1
|
Forestell CA. Does Maternal Diet Influence Future Infant Taste and Odor Preferences? A Critical Analysis. Annu Rev Nutr 2024; 44:313-337. [PMID: 38724030 DOI: 10.1146/annurev-nutr-121222-101404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
The dietary choices a mother makes during pregnancy offer her developing fetus its earliest exposure to the family's culinary preferences. This comprehensive literature review synthesizes five decades of research, which has provided valuable insights into fetal flavor learning. Converging evidence across various species supports the functionality of fetal chemoreceptive systems by the end of gestation, enabling the detection of an extensive array of chemosensory cues derived from the maternal diet and transmitted to the amniotic fluid. The fetus effectively encodes these flavors, resulting in their enhanced acceptance after birth. While existing studies predominantly concentrate on fetal learning about odor volatiles, limited evidence suggests a capacity for learning about gustatory (i.e., taste) properties. Examining whether these prenatal odor, taste, and flavor experiences translate into enduring shifts in dietary behaviors beyond weaning remains a crucial avenue for further investigation.
Collapse
|
2
|
Wen J, Aihemaitijiang S, Li H, Zhou Y, Liu J. Association of maternal body mass index and gestational weight gain rate with cardiometabolic traits in childhood: A prospective cohort study. Nutr Metab Cardiovasc Dis 2024:S0939-4753(24)00279-5. [PMID: 39168803 DOI: 10.1016/j.numecd.2024.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND AND AIMS Evidence on the association of maternal obesity with offspring cardiometabolic health is limited, particularly for the Asian population. We aimed to examine the associations of maternal body mass index (BMI) in early pregnancy and gestational weight gain (GWG) rate in mid- and late-pregnancy with childhood cardiometabolic traits. METHODS AND RESULTS We used data of 1452 mother-child pairs from a population-based prospective cohort study in China. Maternal BMI in early pregnancy and GWG rate in mid- and late-pregnancy were calculated. Childhood cardiometabolic traits were assessed at aged 4-7 years, including BMI, BMI-z, systolic blood pressure (SBP), diastolic blood pressure, high-density lipoprotein-cholesterol, low-density lipoprotein-cholesterol, total cholesterol, triglycerides, fasting glucose, and C-reactive protein. Each 1 kg/m2 increase in maternal BMI in early pregnancy was associated with 0.46% (95% confidence interval, 0.19%-0.72%) higher children BMI, 0.05 (0.02-0.08) higher BMI-z, 0.41% (0.22%-0.59%) higher waist circumference, and 0.24% (0.03%-0.46%) higher SBP. Each 1 kg/week higher GWG rate in mid- and late-pregnancy was associated with higher children SBP (4.58% [1.46%-7.71%]), triglycerides (18.28% [3.13%-33.44%]), and fasting glucose (5.83% [2.64%-9.02%]) and lower BMI-z (-0.45 [-0.82 to -0.08]). Additional adjustment for offspring BMI attenuated the associations for maternal BMI but not for GWG rate. CONCLUSIONS The increase in maternal BMI and GWG are associated with adverse cardiometabolic profiles in childhood. The association between maternal BMI and childhood cardiometabolic traits is likely mediated using the offspring BMI.
Collapse
Affiliation(s)
- Jiaxing Wen
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China; Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China
| | - Sumiya Aihemaitijiang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China; Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China
| | - Hongtian Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China; Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China
| | - Yubo Zhou
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China; Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China.
| | - Jianmeng Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China; Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
3
|
da Silva Rodrigues F, Jantsch J, de Farias Fraga G, Luiza de Camargo Milczarski V, Silva Dias V, Scheid C, de Oliveira Merib J, Giovernardi M, Padilha Guedes R. Cannabidiol improves maternal obesity-induced behavioral, neuroinflammatory and neurochemical dysfunctions in the juvenile offspring. Brain Behav Immun 2024; 119:301-316. [PMID: 38608740 DOI: 10.1016/j.bbi.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/25/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024] Open
Abstract
Maternal obesity is associated with an increased risk of psychiatric disorders such as anxiety, depression, schizophrenia and autism spectrum disorder in the offspring. While numerous studies focus on preventive measures targeting the mothers, only a limited number provide practical approaches for addressing the damages once they are already established. We have recently demonstrated the interplay between maternal obesity and treatment with cannabidiol (CBD) on hypothalamic inflammation and metabolic disturbances, however, little is known about this relationship on behavioral manifestations and neurochemical imbalances in other brain regions. Therefore, here we tested whether CBD treatment could mitigate anxiety-like and social behavioral alterations, as well as neurochemical disruptions in both male and female offspring of obese dams. Female Wistar rats were fed a cafeteria diet for 12 weeks prior to mating, and during gestation and lactation. Offspring received CBD (50 mg/kg) from weaning for 3 weeks. Behavioral tests assessed anxiety-like manifestations and social behavior, while neuroinflammatory and neurochemical markers were evaluated in the prefrontal cortex (PFC) and hippocampus. CBD treatment attenuated maternal obesity-induced anxiety-like and social behavioral alterations, followed by rescuing effects on imbalanced neurotransmitter and endocannabinoid concentrations and altered expression of glial markers, CB1, oxytocin and dopamine receptors, with important differences between sexes. Overall, the findings of this study provide insight into the signaling pathways for the therapeutic benefits of CBD on neuroinflammation and neurochemical imbalances caused by perinatal maternal obesity in the PFC and the hippocampus, which translates into the behavioral manifestations, highlighting the sexual dimorphism encompassing both the transgenerational effect of obesity and the endocannabinoid system.
Collapse
Affiliation(s)
- Fernanda da Silva Rodrigues
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jeferson Jantsch
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Gabriel de Farias Fraga
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Vitória Luiza de Camargo Milczarski
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Victor Silva Dias
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Camila Scheid
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Josias de Oliveira Merib
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Marcia Giovernardi
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170 Rio Grande do Sul, Brazil
| | - Renata Padilha Guedes
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170 Rio Grande do Sul, Brazil.
| |
Collapse
|
4
|
Patel D, Savvidou MD. Maternal Cardiac Function in Pregnancies with Metabolic Disorders. Eur Cardiol 2024; 19:e08. [PMID: 38983578 PMCID: PMC11231816 DOI: 10.15420/ecr.2023.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 01/17/2024] [Indexed: 07/11/2024] Open
Abstract
The obesity epidemic is growing and poses significant risks to pregnancy. Metabolic impairment can be associated with short- and long-term maternal and perinatal morbidity and mortality. The cardiovascular implications are known in those with metabolic disorder outside of pregnancy; however, little is known of the cardiac function in pregnancies complicated by obesity. Maternal cardiac adaptation plays a vital role in normal pregnancy and is known to be involved in the pathophysiology of adverse pregnancy outcomes. Bariatric surgery is the most successful treatment for sustainable weight loss and pre-pregnancy bariatric surgery can drastically change the maternal metabolic profile and pregnancy outcomes. In this review, we discuss the available evidence on maternal cardiac function in pregnancies affected by obesity and its associated consequences of gestational diabetes and hypertension (chronic and hypertensive disorders in pregnancy), as well as pregnancies following bariatric surgery.
Collapse
Affiliation(s)
- Deesha Patel
- Academic Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital, Department of Metabolism, Digestion and Reproduction, Imperial College London, UK
| | - Makrina D Savvidou
- Academic Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital, Department of Metabolism, Digestion and Reproduction, Imperial College London, UK
- Fetal Medicine Unit, Chelsea and Westminster Hospital London, UK
| |
Collapse
|
5
|
Ferreira HU, von Hafe M, Dias H, Gonçalves J, Belo S, Queirós J. Pregnancy After Bariatric Surgery-Experience from a Tertiary Center. Obes Surg 2024; 34:1432-1441. [PMID: 38467900 PMCID: PMC11031611 DOI: 10.1007/s11695-024-07147-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/13/2024]
Abstract
INTRODUCTION It is estimated that most people undergoing bariatric surgery are women of reproductive age; nonetheless, its effects on pregnancy outcomes are not yet fully understood. METHODS Retrospective observational study, conducted in a tertiary center in Portugal, included participants in two groups: (1) pregnant women with a history of bariatric surgery (n = 89) and (2) pregnant women with a BMI ≥ 35 kg/m2, without previous bariatric surgery (n = 176). Data was collected from the medical files. Multivariate analysis was conducted to adjust for confounders. RESULTS Pregnancy after bariatric surgery was associated with lower risk of gestational diabetes (15.7% vs. 30.1%, p = 0.002) and cesarean delivery (20.7% vs. 33.5%, p = 0.007), and a higher gestational weight gain (10.58 ± 9.95 vs. 7.33 ± 6.00 kg, p < 0.001). Participants in the bariatric surgery who experienced a gestational weight gain ≤ 10.0 kg had a higher risk of preterm delivery (16.7% vs. 2.5%, p = 0.031). No significant differences were found regarding hypertensive diseases of pregnancy between groups (4.5% vs 11.4%, p = 0.147). Pregnancy after bariatric surgery was associated with lower neonate weight percentile (34.24 ± 21.09 vs. 48.77 ± 27.94, p < 0.001), higher risk of fetal growth restriction (5.6% vs. 0.6%, p = 0.018), and lower risk of fetal macrosomia (0.0% vs. 7.5%, p = 0.005). There were no significant differences in the risk of SGA (12.5% vs. 7.0%, p = 0.127) or LGA neonates (2.3% vs. 6.4%, p = 0.069). CONCLUSION Pregnancy after bariatric surgery is associated with both risks and benefits, which should be considered by healthcare providers. Pregnancy after bariatric surgery requires individualized care, to ensure adequate gestational weight and avoid micronutrient deficiencies.
Collapse
Affiliation(s)
- Helena Urbano Ferreira
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.
- Instituto de Investigação e Inovação em Saúde (i3s), Faculdade de Medicina da Universidade Do Porto, Porto, Portugal.
| | - Madalena von Hafe
- Instituto de Investigação e Inovação em Saúde (i3s), Faculdade de Medicina da Universidade Do Porto, Porto, Portugal
- Serviço de Pediatria do Centro Hospitalar Universitário São João, Porto, Portugal
| | - Helena Dias
- Instituto de Investigação e Inovação em Saúde (i3s), Faculdade de Medicina da Universidade Do Porto, Porto, Portugal
- Serviço de Obstetrícia do Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Juliana Gonçalves
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3s), Faculdade de Medicina da Universidade Do Porto, Porto, Portugal
| | - Sandra Belo
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Joana Queirós
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| |
Collapse
|
6
|
Hauta-Alus HH, Rosendahl J, Holmlund-Suila EM, Valkama SM, Enlund-Cerullo M, Nurhonen M, Kajantie E, Mäkitie O, Andersson S. Low-grade inflammation from prenatal period to age 6-8 years in a Vitamin D trial. Pediatr Res 2024; 95:1578-1586. [PMID: 38225452 PMCID: PMC11126391 DOI: 10.1038/s41390-024-03019-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 11/29/2023] [Accepted: 12/26/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND Low-grade systemic inflammation measured as high sensitivity C-reactive protein (hs-CRP) has been associated with non-communicable disease risk. We assessed whether prenatal inflammation and early-childhood vitamin D are associated with inflammation until age 6-8. METHODS We analyzed blood hs-CRP and 25-hydroxy vitamin D [25(OH)D] in pregnancy, at birth from umbilical cord blood (UCB), from offspring at ages 1, 2, and 6-8 years in the Vitamin D Intervention in Infants (VIDI) study. VIDI was a randomized-controlled trial of vitamin D supplementation of 10 μg/day or 30 μg/day from age 2 weeks until 2 years in 975 infants recruited in 2013-14, with follow-up at age 6-8 in 2019-21 (n = 283). RESULTS Pregnancy hs-CRP was associated with UCB hs-CRP (r = 0.18, p < 0.001) but not independently with childhood hs-CRP (Estimate [95% CI] 0.04 [<-0.00, 0.09]). Higher UCB hs-CRP was associated independently with higher hs-CRP until 6-8 years (0.20 [0.12, 0.29]). Infant vitamin D dose had no effect on longitudinal hs-CRP (6-8 years, 0.11 [-0.04, 0.25]). Childhood 25(OH)D were associated positively with hs-CRP until age 6-8 (0.01 [>0.00, 0.01]). CONCLUSION Our results indicate that in children, inflammation, assessed by hs-CRP, persists from birth until 6-8 years. We observed positive associations between 25(OH)D and hs-CRP in vitamin D-sufficient children. IMPACT High sensitivity C-reactive protein (hs-CRP) concentrations tract from birth to age 8 years Our novel finding suggests a long-lasting pro-inflammatory phenotype in the child Higher vitamin D concentration - but not dose - is associated with higher childhood hs-CRP Chronic disease risk related to inflammation may in part originate from the prenatal period or early childhood Further studies are needed to investigate the effects of inflammation on long-term clinical health outcomes.
Collapse
Affiliation(s)
- Helena H Hauta-Alus
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Research Program for Clinical and Molecular Metabolism (CAMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Population Health unit, National Institute for Health and Welfare (THL), Helsinki, Finland.
- PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.
| | - Jenni Rosendahl
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism (CAMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Elisa M Holmlund-Suila
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism (CAMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Saara M Valkama
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism (CAMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Maria Enlund-Cerullo
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism (CAMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Markku Nurhonen
- Population Health unit, National Institute for Health and Welfare (THL), Helsinki, Finland
| | - Eero Kajantie
- Population Health unit, National Institute for Health and Welfare (THL), Helsinki, Finland
- PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Outi Mäkitie
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Department of Molecular Medicine and Surgery, Karolinska Institutet, and Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Sture Andersson
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
7
|
Wen J, Lv A, Aihemaitijiang S, Li H, Zhou Y, Liu J. The association of maternal gestational weight gain with cardiometabolic risk factors in offspring: a systematic review and meta-analysis. Nutr Rev 2024:nuae027. [PMID: 38607346 DOI: 10.1093/nutrit/nuae027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024] Open
Abstract
CONTEXT Gestational weight gain (GWG) is known to be a risk factor for offspring obesity, a precursor of cardiometabolic diseases. Accumulating studies have investigated the association of GWG with offspring cardiometabolic risk factors (CRFs), leading to inconsistent results. OBJECTIVE This study synthesized available data from cohort studies to examine the effects of GWG on offspring CRFs. DATA SOURCE Four electronic databases, including PubMed, Web of Science, Scopus, and Embase, were searched through May 2023. DATA EXTRACTION Cohort studies evaluating the association between GWG and CRFs (fat mass [FM], body fat percentage [BF%], waist circumference [WC], systolic blood pressure [SBP] and diastolic blood pressure, high-density-lipoprotein cholesterol [HDL-C] and low-density-lipoprotein cholesterol, triglyceride [TG], total cholesterol, fasting blood glucose, and fasting insulin levels) were included. Regression coefficients, means or mean differences with 95% confidence intervals [CIs], or standard deviations were extracted. DATA ANALYSIS Thirty-three cohort studies were included in the meta-analysis. Higher GWG (per increase of 1 kg) was associated with greater offspring FM (0.041 kg; 95% CI, 0.016 to 0.067), BF% (0.145%; 95% CI, 0.116 to 0.174), WC (0.154 cm; 95% CI, 0.036 to 0.272), SBP (0.040 mmHg; 95% CI, 0.010 to 0.070), and TG (0.004 mmol/L; 95% CI, 0.001 to 0.007), and with lower HDL-C (-0.002 mmol/L; 95% CI, -0.004 to 0.000). Consistently, excessive GWG was associated with higher offspring FM, BF%, WC, and insulin, and inadequate GWG was associated with lower BF%, low-density lipoprotein cholesterol, total cholesterol, and TG, compared with adequate GWG. Most associations went non-significant or attenuated with adjustment for offspring body mass index or FM. CONCLUSIONS Higher maternal GWG is associated with increased offspring adiposity, SBP, TG, and insulin and decreased HDL-C in offspring, warranting a need to control GWG and to screen for cardiometabolic abnormalities of offspring born to mothers with excessive GWG. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42023412098.
