1
|
Takeoka E, Carlson AA, Madan N, Azimirad A, Mahmoud T, Kitano R, Akiyama S, Yun HJ, Tucker R, Im K, O'Tierney-Ginn P, Tarui T. Impact of high maternal body mass index on fetal cerebral cortical and cerebellar volumes. J Perinat Med 2025:jpm-2024-0222. [PMID: 39754513 DOI: 10.1515/jpm-2024-0222] [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: 05/14/2024] [Accepted: 11/27/2024] [Indexed: 01/06/2025]
Abstract
OBJECTIVES Maternal obesity increases a child's risk of neurodevelopmental impairment. However, little is known about the impact of maternal obesity on fetal brain development. METHODS We prospectively recruited 20 healthy pregnant women across the range of pre-pregnancy or first-trimester body mass index (BMI) and performed fetal brain magnetic resonance imaging (MRI) of their healthy singleton fetuses. We examined correlations between early pregnancy maternal BMI and regional brain volume of living fetuses using volumetric MRI analysis. RESULTS Of 20 fetuses, there were 8 males and 12 females (median gestational age at MRI acquisition was 24.3 weeks, range: 19.7-33.3 weeks, median maternal age was 33.3 years, range: 22.0-37.4 years). There were no significant differences in clinical demographics between overweight (OW, 25≤BMI<30)/obese (OB, BMI≥30 kg/m2) (n=12) and normal BMI (18.5≤BMI<25) (n=8) groups. Fetuses in the OW/OB group had significantly larger left cortical plate (p=0.0003), right cortical plate (p=0.0002), and whole cerebellum (p=0.049) compared to the normal BMI group. In the OW/OB BMI group, cortical plate volume was larger relative to other brain regions after 28 weeks. CONCLUSIONS This pilot study supports the concept that maternal obesity impacts fetal brain volume, detectable via MRI in living fetuses using quantitative analysis.
Collapse
Affiliation(s)
- Emiko Takeoka
- Tufts Medical Center, Mother Infant Research Institute, Boston, MA, USA
- Department of Neonatology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Hyogo, Japan
| | - April A Carlson
- Tufts Medical Center, Mother Infant Research Institute, Boston, MA, USA
- Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Neel Madan
- Department of Radiology, Mass General Brigham, Boston, MA, USA
| | - Afshin Azimirad
- Tufts Medical Center, Mother Infant Research Institute, Boston, MA, USA
- Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA, USA
| | - Taysir Mahmoud
- Tufts Medical Center, Mother Infant Research Institute, Boston, MA, USA
| | - Rie Kitano
- Tufts Medical Center, Mother Infant Research Institute, Boston, MA, USA
- Department of Obstetrics and Gynecology, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaragi, Japan
| | - Shizuko Akiyama
- Tufts Medical Center, Mother Infant Research Institute, Boston, MA, USA
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Hyuk Jin Yun
- Fetal-Neonatal Neuroimaging Developmental Science Center, Boston Children's Hospital, Boston, MA, USA
| | - Richard Tucker
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Kiho Im
- Fetal-Neonatal Neuroimaging Developmental Science Center, Boston Children's Hospital, Boston, MA, USA
| | | | - Tomo Tarui
- Tufts Medical Center, Mother Infant Research Institute, Boston, MA, USA
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Providence, RI, USA
- Pediatric Neurology, Hasbro Children's, Providence, RI, USA
| |
Collapse
|
2
|
Ionescu MI, Zahiu CDM, Vlad A, Galos F, Gradisteanu Pircalabioru G, Zagrean AM, O'Mahony SM. Nurturing development: how a mother's nutrition shapes offspring's brain through the gut. Nutr Neurosci 2025; 28:50-72. [PMID: 38781488 DOI: 10.1080/1028415x.2024.2349336] [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: 05/25/2024]
Abstract
Pregnancy is a transformative period marked by profound physical and emotional changes, with far-reaching consequences for both mother and child. Emerging research has illustrated the pivotal role of a mother's diet during pregnancy in influencing the prenatal gut microbiome and subsequently shaping the neurodevelopment of her offspring. The intricate interplay between maternal gut health, nutrition, and neurodevelopmental outcomes has emerged as a captivating field of investigation within developmental science. Acting as a dynamic bridge between mother and fetus, the maternal gut microbiome, directly and indirectly, impacts the offspring's neurodevelopment through diverse pathways. This comprehensive review delves into a spectrum of studies, clarifying putative mechanisms through which maternal nutrition, by modulating the gut microbiota, orchestrates the early stages of brain development. Drawing insights from animal models and human cohorts, this work underscores the profound implications of maternal gut health for neurodevelopmental trajectories and offers a glimpse into the formulation of targeted interventions able to optimize the health of both mother and offspring. The prospect of tailored dietary recommendations for expectant mothers emerges as a promising and accessible intervention to foster the growth of beneficial gut bacteria, potentially leading to enhanced cognitive outcomes and reduced risks of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Mara Ioana Ionescu
- Department of Functional Sciences, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Pediatrics, Marie Curie Emergency Children's Hospital, Bucharest, Romania
| | - Carmen Denise Mihaela Zahiu
- Department of Functional Sciences, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Adelina Vlad
- Department of Functional Sciences, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Felicia Galos
- Department of Pediatrics, Marie Curie Emergency Children's Hospital, Bucharest, Romania
- Department of Pediatrics, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Gratiela Gradisteanu Pircalabioru
- Research Institute of the University of Bucharest, Section Earth, Environmental and Life Sciences, Section-ICUB, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
| | - Ana-Maria Zagrean
- Department of Functional Sciences, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Siobhain M O'Mahony
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
3
|
Guma E, Chakravarty MM. Immune Alterations in the Intrauterine Environment Shape Offspring Brain Development in a Sex-Specific Manner. Biol Psychiatry 2025; 97:12-27. [PMID: 38679357 PMCID: PMC11511788 DOI: 10.1016/j.biopsych.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Exposure to immune dysregulation in utero or in early life has been shown to increase risk for neuropsychiatric illness. The sources of inflammation can be varied, including acute exposures due to maternal infection or acute stress, or persistent exposures due to chronic stress, obesity, malnutrition, or autoimmune diseases. These exposures may cause subtle alteration in brain development, structure, and function that can become progressively magnified across the lifespan, potentially increasing the likelihood of developing a neuropsychiatric conditions. There is some evidence that males are more susceptible to early-life inflammatory challenges than females. In this review, we discuss the various sources of in utero or early-life immune alteration and the known effects on fetal development with a sex-specific lens. To do so, we leveraged neuroimaging, behavioral, cellular, and neurochemical findings. Gaining clarity about how the intrauterine environment affects offspring development is critically important for informing preventive and early intervention measures that may buffer against the effects of these early-life risk factors.
