forms. In this review we summarize the current evidence on the cardiovascular effects of fetal growth restriction, from subcellular to organ structure and function as well as from fetal to early postnatal life. Future research needs to elucidate whether and how early life cardiovascular remodeling persists into adulthood and determines the increased cardiovascular mortality rate described in epidemiologic studies.
Collapse
|
49
|
Wang KW, de Souza RJ, Fleming A, Johnston DL, Zelcer SM, Rassekh SR, Burrow S, Thabane L, Samaan MC. Birth weight and body mass index z-score in childhood brain tumors: A cross-sectional study. Sci Rep 2018; 8:1642. [PMID: 29374278 PMCID: PMC5786044 DOI: 10.1038/s41598-018-19924-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 01/11/2018] [Indexed: 01/31/2023] Open
Abstract
Children with brain tumors (CBT) are at higher risk of cardiovascular disease and type 2 diabetes compared to the general population, in which birth weight is a risk factor for these diseases. However, this is not known in CBT. The primary aim of this study was to explore the association between birth weight and body mass measures in CBT, compared to non-cancer controls. This is a secondary data analysis using cross-sectional data from the CanDECIDE study (n = 78 CBT and n = 133 non-cancer controls). Age, sex, and birth weight (grams) were self-reported, and confirmed through examination of the medical records. Body mass index (BMI) was calculated from height and weight measures and reported as kg/m2. BMI z-scores were obtained for subjects under the age of 20 years. Multivariable linear regression was used to evaluate the relationship between birth weight and BMI and BMI z-score, adjusted for age, sex, puberty, and fat mass percentage. Higher birth weight was associated with higher BMI and BMI z-score among CBT and controls. In conclusion, birth weight is a risk factor for higher body mass during childhood in CBT, and this may help the identification of children at risk of future obesity and cardiometabolic risk.
Collapse
Affiliation(s)
- Kuan-Wen Wang
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Division of Pediatric Endocrinology, McMaster Children's Hospital, Hamilton, Ontario, Canada
- Medical Sciences Graduate Program, McMaster University, Hamilton, Ontario, Canada
| | - Russell J de Souza
- Medical Sciences Graduate Program, McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Adam Fleming
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Medical Sciences Graduate Program, McMaster University, Hamilton, Ontario, Canada
- Division of Pediatric Hematology/Oncology, McMaster Children's Hospital, Hamilton, Ontario, Canada
| | - Donna L Johnston
- Division of Pediatric Hematology/Oncology, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Shayna M Zelcer
- Pediatric Hematology Oncology, Children's Hospital, London Health Sciences Center, London, Ontario, Canada
| | - Shahrad Rod Rassekh
- Division of Pediatric Hematology/Oncology/BMT, Department of Pediatrics, British Columbia's Children's Hospital, Vancouver, BC, Canada
| | - Sarah Burrow
- Division of Orthopedic Surgery, Department of Surgery, McMaster University Medical Centre, Hamilton, Ontario, Canada
| | - Lehana Thabane
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Anesthesia, McMaster University, Hamilton, Ontario, Canada
- Centre for Evaluation of Medicines, St. Joseph's Health Care, Hamilton, Ontario, Canada
- Biostatistics Unit, St Joseph's Healthcare-Hamilton, Hamilton, Ontario, Canada
| | - M Constantine Samaan
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada.
- Division of Pediatric Endocrinology, McMaster Children's Hospital, Hamilton, Ontario, Canada.
- Medical Sciences Graduate Program, McMaster University, Hamilton, Ontario, Canada.
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
50
|
Popova P, Vasilyeva L, Tkachuck A, Puzanov M, Golovkin A, Bolotko Y, Pustozerov E, Vasilyeva E, Li O, Zazerskaya I, Dmitrieva R, Kostareva A, Grineva E. A Randomised, Controlled Study of Different Glycaemic Targets during Gestational Diabetes Treatment: Effect on the Level of Adipokines in Cord Blood and ANGPTL4 Expression in Human Umbilical Vein Endothelial Cells. Int J Endocrinol 2018; 2018:6481658. [PMID: 29861725 PMCID: PMC5976949 DOI: 10.1155/2018/6481658] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/27/2018] [Accepted: 03/14/2018] [Indexed: 12/30/2022] Open
Abstract
Our aim was to study the expression of adipokine-encoding genes (leptin, adiponectin, and angiopoietin-like protein 4 (ANGPTL4)) in human umbilical vein endothelial cells (HUVECs) and adipokine concentration in cord blood from women with gestational diabetes mellitus (GDM) depending on glycaemic targets. GDM patients were randomised to 2 groups per target glycaemic levels: GDM1 (tight glycaemic targets, fasting blood glucose < 5.1 mmol/L and <7.0 mmol/L postprandial, N = 20) and GDM2 (less tight glycaemic targets, <5.3 mmol/L and < 7.8 mmol/L, respectively, N = 21). The control group included 25 women with normal glucose tolerance. ANGPTL4 expression was decreased in the HUVECs from GDM patients versus the control group (23.11 ± 5.71, 21.47 ± 5.64, and 98.33 ± 20.92, for GDM1, GDM2, and controls; p < 0.001) with no difference between GDM1 and GDM2. The level of adiponectin gene expression was low and did not differ among the groups. Leptin gene expression was undetectable in HUVECs. In cord blood, leptin/adiponectin ratio (LAR) was increased in GDM2 compared to controls and GDM1 (p = 0.038) and did not differ between GDM1 and controls. Tight glycaemic targets were associated with normalisation of increased LAR in the cord blood. ANGPTL4 expression was downregulated in HUVECs of newborns from GDM mothers and was not affected by the intensity of glycaemic control.
Collapse
Affiliation(s)
- P. Popova
- Almazov National Medical Research Centre, Saint Petersburg, Russia
- Department of Internal Diseases and Endocrinology, St. Petersburg Pavlov State Medical University, Saint Petersburg, Russia
| | - L. Vasilyeva
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - A. Tkachuck
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - M. Puzanov
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - A. Golovkin
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Y. Bolotko
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - E. Pustozerov
- Almazov National Medical Research Centre, Saint Petersburg, Russia
- Department of Biomedical Engineering, Saint Petersburg State Electrotechnical University, Saint Petersburg, Russia
| | - E. Vasilyeva
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - O. Li
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - I. Zazerskaya
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - R. Dmitrieva
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - A. Kostareva
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - E. Grineva
- Almazov National Medical Research Centre, Saint Petersburg, Russia
- Department of Internal Diseases and Endocrinology, St. Petersburg Pavlov State Medical University, Saint Petersburg, Russia
| |
Collapse
|