Collapse
Affiliation(s)
- Jiaxing Wen
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China
- Department of Epidemiology and Biostatistics, Peking University Health Science Center, Beijing, China
| | - Axing Lv
- School of Public Health, Peking University Health Science Center, Beijing, China
| | - Sumiya Aihemaitijiang
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China
- Department of Epidemiology and Biostatistics, Peking University Health Science Center, Beijing, China
| | - Hongtian Li
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China
- Department of Epidemiology and Biostatistics, Peking University Health Science Center, Beijing, China
| | - Yubo Zhou
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China
- Department of Epidemiology and Biostatistics, Peking University Health Science Center, Beijing, China
| | - Jianmeng Liu
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China
- Department of Epidemiology and Biostatistics, Peking University Health Science Center, Beijing, China
| |
Collapse
|
8
|
Laru J, Pinola P, Ojaniemi M, Korhonen E, Laikari L, Franks S, Piltonen TT, Tapanainen JS, Niinimäki M, Morin-Papunen L. Low testosterone at age 31 associates with maternal obesity and higher body mass index from childhood until age 46: A birth cohort study. Andrology 2024; 12:327-337. [PMID: 37424437 DOI: 10.1111/andr.13492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/28/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND Low testosterone (T) levels in men associate with increased risks of obesity, type 2 diabetes, metabolic syndrome, and cardiovascular diseases. However, most studies are cross-sectional with follow-up-time < 10 years, and data on early growth are limited. OBJECTIVE To compare prenatal factors and body mass index (BMI) development from birth to age 46 in relation to low T at age 31. MATERIALS AND METHODS Men with low T (T < 12.1 nmol/L, n = 132) and men with normal T at age 31 (n = 2561) were derived from the Northern Finland Birth Cohort 1966. Prenatal factors, longitudinal weight and height data from birth to age 14, and cross-sectional weight and height data at ages 31 and 46, and waist-hip-ratio (WHR) and T levels at age 31 were analyzed. Longitudinal modeling and timing of adiposity rebound (AR, second BMI rise at age 5-7 years) were calculated from fitted BMI curves. Results were adjusted for mother's pre-pregnancy BMI and smoking status, birth weight for gestational age, alcohol consumption, education level, smoking status, and WHR at age 31. RESULTS Neither gestational age nor birth weight was associated with low T at age 31; however, maternal obesity during gestation was more prevalent among men with low T (9.8% vs. 3.5%, adjusted aOR: 2.43 [1.19-4.98]). Men with low T had earlier AR (5.28 vs. 5.82, aOR: 0.73 [0.56-0.94]) and higher BMI (p < 0.001) from AR onward until age 46. Men with both early AR and low T had the highest BMI from AR onward. CONCLUSIONS In men, maternal obesity and early weight gain associate with lower T levels at age 31, independently of adulthood abdominal obesity. Given the well-known health risks related to obesity, and the rising prevalence of maternal obesity, the results of the present study emphasize the importance of preventing obesity that may also affect the later reproductive health of the offspring.
Collapse
Affiliation(s)
- Johanna Laru
- Department of Obstetrics and Gynecology, Oulu University Hospital, Wellbeing Services County of North Ostrobothnia, Oulu, Finland
- Medical Research Center, Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
| | - Pekka Pinola
- Department of Obstetrics and Gynecology, Oulu University Hospital, Wellbeing Services County of North Ostrobothnia, Oulu, Finland
- Medical Research Center, Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
| | - Marja Ojaniemi
- Medical Research Center, Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Department of Children and Adolescents, Oulu University Hospital, Wellbeing Services County of North Ostrobothnia Oulu, Oulu, Finland
| | - Elisa Korhonen
- Department of Obstetrics and Gynecology, Oulu University Hospital, Wellbeing Services County of North Ostrobothnia, Oulu, Finland
- Medical Research Center, Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
| | - Lotta Laikari
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Stephen Franks
- Institute of Reproductive and Developmental Biology, Imperial College London, London, UK
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, Oulu University Hospital, Wellbeing Services County of North Ostrobothnia, Oulu, Finland
- Medical Research Center, Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
| | - Juha S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics and Gynaecology, HFR - Cantonal Hospital of Fribourg and University of Fribourg, Fribourg, Switzerland
| | - Maarit Niinimäki
- Department of Obstetrics and Gynecology, Oulu University Hospital, Wellbeing Services County of North Ostrobothnia, Oulu, Finland
- Medical Research Center, Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
| | - Laure Morin-Papunen
- Department of Obstetrics and Gynecology, Oulu University Hospital, Wellbeing Services County of North Ostrobothnia, Oulu, Finland
- Medical Research Center, Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
9
|
Qi Y, Lv R, Zhao M, Huang Y, Zhang Y, Zhang H, Li Z, Jia P, Zhang H, Yang Z, Lai J, Song P, Yuan C. Parental adherence to healthy lifestyles in relation to the risk of obesity in offspring: A prospective cohort study in China. J Glob Health 2023; 13:04181. [PMID: 38115717 PMCID: PMC10731135 DOI: 10.7189/jogh.13.04181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023] Open
Abstract
Background While maternal adherence to a healthy lifestyle was shown to be associated with a lower risk of obesity in offspring, the potential role of overall parental lifestyles has not yet been explored. We aimed to address this gap by exploring whether parental adherence to an overall healthy lifestyle was associated with a lower risk of obesity in offspring. Methods We included 5881 children and adolescents aged 6-15 years at enrolment in the 2010, 2012, and 2014 waves of the China Family Panel Studies (CFPS) who were free of obesity and followed them until 2020. Parental healthy lifestyle score at study baseline was composed of five modifiable lifestyle factors (0-5; 1 for each): never smoking, non-habitual drinking, weekly exercise, modified dietary diversity score ≥5 points, and body mass index (BMI) of 18.5-23.9 kg/m2. We defined obesity according to the age- and gender-specific cutoffs by the BMI percentile curves for Chinese children aged 6-18 years. We used multivariable Cox proportional hazard models to examine the association between parental healthy lifestyle score (both as continuous and categorical variables) and risk of offspring obesity. Results Overall, 597 (10.2%) offspring developed obesity during a median follow-up of 6 years. Compared to the lowest tertile of parental healthy lifestyle score, participants in the highest tertile had a 42% (hazard ratio (HR) = 0.58; 95% confidence interval (CI) = 0.45-0.74) lower risk of obesity. Both maternal (HR = 0.75; 95% CI = 0.61-0.92) and paternal (HR = 0.73; 95% CI = 0.60-0.89) healthy lifestyle scores were associated with lower risks of obesity in offspring. For specific lifestyle factors, we observed beneficial associations for paternal diverse diet (HR = 0.73; 95% CI = 0.60-0.88) and healthy BMI (HR = 0.65; 95% CI = 0.55-0.78). Conclusions Adherence to an overall parental healthier lifestyle was associated with a lower risk of obesity in childhood and adolescence. This finding highlights the potential benefits of promoting a healthy lifestyle among parents for the primary prevention of offspring obesity.
Collapse
Affiliation(s)
- Ye Qi
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Rongxia Lv
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mengjia Zhao
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuhui Huang
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yaodan Zhang
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hangrui Zhang
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhihui Li
- Vanke School of Public Health, Tsinghua University, Beijing, China
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Peng Jia
- School of Resource and Environmental Sciences, Wuhan University, Wuhan, China
- International Institute of Spatial Lifecourse Health, Wuhan University, Wuhan, China
| | - Huanmei Zhang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhenyu Yang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jianqiang Lai
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Peige Song
- School of Public Health and Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Changzheng Yuan
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Costa SO, Chaves WF, Lopes PKF, Silva IM, Burguer B, Ignácio-Souza LM, Torsoni AS, Milanski M, Rodrigues HG, Desai M, Ross MG, Torsoni MA. Maternal consumption of a high-fat diet modulates the inflammatory response in their offspring, mediated by the M1 muscarinic receptor. Front Immunol 2023; 14:1273556. [PMID: 38193079 PMCID: PMC10773672 DOI: 10.3389/fimmu.2023.1273556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 11/27/2023] [Indexed: 01/10/2024] Open
Abstract
Introduction High-fat diet (HFD) consumption is associated with various metabolic disorders and diseases. Both pre-pregnancy and maternal obesity can have long-term consequences on offspring health. Furthermore, consuming an HFD in adulthood significantly increases the risk of obesity and metabolic disorders. However, an intriguing phenomenon known as the obesity paradox suggests that obesity may confer a protective effect on mortality outcomes in sepsis. In sepsis, activation of the cholinergic anti-inflammatory pathway (CAP) can help mitigate systemic inflammation. We employed a metabolic programming model to explore the relationship between maternal HFD consumption and offspring response to sepsis. Methods We fed female mice either a standard diet (SC) or an HFD during the pre-pregnancy, pregnancy, and lactation periods. Subsequently, we evaluated 28-day-old male offspring. Results Notably, we discovered that offspring from HFD-fed dams (HFD-O) exhibited a higher survival rate compared with offspring from SC-fed dams (SC-O). Importantly, inhibition of the m1 muscarinic acetylcholine receptor (m1mAChR), involved in the CAP, in the hypothalamus abolished this protection. The expression of m1mAChR in the hypothalamus was higher in HFD-O at different ages, peaking on day 28. Treatment with an m1mAChR agonist could modulate the inflammatory response in peripheral tissues. Specifically, CAP activation was greater in the liver of HFD-O following agonist treatment. Interestingly, lipopolysaccharide (LPS) challenge failed to induce a more inflammatory state in HFD-O, in contrast to SC-O, and agonist treatment had no additional effect. Analysis of spleen immune cells revealed a distinct phenotype in HFD-O, characterized by elevated levels of CD4+ lymphocytes rather than CD8+ lymphocytes. Moreover, basal Il17 messenger RNA (mRNA) levels were lower while Il22 mRNA levels were higher in HFD-O, and we observed the same pattern after LPS challenge. Discussion Further examination of myeloid cells isolated from bone marrow and allowed to differentiate showed that HFD-O macrophages displayed an anti-inflammatory phenotype. Additionally, treatment with the m1mAChR agonist contributed to reducing inflammatory marker levels in both groups. In summary, our findings demonstrate that HFD-O are protected against LPS-induced sepsis, and this protection is mediated by the central m1mAChR. Moreover, the inflammatory response in the liver, spleen, and bone marrow-differentiated macrophages is diminished. However, more extensive analysis is necessary to elucidate the specific mechanisms by which m1mAChR modulates the immune response during sepsis.
Collapse
Affiliation(s)
- Suleyma Oliveira Costa
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Wenicios Ferreira Chaves
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | | | - Iracema M. Silva
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Beatriz Burguer
- Laboratory of Nutrients and Tissue Repair, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Leticia M. Ignácio-Souza
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas, Limeira, 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, Limeira, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Marciane Milanski
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas, Limeira, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Hosana Gomes Rodrigues
- Laboratory of Nutrients and Tissue Repair, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Mina Desai
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles at Harbor-UCLA, Torrance, CA, United States
| | - Michael Glenn Ross
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles at Harbor-UCLA, Torrance, CA, United States
| | - Marcio Alberto Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas, Limeira, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| |
Collapse
|
11
|
Lecoutre S, Maqdasy S, Lambert M, Breton C. The Impact of Maternal Obesity on Adipose Progenitor Cells. Biomedicines 2023; 11:3252. [PMID: 38137473 PMCID: PMC10741630 DOI: 10.3390/biomedicines11123252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
The concept of Developmental Origin of Health and Disease (DOHaD) postulates that adult-onset metabolic disorders may originate from suboptimal conditions during critical embryonic and fetal programming windows. In particular, nutritional disturbance during key developmental stages may program the set point of adiposity and its associated metabolic diseases later in life. Numerous studies in mammals have reported that maternal obesity and the resulting accelerated growth in neonates may affect adipocyte development, resulting in persistent alterations in adipose tissue plasticity (i.e., adipocyte proliferation and storage) and adipocyte function (i.e., insulin resistance, impaired adipokine secretion, reduced thermogenesis, and higher inflammation) in a sex- and depot-specific manner. Over recent years, adipose progenitor cells (APCs) have been shown to play a crucial role in adipose tissue plasticity, essential for its development, maintenance, and expansion. In this review, we aim to provide insights into the developmental timeline of lineage commitment and differentiation of APCs and their role in predisposing individuals to obesity and metabolic diseases. We present data supporting the possible implication of dysregulated APCs and aberrant perinatal adipogenesis through epigenetic mechanisms as a primary mechanism responsible for long-lasting adipose tissue dysfunction in offspring born to obese mothers.