Collapse
Affiliation(s)
- Elisa Guma
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, Maryland; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.
| | - M Mallar Chakravarty
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
4
|
Samà M, Musillo C, Cirulli F. Counteracting the effects of maternal obesity on offspring neurodevelopment through Omega-3-based nutritional strategies. Neuroscience 2024; 566:142-148. [PMID: 39722288 DOI: 10.1016/j.neuroscience.2024.12.041] [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: 10/01/2024] [Revised: 12/09/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
It is becoming increasingly recognized that, in addition to psychological stress, unbalanced maternal nutritional habits can threaten fetal brain development. Maternal obesity is one of the most pressing public health problems facing the world today, as about 40% of pregnant women are obese or gain excessive weight worldwide. This condition can negatively impact offspring's brain development, increasing the risk for autism spectrum disorders, cognitive deficits, attention deficit hyperactivity disorder, as well as anxiety and depression. In the context of fetal development, nutritional interventions may represent a feasible and safe approach for preventing the negative effects of maternal obesity. We argue that maternal Omega-3 supplementation, among the many dietary strategies available, is especially promising as it buffers oxidative stress and inflammation, both recognized as candidate mechanisms underlying the negative long-term effects of maternal obesity on the offspring. Notwithstanding the current knowledge, both preclinical studies and clinical trials are needed to refine current strategies addressing dietary content and length of administration according to individual characteristics and needs.
Collapse
Affiliation(s)
- Marianna Samà
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299 00161, Rome, Italy
| | - Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299 00161, Rome, Italy.
| | - Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299 00161, Rome, Italy
| |
Collapse
|
5
|
Mahjubi K, Effatpanah M, Parsaei M, Mojtahedi SY, Izadi A, Tavakolizadeh R. Association of anthropometric parameters with intelligence quotient in early school-aged children: a cross-sectional study in Iran. BMC Pediatr 2024; 24:809. [PMID: 39696032 DOI: 10.1186/s12887-024-05303-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND While the link between body growth indices in the first three years of life and neurodevelopment is well established, the relationship between these indicators and intelligence levels in later stages is not well understood. This study aimed to investigate the association between various anthropometric indices and Intelligence Quotient in early school-aged children. METHODS This cross-sectional study recruited healthy students aged 7 to 8 years from four primary schools in Tehran, Iran. Measurements included body weight, height, body mass index, arm circumference, waist circumference, and hip circumference. Intelligence Quotient was assessed using the Raven's Standard Progressive Matrices Test, with scores standardized by age according to the Raven's manual. Separate univariate linear regression analyses evaluated the association between each anthropometric index and children's Intelligence Quotient scores. Multivariate linear regression analyses were then performed to adjust for potential confounding factors, including the child's sex, age, and parental education levels. RESULTS A cohort of 160 children (80 females), with a mean age of 7.5 ± 0.6 years, was included in the study. The participants exhibited the following mean anthropometric values: weight 27.7 ± 6.5 kg, height 1.2 ± 0.1 m, body mass index 17.9 ± 3.4 kg/m2, arm circumference 20.7 ± 2.8 cm, waist circumference 56.4 ± 6.4 cm, and hip circumference 68.1 ± 6.6 cm. Their mean Intelligence Quotient score was 106 ± 16.6. In the univariate analysis, children's weight was not significantly associated with Intelligence Quotient score (P-value = 0.153, β = 0.288 [-0.108, 0.684]), while height was negatively associated (P-value = 0.048, β = -31.685 [-63.142, -0.228]) and body mass index showed a positive association (P-value = 0.001, β = 1.265 [0.524, 2.006]). Abdominal circumference was not significantly associated with Intelligence Quotient (P-value = 0.913, β = 0.051 [-0.870, 0.972]), but both waist circumference (P-value = 0.007, β = 0.542 [0.147, 0.937]) and hip circumference (P-value = 0.013, β = 0.484 [0.102, 0.866]) demonstrated significant positive associations with Intelligence Quotient. However, none of the anthropometric indices maintained statistically significant associations with Intelligence Quotient after adjusting for potential confounders (P-values > 0.05). CONCLUSIONS This study indicated no strong relationship between various body measurements and intelligence levels in school-aged children after adjusting the analyses for confounders. This suggests that intelligence in these children may be largely shaped by genetic and sociodemographic factors, with minimal influence from physical growth patterns.
Collapse
Affiliation(s)
- Kimia Mahjubi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Effatpanah
- Department of Psychiatry, Zaieian Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadamin Parsaei
- Breastfeeding Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sayed Yousef Mojtahedi
- Department of Pediatric Nephrology, Tehran University of Medical Sciences, Tehran, Iran
- Pediatric Chronic Kidney Disease Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Anahita Izadi
- Department of Pediatric Infectious Disease, Tehran University of Medical Science, Tehran, Iran
| | - Reza Tavakolizadeh
- Department of Pediatric Endocrinology and Metabolism, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Gutiérrez MC, Perondi MC, Tortoni GL, Cragnolini AB, Cuadra GR, Valdomero A. Early protein restriction in rats induces anhedonia in adult offspring: A key role of BDNF-TrkB signaling in the nucleus accumbens shell. Neuropharmacology 2024; 258:110099. [PMID: 39098656 DOI: 10.1016/j.neuropharm.2024.110099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
Clinical evidence suggests that early malnutrition promotes symptoms related to psychiatric disorders later in life. Nevertheless, the molecular mechanisms underpinning nutritional injury induce depression remains unknown. The purpose of the present study was to evaluate whether perinatal protein restriction increases vulnerability to developing depressive-like behavior in adulthood by focusing on anhedonia, a core symptom of depression. To this, male adult Wistar rats submitted to a protein restriction schedule at perinatal age (PR-rats), were subjected to the sucrose preference test (SPT), the novel object recognition test (NORT), the forced swim test (FST), and the elevated plus maze (EPM), and compared to animals fed with a normoprotein diet. To investigate neurobiological substrates linked to early protein undernutrition-facilitated depressive-like behavior, we assessed the levels of brain-derived neurotrophic factor (BDNF) and its receptor TrkB in the nucleus accumbens (NAc), and evaluated the reversal of anhedonic-like behavior by infusing ANA-12. We found that early malnutrition decreased sucrose preference, impaired performance in the NORT and increased immobility time in the FST. Furthermore, perinatal protein-restriction-induced anhedonia correlated with increased BDNF and p-TrkB protein levels in the NAc, a core structure in the reward circuit linked with anhedonia. Finally, bilateral infusion of the TrkB antagonist ANA-12 into the NAc shell ameliorated a reduced sucrose preference in the PR-rats. Altogether, these findings revealed that protein restriction during pregnancy and lactation facilitates depressive-like behavior later in life and may increase the risk of developing anhedonia by altering BDNF-TrkB in the NAc shell.
Collapse
Affiliation(s)
- María C Gutiérrez
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Córdoba, Argentina; Instituto de Farmacología Experimental Córdoba (IFEC - CONICET), Córdoba, Argentina
| | - María C Perondi
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| | - Gisella L Tortoni
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| | - Andrea B Cragnolini
- Instituto de Investigaciones Biológicas y Tecnológicas (IIByT - CONICET), Córdoba, Argentina
| | - Gabriel R Cuadra
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Córdoba, Argentina; Instituto de Farmacología Experimental Córdoba (IFEC - CONICET), Córdoba, Argentina
| | - Analía Valdomero
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Córdoba, Argentina; Instituto de Farmacología Experimental Córdoba (IFEC - CONICET), Córdoba, Argentina.