Collapse
Affiliation(s)
- Simon Lecoutre
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, F-75013 Paris, France
| | - Salwan Maqdasy
- Department of Medicine (H7), Karolinska Institutet Hospital, C2-94, 14186 Stockholm, Sweden;
| | - Mélanie Lambert
- U978 Institut National de la Santé et de la Recherche Médicale, F-93022 Bobigny, France;
- Université Sorbonne Paris Nord, Alliance Sorbonne Paris Cité, Labex Inflamex, F-93000 Bobigny, France
| | - Christophe Breton
- Maternal Malnutrition and Programming of Metabolic Diseases, Université de Lille, EA4489, F-59000 Lille, France
- U1283-UMR8199-EGID, Université de Lille, INSERM, CNRS, CHU Lille, Institut Pasteur de Lille, F-59000 Lille, France
| |
Collapse
|
12
|
Kim K, Varghese M, Sun H, Abrishami S, Bowers E, Bridges D, Meijer JL, Singer K, Gregg B. The Influence of Maternal High Fat Diet During Lactation on Offspring Hematopoietic Priming. Endocrinology 2023; 165:bqad182. [PMID: 38048597 PMCID: PMC11032250 DOI: 10.1210/endocr/bqad182] [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: 07/06/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/06/2023]
Abstract
Obesity and metabolic diseases are rising among women of reproductive age, increasing offspring metabolic risk. Maternal nutritional interventions during lactation present an opportunity to modify offspring outcomes. We previously demonstrated in mice that adult male offspring have metabolic impairments and increased adipose tissue macrophages (ATM) when dams are fed high fat diet (HFD) during the postnatal lactation window (HFD PN). We sought to understand the effect of HFD during lactation on early-life inflammation. HFD PN offspring were evaluated at postnatal day 16 to 19 for tissue weight and gene expression. Profiling of adipose tissue and bone marrow immune cells was conducted through lipidomics, in vitro myeloid colony forming unit assays, and flow cytometry. HFD PN mice had more visceral gonadal white adipose tissue (GWAT) and subcutaneous fat. Adipose tissue RNA sequencing demonstrated enrichment of inflammation, chemotaxis, and fatty acid metabolism and concordant changes in GWAT lipidomics. Bone marrow (BM) of both HFD PN male and female offspring had increased monocytes (CD45+Ly6G-CD11b+CD115+) and B cells (CD45+Ly6G-CD11b-CD19+). Similarly, serum from HFD PN offspring enhanced in vitro BM myeloid colonies in a toll-like receptor 4-dependent manner. We identified that male HFD PN offspring had increased GWAT pro-inflammatory CD11c+ ATMs (CD45+CD64+). Maternal exposure to HFD alters milk lipids enhancing adiposity and myeloid inflammation even in early life. Future studies are needed to understand the mechanisms driving this pro-inflammatory state of both BM and ATMs, the causes of the sexually dimorphic phenotypes, and the feasibility of intervening in this window to improve metabolic health.
Collapse
Affiliation(s)
- Katherine Kim
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mita Varghese
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Haijing Sun
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Simin Abrishami
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Emily Bowers
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dave Bridges
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Jennifer L Meijer
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, USA
- Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| | - Kanakadurga Singer
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Brigid Gregg
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| |
Collapse
|
13
|
Jodah R, Arunamata A, Kipps AK, Chen S, Stauffer KJ, Selamet Tierney ES. Maternal Diabetes and Cardiovascular Health in the Offspring. Pediatr Cardiol 2023:10.1007/s00246-023-03333-4. [PMID: 37930377 DOI: 10.1007/s00246-023-03333-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/19/2023] [Indexed: 11/07/2023]
Abstract
Pulse wave velocity (PWV) has been explored to predict cardiovascular health in adults. Less is known about neonatal PWV. We evaluated the association between arterial stiffness of neonates of mothers (NoM) with diabetes and childhood health. Neonatal brachial-femoral PWV (bfPWV) was measured after birth and neonates followed for a median of 5.2 years [1 month-6.6 years]. 36 pregnant women with pregestational diabetes mellitus PGDM (n = 12), gestational diabetes mellitus (GDM) (n = 13), and controls (n = 11) were enrolled. Neonates were similar in weight, gestational age, and delivery mode. 26 neonates had follow-up data including weight, height and blood pressure. More mothers with PGDM had poor glycemic control compared to mothers with GDM (83% vs. 8%; p = 0.0002). PWV was higher in NoM with PGDM than controls (3.4 ± 0.5 vs. 2.6 ± 0.8 m/s; p = 0.04). At follow-up, children of mothers with diabetes (n = 16) had higher weight percentile (78.5 ± 27.9 vs 49.5 ± 34.6%; p = 0.02) and diastolic blood pressure (DBP) (68 ± 13.6 vs 57.3 ± 4.3 mmHg; p = 0.01) than controls (n = 10). No correlation emerged between neonatal PWV and childhood body mass index (BMI) or maternal HbA1c. Results suggest maternal diabetes affect neonatal arterial stiffness and childhood blood pressure; however, the mechanism is unclear. The long-term implications of these findings warrant further investigation.
Collapse
Affiliation(s)
- Riasoya Jodah
- Stanford University School of Medicine, Palo Alto, CA, USA
| | - Alisa Arunamata
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford Children's Health (Lucile Packard Children's Hospital), Palo Alto, CA, USA
| | - Alaina K Kipps
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford Children's Health (Lucile Packard Children's Hospital), Palo Alto, CA, USA
| | - Sharon Chen
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford Children's Health (Lucile Packard Children's Hospital), Palo Alto, CA, USA
| | - Katie Jo Stauffer
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford Children's Health (Lucile Packard Children's Hospital), Palo Alto, CA, USA
| | - Elif Seda Selamet Tierney
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford Children's Health (Lucile Packard Children's Hospital), Palo Alto, CA, USA.
| |
Collapse
|
14
|
Mort EJ, Heritage S, Jones S, Fowden AL, Camm EJ. Sex-Specific Effects of a Maternal Obesogenic Diet High in Fat and Sugar on Offspring Adiposity, Growth, and Behavior. Nutrients 2023; 15:4594. [PMID: 37960247 PMCID: PMC10648016 DOI: 10.3390/nu15214594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
With rising rates of human obesity, this study aimed to determine the relationship between maternal diet-induced obesity, offspring morphometrics, and behavior in mice. Pregnant and lactating female mice fed a diet high in fat and sugar (HFHS) commonly consumed by human populations showed decreased food, calorie, and protein intake but increased adiposity at the expense of lean mass. The pre-weaning body weight of the HFHS offspring was reduced for the first postnatal week but not thereafter, with HFHS female offspring having higher body weights by weaning due to continuing higher fractional growth rates. Post-weaning, there were minor differences in offspring food and protein intake. Maternal diet, however, affected fractional growth rate and total body fat content of male but not female HFHS offspring. The maternal diet did not affect the offspring's locomotor activity or social behavior in either sex. Both the male and female HFHS offspring displayed reduced anxiety-related behaviors, with sex differences in particular aspects of the elevated plus maze task. In the novel object recognition task, performance was impaired in the male but not female HFHS offspring. Collectively, the findings demonstrate that maternal obesity alters the growth, adiposity, and behavior of male and female offspring, with sex-specific differences.
Collapse
Affiliation(s)
- Emily J. Mort
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Sophie Heritage
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Susan Jones
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Abigail L. Fowden
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Emily J. Camm
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia
| |
Collapse
|
15
|
Santos da Silva LL, Malta MB, Lourenço BH, Mosquera PS, de Araújo Damasceno AA, Neves PAR, Cardoso MA. Maternal pre-pregnancy body mass index, gestational weight gain and child weight during the first 2 years of life in an Amazonian birth cohort. J Hum Nutr Diet 2023; 36:1327-1338. [PMID: 36733263 DOI: 10.1111/jhn.13148] [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: 11/22/2022] [Accepted: 01/30/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND In socially vulnerable populations, evidence is needed regarding the role of maternal nutritional status on child weight during the first 2 years of life. OBJECTIVES We aimed to assess the association of pre-pregnancy body mass index (BMI) and gestational weight gain (GWG) with offspring BMI-for-age z-scores (BAZs) during the first 2 years of life. METHODS A population-based birth cohort study was conducted with 900 mother-child pairs. Pre-pregnancy weight and weight at delivery were collected from medical records, and anthropometric data were measured at birth and at 6-month, 1-year and 2-year follow-up visits. Linear regression and linear mixed-effect models assessed associations with pre-pregnancy BMI, GWG and BAZ during the first 2 years of life. RESULTS Pre-pregnancy overweight and obesity and excessive GWG were positively associated with BAZ at birth and at 1- and 2-year follow-up visits. There were no significant additional BAZ changes per year based on the exposures up to age 2 years. CONCLUSIONS Elevated maternal pre-pregnancy BMI and GWG were associated with a child's higher BAZ at birth, and these differences remained constant throughout the first 2 years of life in Amazonian children. These findings highlight the importance of promoting adequate maternal weight before pregnancy and during prenatal care also in socially vulnerable populations.
Collapse
Affiliation(s)
- Lara Lívia Santos da Silva
- Department of Nutrition, University of São Paulo, São Paulo, Brazil
- Department of Collective Health, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil
| | | | | | | | - Ana Alice de Araújo Damasceno
- Postgraduate Program in Public Health, University of São Paulo, São Paulo, Brazil
- Federal University of Acre, Cruzeiro do Sul, Brazil
| | - Paulo Augusto Ribeiro Neves
- Centre for Global Child Health, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
16
|
Fitch J, Dorney E, Tracy M, Black KI. Acceptability and usability of 'One Key Question'® in Australian primary health care. Aust J Prim Health 2023; 29:268-275. [PMID: 36346949 DOI: 10.1071/py22112] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/17/2022] [Indexed: 07/20/2023]
Abstract
BACKGROUND Around one-third of pregnancies in women attending antenatal care are unintended. This means a substantial number of women enter pregnancy without optimising their health prior to conception. Primary care practitioners are uniquely placed to counsel women about how to plan for pregnancy and about how to avoid unintended conception. The One Key Question® (OKQ®) tool facilitates a discussion of pregnancy intention and opens up subsequent discussions regarding preconception or contraception care. This study aimed to assess the acceptability and usability of the OKQ® tool in the Australian primary care setting. METHODS We undertook a pilot study consisting of quantitative and qualitative componentsacross two general practice settings in Sydney, New South Wales, Australia. We documented women's responses to being asked the OKQ® as part of their consultation. We collected data on the characteristics of the participating GPs and their experience of using the OKQ® tool and conducted semi-structured interviews with all participating GPs. RESULTS Fifty-six patients were asked the OKQ®, with the majority stating they were happy to be asked about their reproductive choices and felt it was relevant to their general health. The 10 participating GPs felt the OKQ® was easy to use and although 62.5% reported it extended the consultation time, the medium time taken was 2min. GPs felt framing the OKQ® helped introduce pregnancy intention discussions into a consultation. CONCLUSIONS The OKQ® is acceptable to patients and easy for GPs to use. This tool facilitates a proactive and routine discussion to enhance the delivery of preconception care and contraceptive counselling.
Collapse
Affiliation(s)
- Jessica Fitch
- Speciality of General Practice, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Edwina Dorney
- Faculty of Medicine and Health, Central Clinical School, The Tavern, Medical Foundation Building K25, The University of Sydney, Sydney, NSW 2006, Australia
| | - Marguerite Tracy
- Speciality of General Practice, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Kirsten I Black
- Faculty of Medicine and Health, Central Clinical School, The Tavern, Medical Foundation Building K25, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
17
|
Pavela G, Yi N, Mestre L, Xun P, Allison DB. Birth weight moderates the association between obesity and mortality rate. Ann Epidemiol 2023; 82:26-32. [PMID: 37015307 PMCID: PMC10463462 DOI: 10.1016/j.annepidem.2023.03.010] [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: 07/13/2022] [Revised: 02/14/2023] [Accepted: 03/29/2023] [Indexed: 04/06/2023]
Abstract
PURPOSE The strength of the association between obesity and mortality rate (MR) varies by body mass index (BMI) and sociodemographic groups. We test the hypothesis that the association between obesity and MR varies, in part, due to the moderating effect of parental BMI and birth weight. METHODS Data come from the 1958 National Child Development Study, an ongoing longitudinal dataset initiated in 1958 with baseline measures of birth weight from 18,059 infants born in Great Britain over 1 week. We tested whether the association between BMI and MR was moderated by parental BMI and birth weight using generalized additive proportional hazards models. RESULTS The association between adult BMI and MR was moderated by birth weight and maternal BMI, such that the association between BMI and MR was weaker among individuals with a higher birth weight (P = .0148) and stronger among individuals born to mothers with a higher BMI (P = .032). At any given level of BMI approximately greater than 25, individuals with low birth weight or born to mothers with a higher BMI, had a higher MR. Paternal BMI did not significantly modify the relationship between BMI and MR (P = .5168). CONCLUSIONS Results suggest that the relationship between obesity and MR is modified by birth weight and maternal BMI.
Collapse
Affiliation(s)
- Gregory Pavela
- School of Public Health, University of Alabama at Birmingham, Birmingham.
| | - Nengjun Yi
- School of Public Health, University of Alabama at Birmingham, Birmingham
| | - Luis Mestre
- School of Public Health, Indiana University Bloomington, Bloomington
| | | | - David B Allison
- School of Public Health, Indiana University Bloomington, Bloomington
| |
Collapse
|
18
|
Ahmed N, Kassis A, Malone J, Yang J, Zamzami E, Lin AH, Gordon SM, Gong M, Bardo M, Dalmasso C, Loria AS. Prenatal Morphine Exposure Increases Cardiovascular Disease Risk and Programs Neurogenic Hypertension in the Adult Offspring. Hypertension 2023; 80:1283-1296. [PMID: 37042247 PMCID: PMC10274123 DOI: 10.1161/hypertensionaha.122.20262] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 03/20/2023] [Indexed: 04/13/2023]
Abstract
BACKGROUND The opioid overdose and opioid use disorder epidemics are concomitant with increased metabolic and CVD risk. Although opioid use disorder causes adverse pregnancy outcomes, the offspring's cardiovascular health is understudied. We hypothesized that offspring exposed to in utero morphine exposure (IUME) would show increased CVD risk factors and endogenous opioid system dysregulation. METHODS Sprague Dawley dams were treated with saline (vehicle, n=10) or escalating doses of morphine (5-20 mg/kg per day, SC, n=10) during gestation. Cardiovascular and metabolic parameters were assessed in adult offspring. RESULTS Litter size and pups' birth weight were not different in response to IUME. Female and male IUME offspring showed reduced body length at birth (P<0.05) and body weight from weeks 1 to 3 of life (P<0.05), followed by a catch-up growth effect. By week 16, female and male IUME rats showed reduced tibia length (P<0.05) and fat mass. IUME increases the mean arterial pressure and the depressor response to mecamylamine (5 mg/kg per day, IP) induced by IUME were abolished by a chronic treatment with an alpha-adrenergic receptor blocker (prazosin; 1 mg/kg per day, IP). Although circulating levels of angiotensin peptides were similar between groups, IUME exacerbated maximal ex vivo Ang (angiotensin) II-induced vasoconstriction (P<0.05) and induced endothelial dysfunction in a sex-specific manner (P<0.05). Proenkephalin, an endogenous opioid peptide that lowers blood pressure and sympathetic-mediated vasoconstriction, showed reduced mRNA expression in the heart, aorta, and kidneys from morphine versus vehicle group (P<0.05). CONCLUSIONS Among the effects of IUME, neurogenic hypertension, vascular dysfunction, and metabolic dysfunction could be associated with the dysregulation of the endogenous opioid system.