| |
Collapse
|
7
|
Huang YS, Hung PL, Wang LJ, Tsai CM, Tsai CK, Tiao MM, Yu HR. Distinct Impacts of Prenatal and Postnatal Phthalate Exposure on Behavioral and Emotional Development in Children Aged 1.5 to 3 Years. TOXICS 2024; 12:795. [PMID: 39590974 PMCID: PMC11598217 DOI: 10.3390/toxics12110795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024]
Abstract
Development is a continuous process, but few studies have assessed the simultaneous impact of prenatal and postnatal phthalate exposure on children's behavioral and emotional development. A total of 491 mother-child pairs from the general population in southern Taiwan were studied from 2021 to 2022. Urinary concentrations of bisphenol A (BPA) and phthalate metabolites-mono-ethyl phthalate (MEP), mono-n-butyl phthalate (MnBP), mono-benzyl phthalate (MBzP), and mono-2-ethylhexyl phthalate (MEHP)-were measured in pregnant mothers during the second trimester and in their corresponding children aged 1.5 to 3 years. Behavioral symptoms in children were evaluated using the Child Behavior Checklist (CBCL). Odds ratios (ORs) represent a 1-unit increase in log10-transformed creatinine-corrected maternal urine concentrations. Prenatal maternal urinary MnBP levels were associated with total problems (OR = 19.32, 95% CI: 1.80-43.13, p = 0.04), anxiety (OR = 33.58, 95% CI: 2.16-521.18, p = 0.01), and sleep problems (OR = 41.34, 95% CI: 1.04-1632.84, p = 0.04) in children. Additionally, urinary MnBP levels in children correlated with total problems (OR = 7.06, 95% CI: 1.01-49.05, p = 0.04) and internalizing problems (OR = 11.04, 95% CI: 1.27-95.72, p = 0.01). These findings suggest that prenatal and postnatal exposure to dibutyl phthalate (DBP), metabolized as MnBP, distinctly affects children's behavioral development.
Collapse
Affiliation(s)
- Yi-Siang Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (Y.-S.H.)
| | - Pi-Lien Hung
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (Y.-S.H.)
| | - Liang-Jen Wang
- Department of Child and Adolescent Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Chih-Min Tsai
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (Y.-S.H.)
| | - Chang-Ku Tsai
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (Y.-S.H.)
| | - Mao-Meng Tiao
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (Y.-S.H.)
| | - Hong-Ren Yu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (Y.-S.H.)
- College of Medicine, Chang Gung University, Taoyuan 330, Taiwan
- Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| |
Collapse
|
8
|
Walker CG, Marks E, Fletcher B, Thayer Z, Cha JE, Teng Y, Evans R, Waldie KE. Prenatal determinants of anxiety symptoms in middle childhood. Evidence from Growing Up in New Zealand. J Affect Disord 2024; 363:653-661. [PMID: 39043309 DOI: 10.1016/j.jad.2024.07.114] [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: 12/14/2023] [Revised: 06/26/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Early life environments can have long-lasting impacts on future health and wellbeing. Maternal health during pregnancy, including experiencing stress or mood disorders, has been associated with psychopathology in later life. Anxiety disorders are one of the most prevalent mental health conditions, affecting approximately 7 % of children and adolescents globally, with a lifetime prevalence of 15-20 %. Identifying prenatal risk factors can support future and current public health interventions and maternity care. METHODS Data were obtained from the Growing Up in New Zealand longitudinal study of child development. Prenatally, mothers provided sociodemographic information as well as data on their mental health, potential teratogens, and lifestyle factors such as supplement intake and exercise levels. At 8-years old, 4922 children self-completed the PROMIS-SF anxiety measure. Bivariate analyses and backward stepwise regression were used to determine the best multivariable model. RESULTS Significant prenatal predictors of anxiety symptoms at 8-years old included elevated maternal depression symptoms, body mass index in the overweight/obese range, exercise patterns, and paracetamol, anti-inflammatory and alcohol intake. LIMITATIONS Sample attrition from baseline to 8-year may have affected statistical power. To further untangle the effect of timing and duration of the exposures reported in this study, larger sample sizes would be required. CONCLUSIONS Prenatal mental health and wellbeing was significantly associated with child anxiety symptoms at 8-years of age. This study highlights the importance of supporting expectant mothers' health and wellbeing during pregnancy to ensure children have the best opportunity to have good mental health.
Collapse
Affiliation(s)
- Caroline G Walker
- Centre for Longitudinal Research - He Ara ki Mua, The University of Auckland, Auckland, New Zealand; School of Population Health, The University of Auckland, Auckland, New Zealand.
| | - Emma Marks
- Centre for Longitudinal Research - He Ara ki Mua, The University of Auckland, Auckland, New Zealand; School of Population Health, The University of Auckland, Auckland, New Zealand
| | - Ben Fletcher
- Centre for Longitudinal Research - He Ara ki Mua, The University of Auckland, Auckland, New Zealand; School of Population Health, The University of Auckland, Auckland, New Zealand
| | - Zaneta Thayer
- Dartmouth College, Hanover, NH, United States of America
| | - Jane E Cha
- Centre for Longitudinal Research - He Ara ki Mua, The University of Auckland, Auckland, New Zealand; School of Psychological Medicine, The University of Auckland, Auckland, New Zealand
| | - Yin Teng
- School of Psychology, The University of Auckland, Auckland, New Zealand
| | - Rebecca Evans
- Centre for Longitudinal Research - He Ara ki Mua, The University of Auckland, Auckland, New Zealand; School of Population Health, The University of Auckland, Auckland, New Zealand
| | - Karen E Waldie
- Centre for Longitudinal Research - He Ara ki Mua, The University of Auckland, Auckland, New Zealand; School of Psychology, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
9
|
Ratsika A, Codagnone MG, Bastiaanssen TFS, Hoffmann Sarda FA, Lynch CMK, Ventura-Silva AP, Rosell-Cardona C, Caputi V, Stanton C, Fülling C, Clarke G, Cryan JF. Maternal high-fat diet-induced microbiota changes are associated with alterations in embryonic brain metabolites and adolescent behaviour. Brain Behav Immun 2024; 121:317-330. [PMID: 39032541 DOI: 10.1016/j.bbi.2024.07.020] [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: 02/23/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
The developing central nervous system is highly sensitive to nutrient changes during the perinatal period, emphasising the potential impact of alterations of maternal diet on offspring brain development and behaviour. A growing body of research implicates the gut microbiota in neurodevelopment and behaviour. Maternal overweight and obesity during the perinatal period has been linked to changes in neurodevelopment, plasticity and affective disorders in the offspring, with implications for microbial signals from the maternal gut. Here we investigate the impact of maternal high-fat diet (mHFD)-induced changes in microbial signals on offspring brain development, and neuroimmune signals, and the enduring effects on behaviour into adolescence. We first demonstrate that maternal caecal microbiota composition at term pregnancy (embryonic day 18: E18) differs significantly in response to maternal diet. Moreover, mHFD resulted in the upregulation of microbial genes in the maternal intestinal tissue linked to alterations in quinolinic acid synthesis and elevated kynurenine levels in the maternal plasma, both neuronal plasticity mediators related to glutamate metabolism. Metabolomics of mHFD embryonic brains at E18 also detected molecules linked to glutamate-glutamine cycle, including glutamic acid, glutathione disulphide, and kynurenine. During adolescence, the mHFD offspring exhibited increased locomotor activity and anxiety-like behaviour in a sex-dependent manner, along with upregulation of glutamate-related genes compared to controls. Overall, our results demonstrate that maternal exposure to high-fat diet results in microbiota changes, behavioural imprinting, altered brain metabolism, and glutamate signalling during critical developmental windows during the perinatal period.