Collapse
Affiliation(s)
- Nermin Ahmed
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Alana Kassis
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Jena Malone
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Jodie Yang
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Esraa Zamzami
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - An-Hsuan Lin
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Scott M. Gordon
- SAHA Cardiovascular Center, University of Kentucky, Lexington, KY 40536
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Ming Gong
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Michael Bardo
- Department of Psychology, College of Arts and Sciences, University of Kentucky, Lexington, KY 40536
| | - Carolina Dalmasso
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Analia S. Loria
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
- SAHA Cardiovascular Center, University of Kentucky, Lexington, KY 40536
| |
Collapse
|
19
|
Naomi R, Rusli RNM, Teoh SH, Bahari H, Zakaria ZA. Remodulation Effect of Elateriospermum tapos Yoghurt on Metabolic Profile of Maternal Obesity Induced Cognitive Dysfunction and Anxiety-like Behavior in Female Offspring-An In Vivo Trial on Sprague Dawley Rats. Foods 2023; 12:foods12081613. [PMID: 37107408 PMCID: PMC10137489 DOI: 10.3390/foods12081613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Pre-pregnancy weight gain induces dysregulation in the metabolic profile of the offspring, thereby serving as a key factor for cognitive decline and anxiety status in the offspring. However, early probiotic supplementation during the gestational period is linked with improved metabolic health. At the same time, a natural plant known as Elateriospermum tapos (E. tapos) is proven to improve cognition and modulate the stress hormone due to its high concentration of flavonoids. However, the effects of medicinal plant integrated probiotics in F1 generations warrants further investigation. Thus, this study aimed to study the effect of E. tapos yoghurt on the maternal obesity induced cognitive dysfunction and anxiety in female offspring. In this study, female Sprague Dawley rats were fed with normal chow (n = 8) or high fat diet (n = 40) across pre-pregnancy, gestation, and weaning. The treatment with different concentrations of E. tapos yoghurt (5, 50, and 500 mg/kg/day) were initiated in the obese dams upon post coitum day 0 up to postnatal day 21 (PND 21). Female offspring were weaned on PND 21 and body mass index, waist circumference, lee index, behavior, metabolic parameter, and antioxidant status were analyzed. The result shows that the female offspring of the 500 mg/kg E. tapos yoghurt supplemented group shows a decreased level of insulin, fasting blood glucose, cholesterol, triglycerides, LDL, low fat tissue mass with a high level of HDL, and an increased level of antioxidant status in the hypothalamus. The behavioral assessment proves that the female offspring of the 500 mg/kg E. tapos yoghurt supplemented group exhibits a high recognition index on novel object/place with low anxiety-like behavior in an open field test. In conclusion, our data signify the beneficial effect of early intervention in obese dams on the transgenerational impact on female offspring's metabolic profile, cognitive performance, and anxiety-like behavior.
Collapse
Affiliation(s)
- Ruth Naomi
- Borneo Research on Algesia, Inflammation and Neurodegeneration (BRAIN) Group, Faculty of Medicine and Health Sciences, Sabah Universiti Malaysia, Kota Kinabalu 88400, Malaysia
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Rusydatul Nabila Mahmad Rusli
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Soo Huat Teoh
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang 13200, Malaysia
| | - Hasnah Bahari
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Zainul Amiruddin Zakaria
- Borneo Research on Algesia, Inflammation and Neurodegeneration (BRAIN) Group, Faculty of Medicine and Health Sciences, Sabah Universiti Malaysia, Kota Kinabalu 88400, Malaysia
| |
Collapse
|
20
|
High-fat diet during pregnancy lowers fetal weight and has a long-lasting adverse effect on brown adipose tissue in the offspring. J Dev Orig Health Dis 2023; 14:261-271. [PMID: 36189641 DOI: 10.1017/s2040174422000551] [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: 11/06/2022]
Abstract
Maternal obesity and malnutrition during gestation and lactation have been recognized to increase the risk of obesity and metabolic disorders in the offspring across their lifespan. However, the gestational period during which malnutrition exerts a decisive effect is unclear. Brown adipose tissue (BAT) plays a critical role in energy metabolism owing to its high efficiency in oxidizing glucose and fatty acids. This study aimed to determine the impact of maternal high-fat diet (HFD) consumption only during pregnancy on BAT and energy metabolism in offspring mice. Dams were fed an HFD or a normal chow diet from embryonic day 2.5. HFD consumption during pregnancy induced glucose intolerance and hypertension in dams. In the offspring of HFD-fed dams, maternal HFD lowered fetal weight without affecting placental weight, whereas HFD consumption after birth exacerbated oxygen consumption and cold-induced thermogenesis at 12 months of age, accompanied by increased lipid droplet size in BAT. These data demonstrate that HFD consumption only during pregnancy exerts a long-lasting effect on BAT. Collectively, these findings indicate the importance of nutrition during pregnancy with respect to the energy metabolism of the offspring, and pregnant women should thus ensure proper nutrition during pregnancy to ensure normal energy metabolism in the offspring.
Collapse
|
21
|
Rodrigues FDS, Jantsch J, Fraga GDF, Dias VS, Eller S, De Oliveira TF, Giovenardi M, Guedes RP. Cannabidiol treatment improves metabolic profile and decreases hypothalamic inflammation caused by maternal obesity. Front Nutr 2023; 10:1150189. [PMID: 36969815 PMCID: PMC10033544 DOI: 10.3389/fnut.2023.1150189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/24/2023] [Indexed: 03/11/2023] Open
Abstract
IntroductionThe implications of maternal overnutrition on offspring metabolic and neuroimmune development are well-known. Increasing evidence now suggests that maternal obesity and poor dietary habits during pregnancy and lactation can increase the risk of central and peripheral metabolic dysregulation in the offspring, but the mechanisms are not sufficiently established. Furthermore, despite many studies addressing preventive measures targeted at the mother, very few propose practical approaches to treat the damages when they are already installed.MethodsHere we investigated the potential of cannabidiol (CBD) treatment to attenuate the effects of maternal obesity induced by a cafeteria diet on hypothalamic inflammation and the peripheral metabolic profile of the offspring in Wistar rats.ResultsWe have observed that maternal obesity induced a range of metabolic imbalances in the offspring in a sex-dependant manner, with higher deposition of visceral white adipose tissue, increased plasma fasting glucose and lipopolysaccharides (LPS) levels in both sexes, but the increase in serum cholesterol and triglycerides only occurred in females, while the increase in plasma insulin and the homeostatic model assessment index (HOMA-IR) was only observed in male offspring. We also found an overexpression of the pro-inflammatory cytokines tumor necrosis factor-alpha (TNFα), interleukin (IL) 6, and interleukin (IL) 1β in the hypothalamus, a trademark of neuroinflammation. Interestingly, the expression of GFAP, a marker for astrogliosis, was reduced in the offspring of obese mothers, indicating an adaptive mechanism to in utero neuroinflammation. Treatment with 50 mg/kg CBD oil by oral gavage was able to reduce white adipose tissue and revert insulin resistance in males, reduce plasma triglycerides in females, and attenuate plasma LPS levels and overexpression of TNFα and IL6 in the hypothalamus of both sexes.DiscussionTogether, these results indicate an intricate interplay between peripheral and central counterparts in both the pathogenicity of maternal obesity and the therapeutic effects of CBD. In this context, the impairment of internal hypothalamic circuitry caused by neuroinflammation runs in tandem with the disruptions of important metabolic processes, which can be attenuated by CBD treatment in both ends.
Collapse
Affiliation(s)
- Fernanda da Silva Rodrigues
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Jeferson Jantsch
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Gabriel de Farias Fraga
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Victor Silva Dias
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Sarah Eller
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Tiago Franco De Oliveira
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Márcia Giovenardi
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Renata Padilha Guedes
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
- *Correspondence: Renata Padilha Guedes,
| |
Collapse
|
22
|
Collin DF, Pulvera R, Hamad R. The effect of the 2009 revised U.S. guidelines for gestational weight gain on maternal and infant health: a quasi-experimental study. BMC Pregnancy Childbirth 2023; 23:118. [PMID: 36803304 PMCID: PMC9936770 DOI: 10.1186/s12884-023-05425-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/03/2023] [Indexed: 02/19/2023] Open
Abstract
BACKGROUND Excess gestational weight gain (GWG) has adverse short- and long-term effects on the health of mothers and infants. In 2009, the US Institute of Medicine revised its guidelines for GWG and reduced the recommended GWG for women who are obese. There is limited evidence on whether these revised guidelines affected GWG and downstream maternal and infant outcomes. METHODS We used data from the 2004-2019 waves of the Pregnancy Risk Assessment Monitoring System, a serial cross-sectional national dataset including over 20 states. We conducted a quasi-experimental difference-in-differences analysis to assess pre/post changes in maternal and infant outcomes among women who were obese, while "differencing out" the pre/post changes among a control group of women who were overweight. Maternal outcomes included GWG and gestational diabetes; infant outcomes included preterm birth (PTB), low birthweight (LBW), and very low birthweight (VLBW). Analysis began in March 2021. RESULTS There was no association between the revised guidelines and GWG or gestational diabetes. The revised guidelines were associated with reduced PTB (- 1.19% points, 95%CI: - 1.86, - 0.52), LBW (- 1.38% points 95%CI: - 2.07, - 0.70), and VLBW (- 1.30% points, 95%CI: - 1.68, - 0.92). Results were robust to several sensitivity analyses. CONCLUSION The revised 2009 GWG guidelines were not associated with changes in GWG or gestational diabetes but were associated with improvements in infant birth outcomes. These findings will help inform further programs and policies aimed at improving maternal and infant health by addressing weight gain in pregnancy.
Collapse
Affiliation(s)
- Daniel F Collin
- Philip R. Lee Institute for Health Policy Studies, University of California San Francisco, 995 Potrero Avenue, Building 80, Ward 83, San Francisco, CA, 94110, USA
| | - Richard Pulvera
- School of Public Health, University of California Berkeley, Berkeley, CA, USA
| | - Rita Hamad
- Philip R. Lee Institute for Health Policy Studies, University of California San Francisco, 995 Potrero Avenue, Building 80, Ward 83, San Francisco, CA, 94110, USA.
- Department of Family & Community Medicine, University of California San Francisco, 995 Potrero Avenue, Building 80, Ward 83, San Francisco, CA, 94110, USA.
| |
Collapse
|
23
|
Abruzzese GA, Arbocco FCV, Ferrer MJ, Silva AF, Motta AB. Role of Hormones During Gestation and Early Development: Pathways Involved in Developmental Programming. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:31-70. [PMID: 37466768 DOI: 10.1007/978-3-031-32554-0_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Accumulating evidence suggests that an altered maternal milieu and environmental insults during the intrauterine and perinatal periods of life affect the developing organism, leading to detrimental long-term outcomes and often to adult pathologies through programming effects. Hormones, together with growth factors, play critical roles in the regulation of maternal-fetal and maternal-neonate interfaces, and alterations in any of them may lead to programming effects on the developing organism. In this chapter, we will review the role of sex steroids, thyroid hormones, and insulin-like growth factors, as crucial factors involved in physiological processes during pregnancy and lactation, and their role in developmental programming effects during fetal and early neonatal life. Also, we will consider epidemiological evidence and data from animal models of altered maternal hormonal environments and focus on the role of different tissues in the establishment of maternal and fetus/infant interaction. Finally, we will identify unresolved questions and discuss potential future research directions.
Collapse
Affiliation(s)
- Giselle Adriana Abruzzese
- Laboratorio de Fisio-patología ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Fiorella Campo Verde Arbocco
- Laboratorio de Hormonas y Biología del Cáncer, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, Mendoza, Argentina
- Laboratorio de Reproducción y Lactancia, IMBECU, CONICET, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad de Mendoza, Mendoza, Argentina
| | - María José Ferrer
- Laboratorio de Fisio-patología ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Aimé Florencia Silva
- Laboratorio de Fisio-patología ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Alicia Beatriz Motta
- Laboratorio de Fisio-patología ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| |
Collapse
|
24
|
Schade R, Song L, Cordner ZA, Ding H, Peterson DA, Moran TH, Tamashiro KL, Serre CBDL. Rat offspring's microbiota composition is predominantly shaped by the postnatal maternal diet rather than prenatal diet. Physiol Behav 2023; 258:113987. [PMID: 36198343 PMCID: PMC10088501 DOI: 10.1016/j.physbeh.2022.113987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 01/26/2023]
Abstract
This study assessed the impact of maternal diet during pregnancy versus lactation on offspring gut microbiota. Sprague-Dawley dams were fed high fat (HF) or Chow diets during pregnancy, and their male offspring were raised by a different dam consuming the same or opposite diet (Chow-Chow, Chow-HF, HF-Chow, and HF-HF). Microbiota analysis showed that maternal lactation diet, rather than pregnancy diet, determined offspring microbiota profiles at weaning. Increased abundances of Turicibacter, Staphylococcus , and Ruminococcus were characteristic of chow lactation groups. Lactococcus , Streptococcus , and Parabacteroides were characteristic of HF lactation groups and positively correlated with offspring body weight.
Collapse
Affiliation(s)
- Ruth Schade
- Department of Nutritional Sciences, University of Georgia, Athens, GA, U.S.A; Current address: Department of Microbiology and Immunology, Stanford University, Palo Alto, CA, U.S.A
| | - Lin Song
- Departments of Psychiatry & Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zachary A Cordner
- Departments of Psychiatry & Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A
| | - Hua Ding
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A
| | - Daniel A Peterson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A
| | - Timothy H Moran
- Departments of Psychiatry & Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A
| | - Kellie L Tamashiro
- Departments of Psychiatry & Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A.
| | | |
Collapse
|
25
|
Mankouski A, Miller TA, Dodson RB, Yu B, Yang Y, Liu J, Machin DR, Donato AJ, McKnight RA, Zinkhan EK. Large artery stiffening and mortality in a rat model of early vascular remodeling induced by intrauterine growth restriction and a high-fat diet. Physiol Rep 2022; 10:e15518. [PMID: 36461654 PMCID: PMC9718947 DOI: 10.14814/phy2.15518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/02/2022] [Indexed: 05/31/2023] Open
Abstract
Intrauterine growth restriction (IUGR) and exposure to a high-fat diet (HFD) independently increase the risk of cardiovascular disease (CVD) and hyperlipidemia. In our previous studies, IUGR increased blood pressure and promoted vascular remodeling and stiffness in early life, a finding that persisted and was augmented by a maternal HFD through postnatal day (PND) 60. The impact of these findings with aging and the development of hyperlipidemia and atherosclerosis remain unknown. We hypothesized that the previously noted impact of IUGR on hypertension, vascular remodeling, and hyperlipidemia would persist. Adult female rats were fed either a regular diet (RD) or high fat diet (HFD) prior to conception through lactation. IUGR was induced by uterine artery ligation. Offspring were weaned to either RD or HFD through PND 365. For both control (C) and IUGR (I) and rats, this resulted in the following six groups per sex: offspring from RD dams weaned to an RD (CRR and IRR), or offspring from HFD dams weaned to either an RD (CHR and IHR) or to an HFD (CHH and IHH). IHH male and female rats had increased large artery stiffness, a suggestion of fatty streaks in the aorta, and persistent decreased elastin and increased collagen in the aorta and carotid arteries. Post-weaning HFD intake increased blood lipids regardless of IUGR status. IUGR increased HFD-induced mortality. We speculate that HFD-induced risk of CVD and mortality is potentiated by developmental programming of the ECM.