Collapse
Affiliation(s)
- Anna Ratsika
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Martin G Codagnone
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Thomaz F S Bastiaanssen
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Fabiana A Hoffmann Sarda
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Caoimhe M K Lynch
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Ana Paula Ventura-Silva
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | - Cristina Rosell-Cardona
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | - Valentina Caputi
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | | | - Christine Fülling
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork T12YT20, Ireland
| | - John F Cryan
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland.
| |
Collapse
|
10
|
Nilsson IAK, Ozsvar J, Gissler M, Lavebratt C. Maternal Eating Disorders, Body Mass Index, and Offspring Psychiatric Diagnoses. JAMA Netw Open 2024; 7:e2440517. [PMID: 39436646 PMCID: PMC11581519 DOI: 10.1001/jamanetworkopen.2024.40517] [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: 05/23/2024] [Accepted: 08/28/2024] [Indexed: 10/23/2024] Open
Abstract
Importance Maternal nutrition is essential in fetal development; thus, disordered eating may influence this process and contribute to the development of offspring psychiatric disorders. Objective To investigate the association of maternal eating disorders and prepregnancy body mass index (BMI) with offspring psychiatric diagnoses. Design, Setting, and Participants This population-based cohort study used Finnish national registers to assess all live births from January 1, 2004, through December 31, 2014, with follow-up until December 31, 2021. The data analyses were conducted from September 1, 2023, to September 30, 2024. Exposures Maternal eating disorder and prepregnancy BMI. Main Outcomes and Measures Primary outcomes were 9 neurodevelopmental and psychiatric offspring diagnoses. Cox proportional hazards modeling adjusted for potential risk factors in the development of the outcome disorders was applied in 2 models. Secondary analyses were stratified for adverse birth outcomes (prematurity, small size for gestational age, and low Apgar score) or comorbid offspring eating disorders. Categories of BMI (calculated as weight in kilograms divided by height in meters squared) included underweight (BMI <18.5), normal weight (18.5-24.9), overweight (25.0-29.9), obesity (30.0-34.9), and severe obesity (≥35.0). Results The mean (SD) age of 392 098 included mothers was 30.15 (5.38) years, 42 590 mothers (10.86%) were born outside of Finland, 6273 mothers (1.60%) had a history of an eating disorder, 23 114 mothers (5.89%) had prepregnancy underweight, and 208 335 (53.13%) mothers had overweight or obesity. Among 649 956 included offspring, 332 359 (51.14%) were male, and 106 777 (16.43%) had received a neurodevelopmental or psychiatric diagnosis. Maternal eating disorders, prepregnancy underweight, and overweight or obesity were associated with most of the studied mental diagnoses in offspring, even after adjusting for potential covariates. The largest effect sizes were observed for maternal eating disorders not otherwise specified in association with offspring sleep disorders (hazard ratio [HR], 3.34 [95% CI, 2.39-4.67]) and social functioning and tic disorders (HR, 2.79 [95% CI, 2.21-3.52]), while for maternal severe prepregnancy obesity, offspring intellectual disabilities (HR, 2.04 [95% CI, 1.83-2.28]) had the largest effect size. Adverse birth outcomes further increased the risk of offspring having other feeding disturbances of childhood and infancy (eg, HR, 4.53 [95% CI, 2.97-6.89] for maternal eating disorders) and attention-deficit/hyperactivity disorder and conduct disorder (eg, HR, 2.27 [95% CI, 1.74-2.96] for maternal anorexia nervosa). Conclusions and Relevance In this population-based cohort study including 392 098 mothers and 649 956 offspring, offspring from mothers with an eating disorder history or prepregnancy BMI outside normal weight were at higher risk of psychiatric disorders. The results differed somewhat between the 2 exposures with regard to which offspring diagnoses had associations, and effect sizes were typically larger for maternal eating disorders vs BMI. These findings suggest a need to consider these 2 exposures clinically to help prevent offspring mental illness.
Collapse
Affiliation(s)
- Ida A. K. Nilsson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
- Centre for Eating Disorders Innovation, Karolinska Institutet, Stockholm, Sweden
| | - Judit Ozsvar
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Mika Gissler
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Knowledge Brokers, Finnish Institute for Health and Welfare, Helsinki, Finland
- Research Centre for Child Psychiatry, University of Turku, Turku, Finland
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
11
|
Ben Messaoud K, Khachadourian V, Arildskov E, Hansen SN, Gardner R, Ramlau-Hansen C, Kahn L, Janecka M. Female Infertility and Neurodevelopmental Disorders in Children: associations and evidence for familial confounding in Denmark. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.17.24313638. [PMID: 39371166 PMCID: PMC11451697 DOI: 10.1101/2024.09.17.24313638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
IMPORTANCE Existing research suggests the impact of infertility on the risk of neurodevelopmental disorders in children, however, studies to date have failed to separate the impact of male and female infertility, often blurring the lines with proxies that encompass all forms of infertility. Moreover, while both health conditions co-occurring with infertility and genetic factors operating upstream have been suggested to influence the association between infertility and child outcomes, their roles and potential impact on observed associations remain unclear. OBJECTIVE The objectives of this study are to investigate the relationship between female infertility and autism in the child, differentiating it from the effects of male and the couple infertility; consider the role of various maternal and birth factors in the association; and examine the effects of shared familial confounders on the association. DESIGN SETTING AND PARTICIPANTS Danish population-based cohort study, including all singleton live births in Denmark 1998-2015, their parents and parents' siblings. The cohort was followed up until December 31, 2016. EXPOSURES The exposure was a history of female infertility in the mother and the mother's sister. We examined four definitions of female infertility based on the ICD-10 codes derived from the Danish National Patient Register - any female infertility; specified female infertility; female exclusive infertility; and female or male infertility. MAIN OUTCOME AND MEASURES The outcome was diagnosis of autism spectrum disorder (ASD) in the Danish Psychiatric Central Research Register or the national patient register. A multivariable Cox regression model was used to estimate the associations between female infertility and autism, accounting for child's sex, year of birth, maternal age, education level, chronic comorbidities, and pregnancy and birth complications. The effects of shared familial factors on the association were analyzed using exposure information from the child's maternal aunt. RESULTS The cohort included 1,131,899 mother-child pairs, among which 18,374 children with ASD diagnosis. History of female infertility in the mother (all definitions) was significantly associated with autism in the child, with the association remaining robust after adjustment for covariates (HRadj=1.14 (95% CI, 1.03-1.26) for specified infertility). The diagnosis of infertility in a child's maternal aunt was also significantly linked to the child's autism risk, even after adjustment for maternal infertility (HRadj=1.10 (95% CI, 1.00-1.20). CONCLUSIONS AND RELEVANCE in This population-based birth cohort study, we found a slightly higher risk of autism in children born to mothers with a history of infertility, with the association remaining consistent across various definitions of female infertility and robust to adjustments for demographic, child, and maternal factors. The study suggests for the first time that shared familial factors, possibly both genetic and non-genetic, could be influencing both female infertility and the risk of autism in children, indicating a need for further investigation into these familial effects.