Collapse
Affiliation(s)
| | - Thomas A. Miller
- Department of PediatricsUniversity of UtahSalt Lake CityUtahUSA
- Division of Pediatric CardiologyMaine Medical CenterPortlandMaineUSA
| | - R. Blair Dodson
- Departments of Surgery and BioengineeringThe Pediatric Heart Lung Center and the Laboratory for Fetal and Regenerative Biology and the University of Colorado at Denver Anschutz Medical CampusAuroraColoradoUSA
| | - Baifeng Yu
- Department of PediatricsUniversity of UtahSalt Lake CityUtahUSA
| | - Yueqin Yang
- Department of PediatricsUniversity of UtahSalt Lake CityUtahUSA
| | - Jingtong Liu
- Department of PediatricsUniversity of UtahSalt Lake CityUtahUSA
| | - Daniel R. Machin
- Internal MedicineUniversity of UtahSalt Lake CityUtahUSA
- GRECCVA Medical CenterSalt Lake CityUtahUSA
- Florida State UniversityDepartment of Nutrition and Integrative PhysiologyTallahasseeFloridaUSA
| | - Anthony J. Donato
- Internal MedicineUniversity of UtahSalt Lake CityUtahUSA
- GRECCVA Medical CenterSalt Lake CityUtahUSA
- Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
- BiochemistryUniversity of UtahSalt Lake CityUtahUSA
| | | | - Erin K. Zinkhan
- Department of PediatricsUniversity of UtahSalt Lake CityUtahUSA
| |
Collapse
|
26
|
Kulhanek D, Abrahante Llorens JE, Buckley L, Tkac I, Rao R, Paulsen ME. Female and male C57BL/6J offspring exposed to maternal obesogenic diet develop altered hypothalamic energy metabolism in adulthood. Am J Physiol Endocrinol Metab 2022; 323:E448-E466. [PMID: 36342228 PMCID: PMC9639756 DOI: 10.1152/ajpendo.00100.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/12/2022] [Accepted: 09/12/2022] [Indexed: 11/22/2022]
Abstract
Maternal obesity is exceedingly common and strongly linked to offspring obesity and metabolic disease. Hypothalamic function is critical to obesity development. Hypothalamic mechanisms causing obesity following exposure to maternal obesity have not been elucidated. Therefore, we studied a cohort of C57BL/6J dams, treated with a control or high-fat-high-sugar diet, and their adult offspring to explore potential hypothalamic mechanisms to explain the link between maternal and offspring obesity. Dams treated with obesogenic diet were heavier with mild insulin resistance, which is reflective of the most common metabolic disease in pregnancy. Adult offspring exposed to maternal obesogenic diet had no change in body weight but significant increase in fat mass, decreased glucose tolerance, decreased insulin sensitivity, elevated plasma leptin, and elevated plasma thyroid-stimulating hormone. In addition, offspring exposed to maternal obesity had decreased energy intake and activity without change in basal metabolic rate. Hypothalamic neurochemical profile and transcriptome demonstrated decreased neuronal activity and inhibition of oxidative phosphorylation. Collectively, these results indicate that maternal obesity without diabetes is associated with adiposity and decreased hypothalamic energy production in offspring. We hypothesize that altered hypothalamic function significantly contributes to obesity development. Future studies focused on neuroprotective strategies aimed to improve hypothalamic function may decrease obesity development.NEW & NOTEWORTHY Offspring exposed to maternal diet-induced obesity demonstrate a phenotype consistent with energy excess. Contrary to previous studies, the observed energy phenotype was not associated with hyperphagia or decreased basal metabolic rate but rather decreased hypothalamic neuronal activity and energy production. This was supported by neurochemical changes in the hypothalamus as well as inhibition of hypothalamic oxidative phosphorylation pathway. These results highlight the potential for neuroprotective interventions in the prevention of obesity with fetal origins.
Collapse
Affiliation(s)
- Debra Kulhanek
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
| | | | - Lauren Buckley
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Ivan Tkac
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Raghavendra Rao
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Megan E Paulsen
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
- Minnesota Institute for the Developing Brain, Minneapolis, Minnesota
| |
Collapse
|
27
|
Łoniewska B, Michalczyk K, Podsiadło K, Adamek K, Michalczyk B, Tousty P, Kaczmarczyk M, Łoniewski I. Analysis of the Influence of Pre-Pregnancy BMI and Weight Gain during Pregnancy on the Weight of Healthy Children during the First 2 Years of Life: A Prospective Study. CHILDREN 2022; 9:children9101431. [PMID: 36291367 PMCID: PMC9600872 DOI: 10.3390/children9101431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022]
Abstract
Background: Increased pre-pregnancy maternal BMI (pBMI) and gestational weight gain (GWG) have been found to increase infants’ birthweight and result in the programming of child weight and impact its later weight gain. Aim: To assess the impact of pBMI and GWG on the weight of children from birth to 2 years of age and over the duration of breastfeeding. Methods: Single Centre observational prospective longitudinal cohort study. Data were collected from medical records, and medical history. The analysis of multiple linear and mixed models was involved. Findings: 20% of females were overweight, while 13% were obese before the pregnancy. An overall model, including gender and smoking, indicated a significant impact of pBMI category on a child’s birth mass (p = 0.01). The GWG category affected a child’s birth weight (p = 0.018, Effect size 0.41). pBMI did not affect the breastfeeding duration. Conclusion: pBMI and GWG correlate with birth weight and weight in neonatal period, however they become insignificant in later childhood. Weight assessment methods among children aged up to two years of age require standardization. Maternal weight before the pregnancy nor the weight gain during the pregnancy do not influence the length of breastfeeding. The biggest limitation was the small sample size and the failure to account for weight gain per trimester of pregnancy. Further research on a larger population should be continued.
Collapse
Affiliation(s)
- Beata Łoniewska
- Department of Neonatology and Intensive Neonatal Care, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Kaja Michalczyk
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
- Correspondence:
| | - Konrad Podsiadło
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Karolina Adamek
- Department of Neonatology and Intensive Neonatal Care, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Barbara Michalczyk
- Department of Neonatology and Intensive Neonatal Care, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Piotr Tousty
- Department of Obstetrics and Gynecology, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Mariusz Kaczmarczyk
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Igor Łoniewski
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland
| |
Collapse
|
28
|
Ormazabal V, Nair S, Carrión F, Mcintyre HD, Salomon C. The link between gestational diabetes and cardiovascular diseases: potential role of extracellular vesicles. Cardiovasc Diabetol 2022; 21:174. [PMID: 36057662 PMCID: PMC9441052 DOI: 10.1186/s12933-022-01597-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/05/2022] [Indexed: 11/25/2022] Open
Abstract
Extracellular vesicles are critical mediators of cell communication. They encapsulate a variety of molecular cargo such as proteins, lipids, and nucleic acids including miRNAs, lncRNAs, circular RNAs, and mRNAs, and through transfer of these molecular signals can alter the metabolic phenotype in recipient cells. Emerging studies show the important role of extracellular vesicle signaling in the development and progression of cardiovascular diseases and associated risk factors such as type 2 diabetes and obesity. Gestational diabetes mellitus (GDM) is hyperglycemia that develops during pregnancy and increases the future risk of developing obesity, impaired glucose metabolism, and cardiovascular disease in both the mother and infant. Available evidence shows that changes in maternal metabolism and exposure to the hyperglycemic intrauterine environment can reprogram the fetal genome, leaving metabolic imprints that define life-long health and disease susceptibility. Understanding the factors that contribute to the increased susceptibility to metabolic disorders of children born to GDM mothers is critical for implementation of preventive strategies in GDM. In this review, we discuss the current literature on the fetal programming of cardiovascular diseases in GDM and the impact of extracellular vesicle (EV) signaling in epigenetic programming in cardiovascular disease, to determine the potential link between EV signaling in GDM and the development of cardiovascular disease in infants.
Collapse
Affiliation(s)
- Valeska Ormazabal
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Building 71/918, Herston, QLD, 4029, Australia.,Faculty of Biological Sciences, Pharmacology Department, University of Concepcion, Concepción, Chile
| | - Soumyalekshmi Nair
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Building 71/918, Herston, QLD, 4029, Australia
| | - Flavio Carrión
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| | - H David Mcintyre
- Mater Research, Faculty of Medicine, University of Queensland, Mater Health, South Brisbane, Australia
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Building 71/918, Herston, QLD, 4029, Australia. .,Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| |
Collapse
|
29
|
Maternal high-fat diet promotes calcified atherosclerotic plaque formation in adult offspring by enhancing transformation of VSMCs to osteochondrocytic-like phenotype. Heliyon 2022; 8:e10644. [PMID: 36158105 PMCID: PMC9489965 DOI: 10.1016/j.heliyon.2022.e10644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/17/2022] [Accepted: 09/08/2022] [Indexed: 11/22/2022] Open
Abstract
Aim Maternal high-fat diet (HFD) is associated with the development of cardiovascular disease (CVD) in adult offspring. Atherosclerotic vascular calcification is well documented in patients with CVD. We examined the effect of maternal HFD on calcified plaque formation. Methods and results Seven-week-old female apo-E−/− mice (C57BL6/J) were nourished either an HFD or a normal diet (ND) a week before mating, and during gestation and lactation. Offspring of both the groups were fed a high-cholesterol diet (HCD) from 8 weeks of age. Osteogenic activity of the thoracic aorta, assessed using an ex vivo imaging system, was significantly increased after 3 months of HCD in male offspring of HFD-fed dams (O-HFD) as compared with those of ND-fed dams (O-ND). Alizarin-red-positive area in the aortic root was significantly increased after 6 months of HCD in male O-HFD as compared to that of O-ND. Plaque size and Oil Red O-positive staining were comparable between the two groups. Primary cultured vascular smooth muscle cells (VSMCs) of the thoracic aorta were treated with phosphate and interleukinL-1β (IL-1β) to transform them into an osteochondrocytic-like phenotype. Intracellular calcium content and alkaline phosphatase activity were markedly higher in the VSMCs of O-HFD than in O-ND. IL-1β concentration in the supernatant of bone marrow-derived macrophages was markedly higher in O-HFD than in O-ND. Conclusion Our findings indicate that maternal HFD accelerates the expansion of atherogenic calcification independent of plaque progression. In vitro phosphate- and IL-1β-induced osteochondrocytic transformation of VSMCs was augmented in O-HFD. Inhibition of VSMCs, skewing toward osteochondrocytic-like cells, might be a potential therapeutic strategy for preventing maternal HFD-associated CVD development.
Collapse
|
30
|
Ma Z, Wang Y, Quan Y, Wang Z, Liu Y, Ding Z. Maternal obesity alters methylation level of cytosine in CpG island for epigenetic inheritance in fetal umbilical cord blood. Hum Genomics 2022; 16:34. [PMID: 36045397 PMCID: PMC9429776 DOI: 10.1186/s40246-022-00410-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 08/22/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Over the past few decades, global maternal obesity prevalence has rapidly increased. This condition may induce long-lasting pathophysiological effects on either fetal or infant health that could be attributable to unknown unique changes in the umbilical blood composition. METHODS A total of 34 overweight/obese and 32 normal-weight pregnant women were recruited. Fifteen umbilical blood samples including 8 overweight/obese subjects and 7 normal weight women were sequenced using Targeted Bisulfite Sequencing technology to detect the average methylation level of cytosine and identify the differentially methylated region (DMR). GO and KEGG analyses were then employed to perform pathway enrichment analysis of DMR-related genes and promoters. Moreover, the mRNA levels of methylation-related genes histone deacetylases (HDACs) and DNA methyltransferases (DNMTs) were characterized in the samples obtained from these two groups. RESULTS Average methylated cytosine levels in both the CpG islands (CGI) and promoter significantly decreased in overweight/obese groups. A total of 1669 DMRs exhibited differences in their DNA methylation status between the overweight/obese and control groups. GO and KEGG analyses revealed that DMR-related genes and promoters were enriched in the metabolism, cancer and cardiomyopathy signaling pathways. Furthermore, the HDACs and DNMTs mRNA levels trended to decline in overweight/obese groups. CONCLUSIONS Decreased methylated cytosine levels in overweight/obese women induce the gene expression activity at a higher level than in the control group. DMRs between these two groups in the fetal blood may contribute to the changes in gene transcription that underlie the increased risk of metabolic disorders, cancers and cardiomyopathy in their offspring.
Collapse
Affiliation(s)
- Zhuoyao Ma
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, No.280, Chongqing Road (South), Shanghai, 200025, China
| | - Yingjin Wang
- Department of Obstetrics and Gynecology, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Yanmei Quan
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, No.280, Chongqing Road (South), Shanghai, 200025, China
| | - Zhijie Wang
- Department of Obstetrics and Gynecology, Shanghai Eighth People's Hospital, Shanghai, 200235, China.
| | - Yue Liu
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, No.280, Chongqing Road (South), Shanghai, 200025, China.
| | - Zhide Ding
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, No.280, Chongqing Road (South), Shanghai, 200025, China.
| |
Collapse
|
31
|
Quinte GC, Munhoz TN, Matijasevich A, Santos IS. Maternal pre-pregnancy body mass index and mental health problems in early adolescents from the 2004 Pelotas birth cohort. Sci Rep 2022; 12:14437. [PMID: 36002596 PMCID: PMC9402554 DOI: 10.1038/s41598-022-18032-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/03/2022] [Indexed: 11/10/2022] Open
Abstract
Possible mechanisms by which maternal pre-pregnancy body mass index (BMI) programs offspring mental disorders in late childhood are not fully clarified. To assess the association between maternal BMI and mental health problems at 11 years old, we used data from the 2004 Pelotas birth cohort which comprised 4231 newborns. Maternal pre-pregnancy BMI was analyzed as underweight (< 18.5), normal (18.5-24.9), overweight (25.0-29.9), and obesity (≥ 30.0). Mental health problems were assessed at the child's age of 11 years by the Strengths and Difficulties Questionnaire, total difficulties score and subscale scores (emotional symptoms, conduct problems, hyperactivity/inattention, and peer relationship problems), dichotomized into normal/borderline and abnormal category. The associations between maternal pre-pregnancy BMI and mental health problems in the whole sample and stratified by sex were estimated using crude and adjusted linear and logistic regression. Both linear and logistic regression showed that pre-pregnancy weight was associated with mental problems in early adolescents. Maternal pre-pregnancy obesity was associated with increased odds of total difficulty score among the whole sample. Boys whose mothers were pre-pregnancy overweight had higher odds of conduct problems (OR = 1.56; 95% CI: 1.13, 1.95), when compared to children of normal pre-pregnancy weight mothers, even after adjustments. Maternal pre-pregnancy obesity was associated with mental health problems in early adolescents; specifically, pre-pregnancy overweight increased the risk for conduct problems in 11 years old boys.
Collapse
Affiliation(s)
- Gabriela Callo Quinte
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Rua Marechal Deodoro, 1160 - 3° Piso, Bairro Centro, Caixa Postal 464, Pelotas, RS, Cep: 96020-220, Brazil.
| | - Tiago N Munhoz
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Rua Marechal Deodoro, 1160 - 3° Piso, Bairro Centro, Caixa Postal 464, Pelotas, RS, Cep: 96020-220, Brazil.,School of Psychology, Federal University of Pelotas, Pelotas, Brazil
| | - Alicia Matijasevich
- Departamento de Medicina Preventiva, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Ina S Santos
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Rua Marechal Deodoro, 1160 - 3° Piso, Bairro Centro, Caixa Postal 464, Pelotas, RS, Cep: 96020-220, Brazil
| |
Collapse
|
32
|
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
|
33
|
Murphy CC, Cirillo PM, Krigbaum NY, Singal AG, Lee M, Zaki T, Burstein E, Cohn BA. Maternal obesity, pregnancy weight gain, and birth weight and risk of colorectal cancer. Gut 2022; 71:1332-1339. [PMID: 34429385 PMCID: PMC8866526 DOI: 10.1136/gutjnl-2021-325001] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/28/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Colorectal cancer (CRC) is a leading cause of cancer-related death worldwide. Obesity is a well-established risk factor for CRC, and fetal or developmental origins of obesity may underlie its effect on cancer in adulthood. We examined associations of maternal obesity, pregnancy weight gain, and birth weight and CRC in adult offspring. DESIGN The Child Health and Development Studies is a prospective cohort of women receiving prenatal care between 1959 and 1966 in Oakland, California (N=18 751 live births among 14 507 mothers). Clinical information was abstracted from mothers' medical records 6 months prior to pregnancy through delivery. Diagnoses of CRC in adult (age ≥18 years) offspring were ascertained through 2019 by linkage with the California Cancer Registry. We used Cox proportional hazards models to estimate adjusted HR (aHR); we examined effect measure modification using single-referent models to estimate the relative excess risk due to interaction (RERI). RESULTS 68 offspring were diagnosed with CRC over 738 048 person-years of follow-up, and half (48.5%) were diagnosed younger than age 50 years. Maternal obesity (≥30 kg/m2) increased the risk of CRC in offspring (aHR 2.51, 95% CI 1.05 to 6.02). Total weight gain modified the association of rate of early weight gain (RERI -4.37, 95% CI -9.49 to 0.76), suggesting discordant growth from early to late pregnancy increases risk. There was an elevated association with birth weight (≥4000 g: aHR 1.95, 95% CI 0.8 to 4.38). CONCLUSION Our results suggest that in utero events are important risk factors for CRC and may contribute to increasing incidence rates in younger adults.
Collapse
Affiliation(s)
- Caitlin C Murphy
- School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Piera M Cirillo
- Child Health and Development Studies, Public Health Institute, Oakland, California, USA
| | - Nickilou Y Krigbaum
- Child Health and Development Studies, Public Health Institute, Oakland, California, USA
| | - Amit G Singal
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - MinJae Lee
- Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Timothy Zaki
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ezra Burstein
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Barbara A Cohn
- Child Health and Development Studies, Public Health Institute, Oakland, California, USA
| |
Collapse
|
34
|
Shalitin S, Gat-Yablonski G. Associations of Obesity with Linear Growth and Puberty. Horm Res Paediatr 2022; 95:120-136. [PMID: 34130293 DOI: 10.1159/000516171] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 03/27/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The prevalence of obesity in childhood has increased dramatically in recent decades with increased risk of developing cardiometabolic and other comorbidities. Childhood adiposity may also influence processes of growth and puberty. SUMMARY Growth patterns of obesity during childhood have been shown to be associated with increased linear growth in early childhood, leading to accelerated epiphyseal growth plate (EGP) maturation. Several hormones secreted by the adipose tissue may affect linear growth in the context of obesity, both via the growth hormone IGF-1 axis and via a direct effect on the EGP. The observation that children with obesity tend to mature earlier than lean children has led to the assumption that the degree of body fatness may trigger the neuroendocrine events that lead to pubertal onset. The most probable link between obesity and puberty is leptin and its interaction with the kisspeptin system, which is an important regulator of puberty. However, peripheral action of adipose tissue could also be involved in changes in the onset of puberty. In addition, nutritional factors, epigenetics, and endocrine-disrupting chemicals are potential mediators linking pubertal onset to obesity. In this review, we focused on interactions of obesity with linear growth and pubertal processes, based on basic research and clinical data in humans. KEY MESSAGE Children with obesity are subject to accelerated linear growth with risk of impaired adult height and early puberty, with its psychological consequences. The data highlight another important objective in combatting childhood obesity, for the prevention of abnormal growth and pubertal patterns.
Collapse
Affiliation(s)
- Shlomit Shalitin
- National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Galia Gat-Yablonski
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Felsenstein Medical Research Center, Petach Tikva, Israel
| |
Collapse
|
35
|
Dumolt J, Powell TL, Jansson T, Rosario FJ. Normalization of maternal adiponectin in obese pregnant mice prevents programming of impaired glucose metabolism in adult offspring. FASEB J 2022; 36:e22383. [PMID: 35670755 DOI: 10.1096/fj.202200326r] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 11/11/2022]
Abstract
Infants born to obese mothers have a greater risk for childhood obesity and insulin resistance. However, the underlying biological mechanism remains elusive, which constitutes a significant roadblock for developing specific prevention strategies. Maternal adiponectin levels are lower in obese pregnant women, which is linked with increased placental nutrient transport and fetal overgrowth. We have previously reported that adiponectin supplementation to obese dams during the last four days of pregnancy prevented the development of obesity, glucose intolerance, muscle insulin resistance, and fatty liver in three months old offspring. In the present study, we tested the hypothesis that 6-9-month-old offspring of obese dams show glucose intolerance associated with muscle insulin resistance and mitochondrial dysfunction and that normalization of maternal adiponectin in obese pregnant mice prevents the development of this phenotype in the offspring. Male and female offspring of obese mice exhibited in vivo glucose intolerance and insulin resistance at 6 and 9 months of age. In gastrocnemius muscles ex vivo, male and female offspring of obese dams showed reduced phosphorylation of insulin receptor substrate 1Tyr-608 , AktThr-308 , and decreased Glut4 plasma membrane translocation upon insulin stimulation. These metabolic abnormalities in offspring born to obese mice were largely prevented by normalization of maternal adiponectin levels in late pregnancy. We provide evidence that low circulating maternal adiponectin is a critical mechanistic link between maternal obesity and the development of metabolic disease in offspring. Strategies aimed at improving maternal adiponectin levels may prevent long-term metabolic dysfunction in offspring of obese mothers.
Collapse
Affiliation(s)
- Jerad Dumolt
- Division of Reproductive Sciences, Department of OB/GYN, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Theresa L Powell
- Division of Reproductive Sciences, Department of OB/GYN, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of OB/GYN, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Fredrick J Rosario
- Division of Reproductive Sciences, Department of OB/GYN, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
36
|
Bongers-Karmaoui MN, Jaddoe VW, Roest AA, Helbing WA, Steegers EA, Gaillard R. Associations of maternal angiogenic factors during pregnancy with alterations in cardiac development in childhood at 10 years of age. Am Heart J 2022; 247:100-111. [PMID: 35123935 DOI: 10.1016/j.ahj.2022.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/23/2021] [Accepted: 01/29/2022] [Indexed: 11/27/2022]
Abstract
AIM To examine whether maternal angiogenic factors in the first half of pregnancy are associated with offspring left and right cardiac development. METHODS In a population-based prospective cohort among 2,415 women and their offspring, maternal first and second trimester plasma PlGF and sFlt-1 concentrations were measured. Cardiac MRI was performed in their offspring at 10 years. RESULTS Maternal angiogenic factors were not associated with childhood cardiac outcomes in the total population. In children born small-for-their-gestational-age, higher maternal first trimester PlGF concentrations were associated with a lower childhood left ventricular mass (-0.24 SDS [95%CI -0.42, -0.05 per SDS increase in maternal PlGF]), whereas higher sFlt-1 concentrations were associated with higher childhood left ventricular mass (0.22 SDS [95%CI 0.09, 0.34 per SDS increase in maternal sFlt-1]). Higher second trimester maternal sFlt-1 concentrations were also associated with higher childhood left ventricular mass (P-value <.05). In preterm born children, higher maternal first and second trimester sFlt-1/PlGF ratio were associated with higher childhood left ventricular mass (0.30 SDS [95%CI 0.01, 0.60], 0.22 SDS [95%CI -0.03, 0.40]) per SDS increase in maternal sFlt-1/PlGF ratio in first and second trimester respectively). No effects on other childhood cardiac outcomes were present within these higher-risk children. CONCLUSIONS In a low-risk population, maternal angiogenic factors are not associated with childhood cardiac ventricular structure, and function within the normal range. In children born small for their gestational age or preterm, an imbalance in maternal angiogenic factors in the first half of pregnancy was associated with higher childhood left ventricular mass only.
Collapse
|
37
|
Paternal periconception metabolic health and offspring programming. Proc Nutr Soc 2022; 81:119-125. [DOI: 10.1017/s0029665121003736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The association between maternal metabolic status at the time of conception and subsequent embryogenesis and offspring development has been studied in detail. However, less attention has been given to the significance of paternal nutrition and metabolism in directing offspring health. Despite this disparity, emerging evidence has begun to highlight an important connection between paternal metabolic well-being, semen quality, embryonic development and ultimately adult offspring health. This has established a new component within the Developmental Origins of Health and Disease hypothesis. Building on the decades of understanding and insight derived from the numerous models of maternal programming, attention is now becoming focused on defining the mechanisms underlying the links between paternal well-being, post-fertilisation development and offspring health. Understanding how the health and fitness of the father impact on semen quality is of fundamental importance for providing better information to intending fathers. Furthermore, assisted reproductive practices such as in vitro fertilisation rely on our ability to select the best quality sperm from a diverse and heterogeneous population. With considerable advances in sequencing capabilities, our understanding of the molecular and epigenetic composition of the sperm and seminal plasma, and their association with male metabolic health, has developed dramatically over recent years. This review will summarise our current understanding of how a father's metabolic status at the time of conception can affect sperm quality, post-fertilisation embryonic and fetal development and offspring health.
Collapse
|
38
|
McLennan NM, Hazlehurst J, Thangaratinam S, Reynolds RM. ENDOCRINOLOGY IN PREGNANCY: Targeting metabolic health promotion to optimise maternal and offspring health. Eur J Endocrinol 2022; 186:R113-R126. [PMID: 35380983 PMCID: PMC9066590 DOI: 10.1530/eje-21-1046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 04/05/2022] [Indexed: 11/16/2022]
Abstract
There is an increase in maternal metabolic burden due to the rise in pregnancies complicated by obesity, gestational diabetes, type 2 diabetes and polycystic ovary syndrome. Metabolic dysfunction during pregnancy is associated with increased risks of long-term morbidity and mortality for women and their offspring. Lifestyle interventions in pregnancy in women at risk of metabolic dysfunction have demonstrated short-term improvements such as reduced gestational weight gain and lowered risk of gestational diabetes. It is not known whether these interventions lead to sustained improvements in the metabolic health of the mother and baby. Pharmacological interventions have also shown benefits for the mother and baby in pregnancy, including improvements in glycaemic control, reduction in gestational weight gain and reduction in large for gestational age infants; however, there remains uncertainty over long-term outcomes for mother and child. Existing studies on interventions targeting metabolic health are limited to selected populations in the preconception and postpartum periods and lack follow-up beyond delivery of the intervention. The COVID-19 pandemic has refocused our attention on the effects of maternal metabolic ill-health that play a role in contributing to premature morbidity and mortality. There is an urgent need for strategies to accurately identify the growing number of women and offspring at risk of long-term adverse metabolic health. Strategies which focus on early identification and risk stratification using individualised risk scores in the pre and inter-conception periods must take priority if we are to target and improve the metabolic health of women and their offspring who are at highest risk.
Collapse
Affiliation(s)
- Niamh-Maire McLennan
- MRC Centre for Reproductive Health, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, UK
| | - Jonathan Hazlehurst
- Department of Diabetes and Endocrinology, University Hospital Birmingham Foundation Trust, Birmingham, UK
| | - Shakila Thangaratinam
- WHO Collaborating Centre for Women’s Health, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Birmingham Women’s and Children’s NHS Trust, Birmingham, UK
| | - Rebecca M Reynolds
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, UK
| |
Collapse
|
39
|
Zeng J, Shen F, Zou ZY, Yang RX, Jin Q, Yang J, Chen GY, Fan JG. Association of maternal obesity and gestational diabetes mellitus with overweight/obesity and fatty liver risk in offspring. World J Gastroenterol 2022; 28:1681-1691. [PMID: 35581961 PMCID: PMC9048784 DOI: 10.3748/wjg.v28.i16.1681] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/30/2021] [Accepted: 03/16/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Childhood obesity and fatty liver are associated with adverse outcomes such as diabetes, metabolic syndrome, and cardiovascular diseases in adulthood. It is very important to identify relevant risk factors and intervene as early as possible. At present, the relationship between maternal and offspring metabolic factors is conflicting.
AIM To estimate the association of maternal obesity and gestational diabetes mellitus (GDM) with overweight/obesity and fatty liver risk in offspring at 8 years of age.
METHODS The prospective study included mothers who all had a 75-g oral glucose tolerance test at 24-28 wk of gestation and whose offspring completed follow-up at 8 years of age. Offspring birth weight, sex, height, weight, and body mass index (BMI) were measured and calculated. FibroScan-502 examination with an M probe (Echosens, Paris, France) was prospectively conducted in offspring aged 8 years from the Shanghai Prenatal Cohort Study.