Collapse
Affiliation(s)
- Khaoula Ben Messaoud
- Department of Child and Adolescent Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
| | - Vahe Khachadourian
- Department of Child and Adolescent Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
| | - Elias Arildskov
- Department of Biomedecine, Aarhus University, Aarhus, Denmark
| | - Stefan N Hansen
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Renee Gardner
- Department of Public Health, Aarhus University, Aarhus, Denmark
| | | | - Linda Kahn
- Department of Pediatrics, New York University School of Medicine, New York, NY, USA
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Magdalena Janecka
- Department of Child and Adolescent Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
12
|
Cho NA, Giesbrecht GF, Dewey D, Reimer RA. Early Life Surgency, but not Effortful Control or Negative Affectivity, Is a Mediating Variable Between Maternal Pre-Pregnancy Body Mass Index and Childhood Obesity Risk. Dev Psychobiol 2024; 66:e22517. [PMID: 38924077 DOI: 10.1002/dev.22517] [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: 07/13/2023] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024]
Abstract
Maternal gestational obesity is related to risk of obesity in the child. This risk may be in part mediated by altered child temperament, which can affect mother-child interactions, including feeding and soothing behaviors that affect obesity risk. Our objective was to examine the association between maternal pre-pregnancy BMI and child zBMI and determine if child temperament, specifically positive Affectivity/Surgency, mediates this association. Using conditional process modeling, we analyzed data from 408 mother-child dyads enrolled in the Alberta Pregnancy Outcomes and Nutrition (APrON) study. Child temperament was assessed at 3 years of age via a parent report measure, the Child Behavior Questionnaire (CBQ), and child zBMI was calculated from in-person measurements of child height and weight at 4-5 years of age. Bivariate correlations showed that there was a significant positive correlation between zBMI and Surgency (r = 0.11, p = 0.03), and zBMI was also correlated with maternal pre-pregnancy BMI (r = 0.12, p = 0.02). Multivariable regression revealed that maternal pre-pregnancy BMI (adjusted β = 0.15, 95% confidence interval [CI]; 0.00-0.05, p = 0.02) and Surgency scores (adjusted β = 0.14, 95% CI; 0.02-0.28, p = 0.03) were associated with higher child zBMI at 4-5 years of age. Mediation analysis showed that Surgency mediated the association between pre-pregnancy BMI and child zBMI. Our models controlled for maternal gestational weight gain, gestational diabetes, socioeconomic status, maternal anxiety and depression, and gestational age at birth. Overall, maternal pre-pregnancy BMI was positively associated with child zBMI, and this association was mediated by higher child Surgency scores.
Collapse
Affiliation(s)
- Nicole A Cho
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Gerald F Giesbrecht
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute (ACHRI), University of Calgary, Calgary, Alberta, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Deborah Dewey
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute (ACHRI), University of Calgary, Calgary, Alberta, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, Alberta, Canada
| | - Raylene A Reimer
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute (ACHRI), University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, Calgary, Alberta, Canada
| |
Collapse
|
13
|
Cattaneo A, Begni V, Zonca V, Riva MA. Early life adversities, psychopathologies and novel pharmacological strategies. Pharmacol Ther 2024; 260:108686. [PMID: 38969307 DOI: 10.1016/j.pharmthera.2024.108686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/05/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Exposure to adversities during early life stages (early life adversities - ELA), ranging from pregnancy to adolescence, represents a major risk factor for the vulnerability to mental disorders. Hence, it is important to understand the molecular and functional underpinning of such relationship, in order to develop strategies aimed at reducing the psychopathologic burden associated with ELA, which may eventually lead to a significant improvement in clinical practice. In this review, we will initially recapitulate clinical and preclinical evidence supporting the link between ELA and psychopathology and we will primarily discuss the main biological mechanisms that have been described as potential mediators of the effects of ELA on the psychopathologic risk, including the role for genetic factors as well as sex differences. The knowledge emerging from these studies may be instrumental for the development of novel therapeutic strategies aimed not only at correcting the deficits that emerge from ELA exposure, but also in preventing the manifestation of a full-blown psychopathologic condition. With this respect, we will specifically focus on adolescence as a key time frame for disease onset as well as for early therapeutic intervention. We believe that incorporating clinical and preclinical research data in the context of early life adversities can be instrumental to elucidate the mechanisms contributing to the risk for psychopathology or that may promote resilience. This will ultimately allow the identification of 'at risk' individuals who may benefit from specific forms of interventions that, by interfering with disease trajectories, could result in more benign clinical outcomes.
Collapse
Affiliation(s)
- Annamaria Cattaneo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Valentina Zonca
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Marco A Riva
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| |
Collapse
|
14
|
Sullivan EL, Molloy KR, Dunn GA, Balanzar AL, Young AS, Loftis JM, Ablow JC, Nigg JT, Gustafsson HC. Adipokines measured during pregnancy and at birth are associated with infant negative affect. Brain Behav Immun 2024; 120:34-43. [PMID: 38772428 PMCID: PMC11401062 DOI: 10.1016/j.bbi.2024.05.018] [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: 10/22/2023] [Revised: 03/18/2024] [Accepted: 05/18/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Increased adiposity during pregnancy may be related to offspring risk for mental health disorders, although the biological mechanisms are poorly understood. One promising hypothesis is that factors secreted from adipocytes such as leptin and adiponectin may explain this association. The current study examined whether pregnancy or umbilical cord blood concentrations of leptin and/or adiponectin a) predict elevated infant negative affect at 6 months (an early life marker of risk for psychopathology); and b) help explain the association between pregnancy adiposity and increased infant negative affect. METHODS Data came from a prospective cohort (N = 305) of pregnant individuals and their offspring. Second trimester adiposity was assessed using air displacement plethysmography. Concentrations of leptin and adiponectin were measured in second trimester plasma and umbilical cord plasma. Infant negative affect was assessed by standardized observation at 6 months. Second trimester inflammation was assessed using a comprehensive panel of cytokines. RESULTS Lower second trimester adiponectin was associated with elevated infant negative affect, and mediated the effect of pregnancy adiposity on infant negative affect. This association was independent of the effect of second trimester inflammation. Umbilical cord leptin also predicted higher infant negative affect and mediated the association between pregnancy adiposity and infant negative affect. CONCLUSIONS This is the first study to link pregnancy adiponectin or cord blood leptin to infant markers of risk for psychopathology, and the first to demonstrate that these adipokines mediate the association between pregnancy adiposity and offspring behavioral outcomes, suggesting novel markers of risk and potential mechanisms of effect.
Collapse
Affiliation(s)
- Elinor L Sullivan
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States; Division of Neuroscience, Oregon National Primary Research Center, United States.