RESULTS A total of 430 mother-child pairs were included in the analysis. A total of 62 (14.2%) mothers were classified as obese, and 48 (11.1%) were classified as having GDM. The mean age of the offspring at follow-up was 8 years old. Thirty-seven (8.6%) offspring were overweight, 14 (3.3%) had obesity, and 60 (14.0%) had fatty liver. The prevalence of overweight, obesity and fatty liver in offspring increased significantly across maternal BMI quartiles (all P < 0.05). Among offspring of mothers with GDM, 12 (25.0%) were overweight, 4 (8.3%) were obese, and 12 (25.0%) had fatty liver vs. 25 (6.5%), 10 (2.6%) and 48 (12.6%), respectively, for offspring of mothers without GDM (all P < 0.05). In multiple logistic regression, after adjustment for variables, the OR for fatty liver in offspring was 8.26 (95%CI: 2.38-28.75) for maternal obesity and GDM.
CONCLUSION This study showed that maternal obesity can increase the odds of overweight/obesity and fatty liver in offspring, and GDM status also increases the odds of overweight/obesity in offspring. Weight management and glycemic control before and during pregnancy need to be highlighted in primary prevention of pediatric obesity and fatty liver.
Collapse
Affiliation(s)
- Jing Zeng
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Feng Shen
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Zi-Yuan Zou
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Rui-Xu Yang
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Qian Jin
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jing Yang
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Guang-Yu Chen
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
| |
Collapse
|
40
|
Pippen J, Stetson B, Doherty L, Varner MW, Casey BM, Reddy UM, Wapner RJ, Rouse DJ, Tita ATN, Thorp JM, Chien EK, Saade GR, Blackwell SC. Neonatal Birthweight, Infant Feeding, and Childhood Metabolic Markers. Am J Perinatol 2022; 39:584-591. [PMID: 34918330 PMCID: PMC9106839 DOI: 10.1055/s-0041-1740056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Antenatal and early neonatal nutritional environment may influence later metabolic health. Infants of mothers with gestational diabetes mellitus (GDM) have higher risk for childhood obesity and metabolic syndrome (MetS). Leptin and adiponectin are known biomarkers for MetS and may guide interventions to reduce later obesity. We sought to examine the relationship between birthweight, early infancy feeding practices, and biomarkers for MetS in offspring of women with mild GDM. STUDY DESIGN Secondary analysis of a prospective observational follow-up study on the offspring of women who participated in a multicenter randomized treatment trial on mild GDM. Children were evaluated by research coordinators and biospecimens collected at the age of 5 to 10. Plasma concentrations of leptin and adiponectin were compared between large for gestational age (LGA) and average birthweight (AGA) infants, and according to whether solid foods were introduced early (<6 months of age) or at the recommended age (≥6 months of age). Multivariable analysis adjusted for fetal sex, race/ethnicity, and maternal body mass index. RESULTS Leptin and adiponectin were measured in 336 plasma samples. In bivariate analysis, compared with AGA children, LGA children had lower leptin (5.0 ng/mL [3.6-6.0] vs. 5.8 ng/mL [4.5 = 6.6], p = 0.01) and similar adiponectin (6.3 µg/mL [5.1-7.9] vs. 6.4 µg/mL [5.3-8.6], p = 0.49) concentrations. Maternal/child characteristics were similar between the early/delayed solid feeding groups. Leptin and adiponectin concentrations were similar in the early fed and delayed feeding groups (5.8 ng/mL [4.6-6.7] vs. 5.6 ng/mL [4.2-6.6], p = 0.50 and 6.4 µg/mL [5.4-8.1] vs. 6.4 µg/mL [5.1-8.8], p = 0.85, respectively). After controlling for covariates, children who were LGA and AGA at birth had similar leptin concentrations. CONCLUSION Birthweight and early infancy feeding practice are not associated with alterations in leptin and adiponectin in children of women with mild GDM. KEY POINTS · Adipocytokines are markers of metabolic status.. · Children of women with mild GDM may be at risk for MetS.. · Biomarkers similar in LGA and AGA groups.. · Biomarkers similar in early and delayed solid-fed groups.. · Nonhuman milk does not modify effect of feeding practice..
Collapse
Affiliation(s)
- Jessica Pippen
- Department of Obstetrics and Gynecology of The Ohio State University, Columbus, Ohio
| | - Bethany Stetson
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, Illinois
| | - Lindsay Doherty
- Department of Obstetrics and Gynecology, The George Washington University Biostatistics Center, Washington, Dist. of Columbia
| | - Michael W. Varner
- Department of Obstetrics and Gynecology, The University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Brian M. Casey
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Uma M. Reddy
- Department of Obstetrics and Gynecology, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Rockville, Maryland
| | - Ronald J. Wapner
- Department of Obstetrics and Gynecology, Columbia University, New York, New York
| | - Dwight J. Rouse
- Department of Obstetrics and Gynecology, Brown University, Providence, Rhode Island
| | | | - John M. Thorp
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Edward K. Chien
- MetroHealth Medical Center-Case Western Reserve University, Cleveland, Ohio
| | | | - Sean C. Blackwell
- University of Texas Health Science Center at Houston-Children’s Memorial Hermann Hospital, Houston, Texas
| | | |
Collapse
|
41
|
Perichart-Perera O, Avila-Sosa V, Solis-Paredes JM, Montoya-Estrada A, Reyes-Muñoz E, Rodríguez-Cano AM, González-Leyva CP, Sánchez-Martínez M, Estrada-Gutierrez G, Irles C. Vitamin D Deficiency, Excessive Gestational Weight Gain, and Oxidative Stress Predict Small for Gestational Age Newborns Using an Artificial Neural Network Model. Antioxidants (Basel) 2022; 11:574. [PMID: 35326224 PMCID: PMC8944993 DOI: 10.3390/antiox11030574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/10/2022] [Accepted: 03/14/2022] [Indexed: 02/04/2023] Open
Abstract
(1) Background: Size at birth is an important early determinant of health later in life. The prevalence of small for gestational age (SGA) newborns is high worldwide and may be associated with maternal nutritional and metabolic factors. Thus, estimation of fetal growth is warranted. (2) Methods: In this work, we developed an artificial neural network (ANN) model based on first-trimester maternal body fat composition, biochemical and oxidative stress biomarkers, and gestational weight gain (GWG) to predict an SGA newborn in pregnancies with or without obesity. A sensibility analysis to classify maternal features was conducted, and a simulator based on the ANN algorithm was constructed to predict the SGA outcome. Several predictions were performed by varying the most critical maternal features attained by the model to obtain different scenarios leading to SGA. (3) Results: The ANN model showed good performance between the actual and simulated data (R2 = 0.938) and an AUROC of 0.8 on an independent dataset. The top-five maternal predictors in the first trimester were protein and lipid oxidation biomarkers (carbonylated proteins and malondialdehyde), GWG, vitamin D, and total antioxidant capacity. Finally, excessive GWG and redox imbalance predicted SGA newborns in the implemented simulator. Significantly, vitamin D deficiency also predicted simulated SGA independently of GWG or redox status. (4) Conclusions: The study provided a computational model for the early prediction of SGA, in addition to a promising simulator that facilitates hypothesis-driven constructions, to be further validated as an application.
Collapse
Affiliation(s)
- Otilia Perichart-Perera
- Nutrition and Bioprogramming Coordination, Instituto Nacional de Perinatologia, Mexico City 11000, Mexico; (O.P.-P.); (A.M.R.-C.); (C.P.G.-L.)
| | - Valeria Avila-Sosa
- Department of Physiology and Cellular Development, Instituto Nacional de Perinatologia, Mexico City 11000, Mexico;
| | - Juan Mario Solis-Paredes
- Department of Human Genetics and Genomics, Instituto Nacional de Perinatologia, Mexico City 11000, Mexico;
| | - Araceli Montoya-Estrada
- Coordination of Gynecological and Perinatal Endocrinology, Instituto Nacional de Perinatologia, Mexico City 11000, Mexico; (A.M.-E.); (E.R.-M.)
| | - Enrique Reyes-Muñoz
- Coordination of Gynecological and Perinatal Endocrinology, Instituto Nacional de Perinatologia, Mexico City 11000, Mexico; (A.M.-E.); (E.R.-M.)
| | - Ameyalli M. Rodríguez-Cano
- Nutrition and Bioprogramming Coordination, Instituto Nacional de Perinatologia, Mexico City 11000, Mexico; (O.P.-P.); (A.M.R.-C.); (C.P.G.-L.)
| | - Carla P. González-Leyva
- Nutrition and Bioprogramming Coordination, Instituto Nacional de Perinatologia, Mexico City 11000, Mexico; (O.P.-P.); (A.M.R.-C.); (C.P.G.-L.)
| | - Maribel Sánchez-Martínez
- Department of Immunobiochemistry, Instituto Nacional de Perinatologia, Mexico City 11000, Mexico;
| | | | - Claudine Irles
- Department of Physiology and Cellular Development, Instituto Nacional de Perinatologia, Mexico City 11000, Mexico;
| |
Collapse
|
42
|
Haby K, Gyllensten H, Hanas R, Berg M, Premberg Å. A Lifestyle Intervention During Pregnancy and Its Effects on Child Weight 2.5 Years Later. Matern Child Health J 2022; 26:1881-1890. [PMID: 35253077 PMCID: PMC9374787 DOI: 10.1007/s10995-022-03395-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2022] [Indexed: 11/27/2022]
Abstract
Aim The aim of this study was to evaluate if overweight and obesity in the offspring is reduced by a low-intensity antenatal primary care intervention with focus on diet and physical activity for pregnant women with obesity, comparing children to mothers receiving the intervention with children to mothers who did not. Methods This study is a follow-up of children 2.5 years of age after their mothers’ participation in a non-randomised controlled intervention intending to limit gestational weight gain. All study participants received standard antenatal care. The intervention group received lifestyle support via motivational talks with midwife and support from dietician. Data on child weight were collected by medical records, letter and phone. Results There was no significant difference between the groups 2.5 years after intervention (International Obesity Task Force ISO-BMI 25 (child BMI corresponding to adult BMI of 25): 20% vs. 21%; ISO-BMI 30: 4.6% vs. 1.3%). The mother’s BMI at the beginning of pregnancy significantly influenced child BMI at 2.5 years (r = 0.13, p = 0.014, r2 = 0.017). For each unit of increase in maternal BMI at enrollment, the probability of child ISO-BMI ≥ 25 increased by 7.5% (p = 0.021) and of ≥ 30, by 12.9% (p = 0.017). Conclusion The frequency of overweight and obesity of the children at 2.5 years of age was significantly correlated to the mother’s BMI, but not correlated to the mothers’ participation in the antenatal lifestyle intervention. Thus, it seems important to address obesity and lifestyle issues before and between pregnancies. Trial registration The study has been registered at ClinicalTrials.gov, Identifier: NCT03147079. Supplementary Information The online version contains supplementary material available at 10.1007/s10995-022-03395-5.
Collapse
Affiliation(s)
- Karin Haby
- Antenatal Health Care, Primary Health Care, Research and Development Unit, Regionhälsan, Gothenburg, Sweden.
- Institute of Health and Care Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Primary Health Care and Research and Development Unit, Regionhälsan, Gothenburg, Region Västra Götaland, Sweden.
| | - Hanna Gyllensten
- Institute of Health and Care Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- University of Gothenburg Centre for Person-Centred Care (GPCC), Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ragnar Hanas
- Department of Paediatrics, NU Hospital Group, Uddevalla, Sweden
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Marie Berg
- Institute of Health and Care Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- University of Gothenburg Centre for Person-Centred Care (GPCC), Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics and Gynaecology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Åsa Premberg
- Institute of Health and Care Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Primary Health Care and Research and Development Unit, Regionhälsan, Gothenburg, Region Västra Götaland, Sweden
| |
Collapse
|
43
|
Brodowski L, Rochow N, Yousuf EI, Kohls F, von Kaisenberg CS, Berlage S, Voigt M. The impact of parity and maternal obesity on the fetal outcomes of a non-selected Lower Saxony population. J Perinat Med 2022; 50:167-175. [PMID: 34695308 DOI: 10.1515/jpm-2020-0614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 10/05/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Maternal obesity during pregnancy is associated with adverse intrauterine events and fetal outcomes and may increase the risk of obesity and metabolic disease development in offspring. Higher parity, regardless of socioeconomic status, is associated with increased maternal body mass index (BMI). In this study, we examined the relationship between parity, maternal obesity, and fetal outcomes in a large sample of mother-neonate pairs from Lower Saxony, Germany. METHODS This retrospective cohort study examined pseudonymized data of a non-selected singleton cohort from Lower Saxony's statewide quality assurance initiative. 448,963 cases were included. Newborn outcomes were assessed in relation to maternal BMI and parity. RESULTS Maternal obesity was associated with an increased risk of placental insufficiency, chorioamnionitis, and fetal distress while giving birth. This effect was present across all parity groups. Fetal presentation did not differ between BMI groups, except for the increased risk of high longitudinal position and shoulder dystocia in obese women. Maternal obesity was also associated with an increased risk of premature birth, low arterial cord blood pH and low 5-min APGAR scores. CONCLUSIONS Maternal obesity increases the risk of adverse neonatal outcomes. There is a positive correlation between parity and increased maternal BMI. Weight-dependent fetal risk factors increase with parity, while parity-dependent outcomes occur less frequently in multipara. Prevention and intervention programs for women planning to become pregnant can be promising measures to reduce pregnancy and birth complications.