| | - Kelly R Molloy
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| | - Geoffrey A Dunn
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| | - Adriana L Balanzar
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| | - Anna S Young
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| | - Jennifer M Loftis
- Department of Psychiatry, Oregon Health & Science University, United States; VA Portland Health Care System, United States
| | | | - Joel T Nigg
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| | - Hanna C Gustafsson
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| |
Collapse
|
15
|
Leonard KM, Schmiedecke SS, Talley RL, Damicis JR, Walton RB, Burd I, Napolitano PG, Ieronimakis N. Maternal obesity alters fetal neuroinflammation in a murine model of preterm birth. AJOG GLOBAL REPORTS 2024; 4:100361. [PMID: 39072339 PMCID: PMC11278798 DOI: 10.1016/j.xagr.2024.100361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Preterm birth from intrauterine infection is a leading cause of neonatal neurologic morbidity. Likewise, maternal obesity is associated with intra-amniotic infection and inflammation. Whether maternal obesity is a risk factor for fetal brain injury that occurs with premature birth remains unknown. This study hypothesized that maternal obesity intensifies fetal neuroinflammation in the setting of premature delivery. OBJECTIVE This study aimed to examine the influence of maternal obesity on perinatal neuroinflammatory responses that arise with preterm birth using a murine model. STUDY DESIGN Dams with obesity were generated via a high-fat diet that was maintained throughout pregnancy. In parallel, dams without obesity (normal) received a control diet. All dams were paired with males on normal diet. Pregnant dams were randomized to receive an intrauterine administration of bacterial endotoxin (lipopolysaccharide) or the vehicle (phosphate-buffered saline) on embryo day 15.5 of what is typically a 19- to 21-day gestation. Fetal brains were harvested 6 hours after intrauterine administrations, and the expressions of key inflammatory cytokines (Il1b, Il6, and Tnf) and panels of metabolic, immune, and inflammatory genes were analyzed. RESULTS With the phosphate-buffered saline, there was no difference in gene expression related to maternal obesity. There were substantial differences in Il6 and immune/inflammatory expression profiles in fetal brains from dams with obesity vs normal dams that received lipopolysaccharide. Few differences were observed among the metabolic genes examined under these conditions. The gene expression pattern associated with maternal obesity correlated with pathways related to white matter injury. CONCLUSION The expression of neuroinflammatory markers instigated by bacterial endotoxin via intrauterine lipopolysaccharide was greater in embryo brains obtained from dams with obesity. Expression profiles suggest that in combination with intrauterine inflammation, maternal obesity may increase the risk of fetal white matter injury. Further investigation is warranted to understand the relationship between maternal health and neurologic outcomes associated with prematurity.
Collapse
Affiliation(s)
- Katherine M. Leonard
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Madigan Army Medical Center, Tacoma, WA (Leonard, Schmiedecke, Walton, and Ieronimakis)
| | - Stacey S. Schmiedecke
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Madigan Army Medical Center, Tacoma, WA (Leonard, Schmiedecke, Walton, and Ieronimakis)
| | - Rebecca L. Talley
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA (Talley, Damicis, and Ieronimakis)
| | - Jennifer R. Damicis
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA (Talley, Damicis, and Ieronimakis)
| | - Robert B. Walton
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Madigan Army Medical Center, Tacoma, WA (Leonard, Schmiedecke, Walton, and Ieronimakis)
| | - Irina Burd
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland, Baltimore, MD (Burd)
| | - Peter G. Napolitano
- Department of Obstetrics and Gynecology, University of Washington Medical Center, Seattle, WA (Napolitano)
| | - Nicholas Ieronimakis
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Madigan Army Medical Center, Tacoma, WA (Leonard, Schmiedecke, Walton, and Ieronimakis)
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA (Talley, Damicis, and Ieronimakis)
| |
Collapse
|
16
|
Nakaki A, Gomez Y, Darecka K, Borras R, Vellvé K, Paules C, Boutet ML, Basso A, Casu G, Traversi P, Youssef L, Casas I, Genero M, Benitez L, Larroya M, Casas R, Miranda J, Castro-Barquero S, Rodríguez-Sureda V, Arranz A, Pozo ÓJ, Gomez-Gomez A, Vieta E, Estruch R, Izquierdo Renau M, Eixarch E, Crispi F, Crovetto F, Gratacós E. Effects of Mediterranean Diet or Mindfulness-Based Stress Reduction during Pregnancy on Fetal Brain Development Detected by Neurosonography: A Secondary Analysis of a Randomized Clinical Trial (IMPACT BCN). Fetal Diagn Ther 2024:1-13. [PMID: 39079502 DOI: 10.1159/000540580] [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: 03/04/2024] [Accepted: 07/23/2024] [Indexed: 09/10/2024]
Abstract
INTRODUCTION We investigated whether structured maternal lifestyle interventions based on Mediterranean diet or stress reduction influence fetal-infant neurodevelopment detected by detailed fetal neurosonography and Ages and Stages Questionnaires 3rd edition (ASQ) at 12 months old. METHODS This was a secondary analysis of a randomized clinical trial (2017-2020), including 1,221 singleton pregnancies at high risk for small-for-gestational age. Participants were randomized into three groups at 19-23 weeks' gestation: Mediterranean diet intervention, stress reduction program, or usual care. A detailed neurosonography was performed on 881 participants at mean (SD) 33.4 (1.1) weeks' gestation. Neurosonographic measurements were done offline. ASQ was performed on 276 infants at 1 year of corrected age. RESULTS Biparietal diameter was similar among study groups. Mediterranean diet group fetuses had deeper insula (26.80 [1.68] versus 26.63 [1.75], mm, p = 0.02) and longer corpus callosum (42.98 [2.44] versus 42.62 [2.27], mm, p = 0.04), with a lower rate of suboptimal score infants in ASQ problem-solving domain (6.2 vs. 16.3%, p = 0.03). Stress reduction group fetuses had deeper insula (26.90 [1.75] versus 26.63 [1.75], mm, p = 0.04) and lower rates of suboptimal score infants in ASQ fine motor domain (4.3 vs. 12.8%, p = 0.04), compared to usual care group fetuses. CONCLUSION Maternal structured intervention during pregnancy of the trial has the potential to modify offspring's neurodevelopment.
Collapse
Affiliation(s)
- Ayako Nakaki
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain,
- Fundació de Recerca Clínic Barcelona - IDIBAPS, Barcelona, Spain,
| | - Yvan Gomez
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
| | - Katarzyna Darecka
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
| | - Roger Borras
- Cardiovascular Institute, Hospital Clínic, IDIBAPS, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Kilian Vellvé
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
| | - Cristina Paules
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
- Instituto de Investigación Sanitaria Aragón (IISAragon), Red de Salud Materno Infantil y del Desarrollo (SAMID), RETICS, Instituto de Salud Carlos III (ISCIII), Subdirección General de Evaluación y Fomento de la Investigación y Fondo Europeo de Desarrollo Regional (FEDER), Zaragoza, Spain
| | - Maria Laura Boutet
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
| | - Annachiara Basso
- Department of Obstetrics and Pediatrics ASST Lecco, A. Manzoni Hospital, Lecco, Italy
| | - Giulia Casu
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
| | - Paola Traversi
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
| | - Lina Youssef
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
- Fundació de Recerca Clínic Barcelona - IDIBAPS, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/Universitat de Barcelona (UB) Campus, Barcelona, Spain
| | - Irene Casas
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
- Fundació de Recerca Clínic Barcelona - IDIBAPS, Barcelona, Spain
| | - Mariona Genero
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Leticia Benitez
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
- Fundació de Recerca Clínic Barcelona - IDIBAPS, Barcelona, Spain
| | - Marta Larroya
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
- Fundació de Recerca Clínic Barcelona - IDIBAPS, Barcelona, Spain
| | - Rosa Casas
- Department of Internal Medicine Hospital Clinic, IDIBAPS, Universitat de Barcelona (UB), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERON), Comunidad de Madrid, Madrid, Spain
- Institut de Recerca en Nutrició i Seguretat Alimentaria (INSA-UB), Universitat de Barcelona (UB), Barcelona, Spain
| | - Jezid Miranda
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Cartagena, Cartagena de Indias, Colombia
| | - Sara Castro-Barquero
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERON), Comunidad de Madrid, Madrid, Spain
- Institut de Recerca en Nutrició i Seguretat Alimentaria (INSA-UB), Universitat de Barcelona (UB), Barcelona, Spain
| | - Víctor Rodríguez-Sureda
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
- Fundació de Recerca Clínic Barcelona - IDIBAPS, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| | - Angela Arranz
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
| | - Óscar J Pozo
- Applied Metabolomics Research Group, IMIM-Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| | - Alex Gomez-Gomez
- Applied Metabolomics Research Group, IMIM-Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| | - Eduard Vieta
- Hospital Clinic, Department of Psychiatry and Psychology, Neuroscience Institute, IDIBAPS, Universitat de Barcelona (UB), CIBERSAM, Barcelona, Spain
| | - Ramon Estruch
- Department of Internal Medicine Hospital Clinic, IDIBAPS, Universitat de Barcelona (UB), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERON), Comunidad de Madrid, Madrid, Spain
- Institut de Recerca en Nutrició i Seguretat Alimentaria (INSA-UB), Universitat de Barcelona (UB), Barcelona, Spain
| | - Montserrat Izquierdo Renau
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
- Neonatology Department, Hospital Sant Joan de Déu, Universitat de Barcelona (UB), Barcelona, Spain
| | - Elisenda Eixarch
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
- Fundació de Recerca Clínic Barcelona - IDIBAPS, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Fàtima Crispi
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
- Fundació de Recerca Clínic Barcelona - IDIBAPS, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| | - Francesca Crovetto
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin RD21/0012/0003, Instituto de Salud Carlos III, Madrid, Spain
| | - Eduard Gratacós
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona (UB), Barcelona, Spain
- Fundació de Recerca Clínic Barcelona - IDIBAPS, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| |
Collapse
|
17
|
Parsaei M, Hashemi SM, Moghaddam HS, Peterson BS. A systematic review of MRI studies on the effects of maternal obesity on offspring brain structure and function. J Neurosci Res 2024; 102:e25368. [PMID: 39007363 DOI: 10.1002/jnr.25368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 06/27/2024] [Accepted: 06/30/2024] [Indexed: 07/16/2024]
Abstract
Maternal obesity before or during pregnancy has been associated previously in offspring with a wide range of poor neurodevelopmental outcomes and mental health problems. The effects of maternal obesity on offspring brain structure and function that may be responsible for these poor outcomes are not well understood. We, therefore, undertook a systematic review of magnetic resonance imaging (MRI) studies that have assessed the associations of maternal obesity with brain measures in offspring. A systematic search was conducted in PubMed, Web of Science, Scopus, and PsycINFO on August 20, 2023. Of 15 eligible studies, seven employed functional MRI (fMRI), five diffusion tensor imaging (DTI), and four anatomical MRI (one used both DTI and anatomical MRI) in the offspring. The ages of offspring varied widely: one was a study of fetuses in utero, five of neonates, one of infants, five of school-aged children, two of both neonates and infants, and one of both children and adults. Collectively, 12 studies reported significant associations of maternal obesity with structural or functional alterations of the offspring's brain, most frequently in the prefrontal cortex and limbic system. In conclusion, maternal obesity appears to have a profound influence on offspring brain development, particularly within the prefrontal and limbic networks that regulate emotion and behavior. Further studies are needed to identify how changes in brain structure and function mediate the effects of maternal obesity on long-term emotional and behavioral outcomes, as well as the molecular pathways through which maternal obesity alters offspring brain development.
Collapse
Affiliation(s)
- Mohammadamin Parsaei
- Maternal, Fetal & Neonatal Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hossein Sanjari Moghaddam
- Psychiatry and Psychology Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Bradley S Peterson
- Institute for the Developing Mind, Children's Hospital, Los Angeles, California, USA
- Department of Psychiatry, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
18
|
Akgun Aktaş B, Ecem K, Murat H, Gökçe Ozcan K, Ozgür K, Atakan T, Dilek S. Fetal cortical development and neurosonographic findings in obese pregnant women: a case control study from a tertiary hospital. Arch Gynecol Obstet 2024; 310:83-91. [PMID: 37831178 DOI: 10.1007/s00404-023-07258-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/05/2023] [Indexed: 10/14/2023]
Abstract
PURPOSE To investigate the effect of obesity on fetal cortical development. METHODS This prospective cross-sectional study was conducted with 91 pregnant women. Fetal neurosonography scans were performed in the third trimester, and according to body mass index (BMI) values, the patients were evaluated in two groups: obese (BMI ≥ 30 kg/m2) and normal weight (BMI < 30 kg/m2). During neurosonography, fetal insular depth and Sylvian fissures, parieo-occipital and cingulate fissure depth, frontal lobe length, and the sizes of the corpus callosum and cavum septum pellucidum were measured using a transvaginal approach. Fetal cortical development and Sylvian fissure operculization were graded. RESULTS The number of patients with grade 2 fetal cortical development was significantly higher among the pregnant women in the obese group compared to the normal weight group (n = 17, 41.5% and n = 8, 16.0%, respectively; p = 0.007). In the obese group, the number of pregnant women with grade 4 or below fetal Sylvian fissure operculization was significantly higher (n = 13, 31.7%), and the number of those with grade 9 or above operculization was significantly lower (n = 1, 2.5%) (p = 0.003). The fetal insular depth, frontal lobe anterior-posterior diameter, cingulate fissure depth, and corpus callosum thickness were lower in the obese group, albeit with no significant difference. Parieto-occipital depth significantly decreased in the obese group [6.8 (6) mm)] compared to the control group [10.5 (7.2) mm)] (p = 0.008). The fetal Sylvian fissure ratio and the cavum septum pellucidum ratio were found to be similar between the groups. CONCLUSION The data obtained from this study showed that obesity caused fetal cortical changes in pregnant women.
Collapse
Affiliation(s)
- Betül Akgun Aktaş
- Department of Obstetrics and Gynecology, Ministry of Health, Ankara City Hospital, Üniversiteler Mahallesi 1604. Cadde No: 9 Çankaya, Ankara, Turkey.