Collapse
Affiliation(s)
- Lars Brodowski
- Department of Obstetrics, Gynecology and Reproductive Medicine, Hannover Medical School, Hannover, Germany
| | - Niels Rochow
- Department of Pediatrics, Paracelsus Medical University, Nuremberg, Germany.,Department of Pediatrics, Univesity Hospital Rostock, Rostock, Germany.,Department of Pediatrics, McMaster University, Hamilton, ON, Canada
| | - Efrah I Yousuf
- Department of Pediatrics, McMaster University, Hamilton, ON, Canada.,Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON, Canada
| | - Fabian Kohls
- Department of Obstetrics, Gynecology and Reproductive Medicine, Hannover Medical School, Hannover, Germany
| | | | - Silvia Berlage
- Center for Quality and Management in Health Care, Ärztekammer Niedersachsen, Hannover, Germany
| | - Manfred Voigt
- Faculty of Medicine, Department of Obstetrics and Gynecology, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany.,Biological Anthropology, Medical Faculty, University of Freiburg, Freiburg, Germany
| |
Collapse
|
44
|
Fayed A, Wahabi HA, Esmaeil S, Elkouny R, Elmorshedy H, Bakhsh H. Independent effect of gestational weight gain and prepregnancy obesity on pregnancy outcomes among Saudi women: A sub-cohort analysis from Riyadh mother and baby cohort study (RAHMA). PLoS One 2022; 17:e0262437. [PMID: 35015784 PMCID: PMC8751991 DOI: 10.1371/journal.pone.0262437] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 12/22/2021] [Indexed: 12/02/2022] Open
Abstract
Background Gestational weight gain (GWG) and prepregnancy obesity are garnering more attention as determining factors of pregnancy outcomes when it comes to the wellbeing of both the mother and her baby. This study was conducted to describe the pattern of GWG among participants of Riyadh Mother and Baby Multicenter Cohort Study (RAHMA) and to investigate the detrimental effects of excessive GWG and prepregnancy obesity on pregnancy outcomes. Methods RAHMA is a multicentre cohort study conducted in three hospitals in Riyadh, Saudi Arabia. Participants were categorized according to the Institute of Medicine into inadequate, adequate, and excessive GWG, and stratified by body mass index (BMI) into under/normal weight, overweight, and obese. To examine the independent effect of maternal prepregnancy obesity and GWG, a multivariate regression model was used and adjusted odds ratio (AOR) and 95% Confidence Interval (CI) for each outcome were calculated. Results A total of 7029 participants were included in this study; 31.8% had adequate GWG, 25.9% had excessive GWG and 42.3% had inadequate GWG, while 29.7% had normal BMI, 33.3% were overweight, 34.8% were obese, and 2.2% were underweight. Excessive GWG was independently associated with increased risk of hypertensive events, (AOR = 1.77, 95% CI 1.20–2.63). Obesity was associated with higher risk of gestational diabetes (AOR 2.11, 95% CI 1.76–2.53), hypertensive events (AOR 2.06, 95% CI 1.48–3.01), and delivery by emergency caesarean section (AOR = 1.63, 95% CI 1.35–1.97). Infants of obese women had increased odds of macrosomia (AOR 3.11, 95% CI 1.94–4.99) and lower odds of low birth weight (AOR = 0.68, 95% CI 0.53–0.88). Conclusion In comparison to excessive GWG, which increases the risk of hypertensive events during pregnancy, prepregnancy obesity is associated with more adverse outcomes including GDM, hypertensive events in pregnancy and emergency CS.
Collapse
Affiliation(s)
- Amel Fayed
- Clinical Sciences Department, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Hayfaa A. Wahabi
- Research Chair of Evidence-Based Healthcare and Knowledge Translation, King Saud University, Riyadh, Saudi Arabia
- Department of Family and Community Medicine, King Saud University Medical City and College of Medicine, Riyadh, Saudi Arabia
- * E-mail:
| | - Samia Esmaeil
- Research Chair of Evidence-Based Healthcare and Knowledge Translation, King Saud University, Riyadh, Saudi Arabia
| | - Roaa Elkouny
- College of Medicine, AlFaisal University, Riyadh, Saudi Arabia
| | - Hala Elmorshedy
- Clinical Sciences Department, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Hanadi Bakhsh
- Clinical Sciences Department, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| |
Collapse
|
45
|
Tajaddini A, Kendig MD, Prates KV, Westbrook RF, Morris MJ. Male Rat Offspring Are More Impacted by Maternal Obesity Induced by Cafeteria Diet than Females-Additive Effect of Postweaning Diet. Int J Mol Sci 2022; 23:ijms23031442. [PMID: 35163366 PMCID: PMC8835941 DOI: 10.3390/ijms23031442] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 01/07/2023] Open
Abstract
Maternal obesity increases the risk of health complications in offspring, but whether these effects are exacerbated by offspring exposure to unhealthy diets warrants further investigation. Female Sprague-Dawley rats were fed either standard chow (n = 15) or ‘cafeteria’ (Caf, n = 21) diets across pre-pregnancy, gestation, and lactation. Male and female offspring were weaned onto chow or Caf diet (2–3/sex/litter), forming four groups; behavioural and metabolic parameters were assessed. At weaning, offspring from Caf dams were smaller and lighter, but had more retroperitoneal (RP) fat, with a larger effect in males. Maternal Caf diet significantly increased relative expression of ACACA and Fasn in male and female weanling liver, but not CPT-1, SREBP and PGC1; PPARα was increased in males from Caf dams. Maternal obesity enhanced the impact of postweaning Caf exposure on adult body weight, RP fat, liver mass, and plasma leptin in males but not females. Offspring from Caf dams appeared to exhibit reduced anxiety-like behaviour on the elevated plus maze. Hepatic CPT-1 expression was reduced only in adult males from Caf fed dams. Post weaning Caf diet consumption did not alter liver gene expression in the adult offspring. Maternal obesity exacerbated the obesogenic phenotype produced by postweaning Caf diet in male, but not female offspring. Thus, the impact of maternal obesity on adiposity and liver gene expression appeared more marked in males. Our data underline the sex-specific detrimental effects of maternal obesity on offspring.
Collapse
Affiliation(s)
- Aynaz Tajaddini
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; (A.T.); (M.D.K.); (K.V.P.)
| | - Michael D. Kendig
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; (A.T.); (M.D.K.); (K.V.P.)
| | - Kelly V. Prates
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; (A.T.); (M.D.K.); (K.V.P.)
| | | | - Margaret J. Morris
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; (A.T.); (M.D.K.); (K.V.P.)
- Correspondence:
| |
Collapse
|
46
|
The postnatal leptin surge in mice is variable in both time and intensity and reflects nutritional status. Int J Obes (Lond) 2022; 46:39-49. [PMID: 34475504 PMCID: PMC8748198 DOI: 10.1038/s41366-021-00957-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 07/27/2021] [Accepted: 08/20/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND/OBJECTIVES The murine postnatal leptin surge occurs within the first 4 weeks of life and is critical for neuronal projection development within hypothalamic feeding circuits. Here we describe the influence of nutritional status on the timing and magnitude of the postnatal leptin surge in mice. METHODS Plasma leptin concentrations were measured 1-3 times per week for the first 4 weeks of life in C57BL/6J pups reared in litters adjusted to 3 (small), 7-8 (normal), or 11-12 (large) pups per dam fed breeder chow or raised in litters of 7-8 by dams fed high-fat diet (HFD) ad libitum starting either prior to conception or at parturition. RESULTS Mice raised in small litters become fatter than pups raised in either normal or large litters. The leptin surge in small litter pups starts earlier, lasts longer, and is dramatically larger in magnitude compared to normal litter pups, even when leptin concentrations are normalized to fat mass. In mice reared in large litters, weight gain is diminished and the surge is both significantly delayed and lower in magnitude compared to control pups. Pups reared by HFD-fed dams (starting preconception or at parturition) are fatter and have augmented leptin surge magnitude compared to pups suckled by chow-fed dams. Surge timing varies depending upon nutritional status of the pup; the source of the surge is primarily subcutaneous adipose tissue. At peak leptin surge, within each group, fat mass and plasma leptin are uncorrelated; in comparison with adults, pups overproduce leptin relative to fat mass. Plasma leptin elevation persists longer than previously described; at postnatal day 27 mice continue overproducing leptin relative to fat mass. CONCLUSIONS In mice, small litter size and maternal HFD feeding during the perinatal period augment the plasma leptin surge whereas large litter size is associated with a delayed surge of reduced magnitude.
Collapse
|
47
|
Anwer H, Morris MJ, Noble DWA, Nakagawa S, Lagisz M. Transgenerational effects of obesogenic diets in rodents: A meta-analysis. Obes Rev 2022; 23:e13342. [PMID: 34595817 DOI: 10.1111/obr.13342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/22/2022]
Abstract
Obesity is a major health condition that affects millions worldwide. There is an increased interest in understanding the adverse outcomes associated with obesogenic diets. A multitude of studies have investigated the transgenerational impacts of maternal and parental obesogenic diets on subsequent generations of offspring, but results have largely been mixed. We conducted a systematic review and meta-analysis on rodent studies to elucidate how obesogenic diets impact the mean and variance of grand-offspring traits. Our study focused on transgenerational effects (i.e., F2 and F3 generations) in one-off and multigenerational exposure studies. From 33 included articles, we obtained 407 effect sizes representing pairwise comparisons of control and treatment grand-offspring groups pertaining to measures of body weight, adiposity, glucose, insulin, leptin, and triglycerides. We found evidence that male and female grand-offspring descended from grandparents exposed to an obesogenic diet displayed phenotypes consistent with metabolic syndrome, especially in cases where the obesogenic diet was continued across generations. Further, we found stronger evidence for the effects of grand-maternal than grand-paternal exposure on grand-offspring traits. A high-fat diet in one-off exposure studies did not seem to impact phenotypic variation, whereas in multigenerational exposure studies it reduced variation in several traits.
Collapse
Affiliation(s)
- Hamza Anwer
- Evolution and Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Margaret J Morris
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Daniel W A Noble
- Evolution and Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
- Division of Ecology and Evolution, Research School of Biology, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Shinichi Nakagawa
- Evolution and Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Malgorzata Lagisz
- Evolution and Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
48
|
Grzęda E, Matuszewska J, Ziarniak K, Gertig-Kolasa A, Krzyśko- Pieczka I, Skowrońska B, Sliwowska JH. Animal Foetal Models of Obesity and Diabetes - From Laboratory to Clinical Settings. Front Endocrinol (Lausanne) 2022; 13:785674. [PMID: 35197931 PMCID: PMC8858803 DOI: 10.3389/fendo.2022.785674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/11/2022] [Indexed: 12/26/2022] Open
Abstract
The prenatal period, during which a fully formed newborn capable of surviving outside its mother's body is built from a single cell, is critical for human development. It is also the time when the foetus is particularly vulnerable to environmental factors, which may modulate the course of its development. Both epidemiological and animal studies have shown that foetal programming of physiological systems may alter the growth and function of organs and lead to pathology in adulthood. Nutrition is a particularly important environmental factor for the pregnant mother as it affects the condition of offspring. Numerous studies have shown that an unbalanced maternal metabolic status (under- or overnutrition) may cause long-lasting physiological and behavioural alterations, resulting in metabolic disorders, such as obesity and type 2 diabetes (T2DM). Various diets are used in laboratory settings in order to induce maternal obesity and metabolic disorders, and to alter the offspring development. The most popular models are: high-fat, high-sugar, high-fat-high-sugar, and cafeteria diets. Maternal undernutrition models are also used, which results in metabolic problems in offspring. Similarly to animal data, human studies have shown the influence of mothers' diets on the development of children. There is a strong link between the maternal diet and the birth weight, metabolic state, changes in the cardiovascular and central nervous system of the offspring. The mechanisms linking impaired foetal development and adult diseases remain under discussion. Epigenetic mechanisms are believed to play a major role in prenatal programming. Additionally, sexually dimorphic effects on offspring are observed. Therefore, further research on both sexes is necessary.
Collapse
Affiliation(s)
- Emilia Grzęda
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
| | - Julia Matuszewska
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
| | - Kamil Ziarniak
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
- Molecular and Cell Biology Unit, Poznań University of Medical Sciences, Poznań, Poland
| | - Anna Gertig-Kolasa
- Department of Paediatric Diabetes and Obesity, Poznań University of Medical Sciences, Poznań, Poland
| | - Izabela Krzyśko- Pieczka
- Department of Paediatric Diabetes and Obesity, Poznań University of Medical Sciences, Poznań, Poland
| | - Bogda Skowrońska
- Department of Paediatric Diabetes and Obesity, Poznań University of Medical Sciences, Poznań, Poland
| | - Joanna H. Sliwowska
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
- *Correspondence: Joanna H. Sliwowska,
| |
Collapse
|
49
|
Incollingo Rodriguez AC, Nagpal TS. The WOMBS Framework: A review and new theoretical model for investigating pregnancy-related weight stigma and its intergenerational implications. Obes Rev 2021; 22:e13322. [PMID: 34288364 DOI: 10.1111/obr.13322] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/30/2021] [Indexed: 11/30/2022]
Abstract
As the growing weight stigma literature has developed, one critically relevant and vulnerable population has received little consideration-pregnant and postpartum women. Because weight fluctuations are inherent to this life phase, and rates of prepregnancy overweight and obesity are already high, this gap is problematic. More recently, however, there has been a rising interest in pregnancy-related weight stigma and its consequences. This paper therefore sought to (a) review the emerging research on pregnancy-related weight stigma phenomenology and (b) integrate this existing evidence to present a novel theoretical framework for studying pregnancy-related weight stigma. The Weight gain, Obesity, Maternal-child Biobehavioral pathways, and Stigma (WOMBS) Framework proposes psychophysiological mechanisms linking pregnancy-related weight stigmatization to increased risk of weight gain and, in turn, downstream childhood obesity risk. This WOMBS Framework highlights pregnant and postpartum women as a theoretically unique at-risk population for whom this social stigma engages maternal physiology and transfers obesity risk to the child via social and physiological mechanisms. The WOMBS Framework provides a novel and useful tool to guide the emerging pregnancy-related weight stigma research and, ultimately, support stigma-reduction efforts in this critical context.
Collapse
Affiliation(s)
- Angela C Incollingo Rodriguez
- Psychological & Cognitive Sciences, Department of Social Science & Policy Studies, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Taniya S Nagpal
- Department of Kinesiology, Faculty of Applied Health Sciences, Brock University, St. Catharines, Ontario, Canada
| |
Collapse
|
50
|
Imprinted lncRNA Dio3os preprograms intergenerational brown fat development and obesity resistance. Nat Commun 2021; 12:6845. [PMID: 34824246 PMCID: PMC8617289 DOI: 10.1038/s41467-021-27171-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 11/08/2021] [Indexed: 12/17/2022] Open
Abstract
Maternal obesity (MO) predisposes offspring to obesity and metabolic disorders but little is known about the contribution of offspring brown adipose tissue (BAT). We find that MO impairs fetal BAT development, which persistently suppresses BAT thermogenesis and primes female offspring to metabolic dysfunction. In fetal BAT, MO enhances expression of Dio3, which encodes deiodinase 3 (D3) to catabolize triiodothyronine (T3), while a maternally imprinted long noncoding RNA, Dio3 antisense RNA (Dio3os), is inhibited, leading to intracellular T3 deficiency and suppression of BAT development. Gain and loss of function shows Dio3os reduces D3 content and enhances BAT thermogenesis, rendering female offspring resistant to high fat diet-induced obesity. Attributing to Dio3os inactivation, its promoter has higher DNA methylation in obese dam oocytes which persists in fetal and adult BAT, uncovering an oocyte origin of intergenerational obesity. Overall, our data uncover key features of Dio3os activation in BAT to prevent intergenerational obesity and metabolic dysfunctions. Maternal obesity predisposes offspring to obesity and metabolic disorders through incompletely understood mechanisms. Here the authors report that Dio3os is an imprinted long-coding RNA that modulates brown adipose tissue development and obesity resistance in the offspring.
Collapse
|