| | - Kaya Ecem
- Department of Obstetrics and Gynecology, Ministry of Health, Ankara City Hospital, Üniversiteler Mahallesi 1604. Cadde No: 9 Çankaya, Ankara, Turkey
| | - Haksever Murat
- Department of Obstetrics and Gynecology, Ministry of Health, Ankara City Hospital, Üniversiteler Mahallesi 1604. Cadde No: 9 Çankaya, Ankara, Turkey
| | - Kılınç Gökçe Ozcan
- Department of Obstetrics and Gynecology, Ministry of Health, Ankara City Hospital, Üniversiteler Mahallesi 1604. Cadde No: 9 Çankaya, Ankara, Turkey
| | - Kara Ozgür
- Department of Obstetrics and Gynecology, Ministry of Health, Ankara City Hospital, Üniversiteler Mahallesi 1604. Cadde No: 9 Çankaya, Ankara, Turkey
- Department of Obstetrics and Gynecology, University of Health Sciences, Istanbul, Turkey
| | - Tanacan Atakan
- Department of Obstetrics and Gynecology, Ministry of Health, Ankara City Hospital, Üniversiteler Mahallesi 1604. Cadde No: 9 Çankaya, Ankara, Turkey
- Department of Obstetrics and Gynecology, University of Health Sciences, Istanbul, Turkey
| | - Sahin Dilek
- Department of Obstetrics and Gynecology, Ministry of Health, Ankara City Hospital, Üniversiteler Mahallesi 1604. Cadde No: 9 Çankaya, Ankara, Turkey
- Department of Obstetrics and Gynecology, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
19
|
Batorsky R, Ceasrine AM, Shook LL, Kislal S, Bordt EA, Devlin BA, Perlis RH, Slonim DK, Bilbo SD, Edlow AG. Hofbauer cells and fetal brain microglia share transcriptional profiles and responses to maternal diet-induced obesity. Cell Rep 2024; 43:114326. [PMID: 38848212 DOI: 10.1016/j.celrep.2024.114326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/25/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
Maternal immune activation is associated with adverse offspring neurodevelopmental outcomes, many mediated by in utero microglial programming. As microglia remain inaccessible throughout development, identification of noninvasive biomarkers reflecting fetal brain microglial programming could permit screening and intervention. We used lineage tracing to demonstrate the shared ontogeny between fetal brain macrophages (microglia) and fetal placental macrophages (Hofbauer cells) in a mouse model of maternal diet-induced obesity, and single-cell RNA-seq to demonstrate shared transcriptional programs. Comparison with human datasets demonstrated conservation of placental resident macrophage signatures between mice and humans. Single-cell RNA-seq identified common alterations in fetal microglial and Hofbauer cell gene expression induced by maternal obesity, as well as sex differences in these alterations. We propose that Hofbauer cells, which are easily accessible at birth, provide insights into fetal brain microglial programs and may facilitate the early identification of offspring vulnerable to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Rebecca Batorsky
- Data Intensive Studies Center, Tufts University, Medford, MA, USA
| | - Alexis M Ceasrine
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Lydia L Shook
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Sezen Kislal
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Evan A Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Benjamin A Devlin
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Roy H Perlis
- Department of Psychiatry and Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Donna K Slonim
- Department of Computer Science, Tufts University, Medford, MA, USA
| | - Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA; Department of Neurobiology, Duke University, Durham, NC, USA; Lurie Center for Autism, Massachusetts General Hospital, Boston, MA, USA
| | - Andrea G Edlow
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
20
|
Nel NH, Haddad EN, Kerver JM, Cassidy-Bushrow AE, Comstock SS. Maternal Body Mass Index Associates with Prenatal Characteristics and Fecal Microbial Communities. Nutrients 2024; 16:1881. [PMID: 38931236 PMCID: PMC11206496 DOI: 10.3390/nu16121881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
The maternal microbiome plays a vital role in shaping pregnancy outcomes, but there remains a substantial gap in understanding its precise relationships to maternal health, particularly in relation to potential effects of body mass index (BMI) on gut microbial diversity. The aim of this observational study was to assess maternal characteristics in association with pre-pregnancy BMI and to further assess microbial diversity in association with specific maternal characteristics. Eighty-four pregnant women were recruited during their third trimester of pregnancy from various prenatal clinics across the state of Michigan. The participants completed an enrollment questionnaire including self-reported pre-pregnancy BMI; stool samples were collected to assess the fecal microbial community composition. Pre-pregnancy obesity (BMI 30+) was associated (univariably) with antibiotic use before pregnancy, ever smoked, lower education level, and being unmarried. The gut microbiota alpha diversity was significantly different for pregnant women by pre-pregnancy BMI category (normal, overweight, obese). The beta diversity was unique for the gut microbiotas of pregnant women within each BMI category, by education level, and by marital status. Multivariable models revealed that pre-pregnancy BMI, maternal education, marital status, and maternal age were associated with the microbial diversity of the gut microbiota during pregnancy. These results give new insight into the relationship between a woman's microbiome during pregnancy and their prenatal health, along with an understanding of the relationships between socioeconomic factors and microbial diversity.
Collapse
Affiliation(s)
- Nikita H. Nel
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA
| | - Eliot N. Haddad
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA
| | - Jean M. Kerver
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Andrea E. Cassidy-Bushrow
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Department of Public Health Sciences, Henry Ford Health, Detroit, MI 48202, USA
| | - Sarah S. Comstock
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
21
|
Wang H, Yin W, Ma S, Wang P, Zhang L, Li P, Shao Z, Chen X, Zhu P. Prenatal environmental adversity and child neurodevelopmental delay: the role of maternal low-grade systemic inflammation and maternal anti-inflammatory diet. Eur Child Adolesc Psychiatry 2024; 33:1771-1781. [PMID: 37596369 DOI: 10.1007/s00787-023-02267-9] [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: 02/18/2023] [Accepted: 07/22/2023] [Indexed: 08/20/2023]
Abstract
Maternal inflammation has been proposed as a possible pathway connecting prenatal environmental adversity (PEA), which includes maternal overweightness or obesity, diabetes, hypertensive disorders, and mood or anxiety disorders, to child neurodevelopmental delay. However, effective preventive measures have not yet been reported. Herein, we aimed to investigate whether a maternal anti-inflammatory diet reduced the risk of PEA-induced neurodevelopmental delay, by inhibiting inflammation. This prospective study included 7438 mother-child pairs. Maternal overweightness or obesity, diabetes, and hypertensive disorders were diagnosed before 28 week gestation. Maternal depression disorders were identified using the Edinburgh postnatal depression survey (EPDS) during mid-pregnancy. During mid- and late pregnancy, maternal high-sensitivity C-reactive protein (hs-CRP) levels were measured to evaluate systemic inflammation. The inflammatory potential of the diet was evaluated using the food-based empirical dietary inflammatory pattern (EDIP) score during mid-pregnancy. Pregnant women were classified into high- or low-score groups based on the median EDIP score. The outcomes of neurodevelopmental delay at 6-36 month postpartum were extracted from the Register of Child Healthcare. Among the 7438 mother-child pairs, 2937 (39.5%) were exposed to PEA, and neurodevelopmental delay occurred in 540 (7.3%). Children exposed to PEA had a higher risk of neurodevelopmental delay than those not exposed. PEA exposure was associated with increased hs-CRP during pregnancy in a PEA monotonic manner, an interquartile range increase in hs-CRP in mid- and late pregnancy was associated with an increased risk of child neurodevelopmental delay. Higher maternal persistent inflammation partially mediated the effect of PEA exposure on child neurodevelopmental delay by 17.19%. An increased risk of PEA-related neurodevelopmental delay was observed only in the children of mothers with high-EDIP rather than low-EDIP. These results suggest that increased systemic inflammation through mid- and late pregnancy mediates the association between PEA and child neurodevelopmental delay. A maternal anti-inflammatory diet may improve PEA-induced neurodevelopmental delay, by inhibiting inflammation.
Collapse
Affiliation(s)
- Haixia Wang
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, China
- MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, China
| | - Wanjun Yin
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, China
- MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, China
| | - Shuangshuang Ma
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, China
- Anhui Mental Health Centre, Hefei, China
| | - Peng Wang
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, China
- MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, China
| | - Lei Zhang
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, China
- MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, China
| | - Peipei Li
- Hefei Maternal and Child Health Service Centre, Hefei, China
| | - Ziyu Shao
- Hefei Maternal and Child Health Service Centre, Hefei, China
| | - Xianxia Chen
- Department of Obstetrics and Gynecology, Anhui Maternal and Child Health Hospital, 15 Yimin Street, Hefei, China.
| | - Peng Zhu
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, China.
- MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, China.
| |
Collapse
|
22
|
Alves FCR, Moreira A, Moutinho O. Maternal and long-term offspring outcomes of obesity during pregnancy. Arch Gynecol Obstet 2024; 309:2315-2321. [PMID: 38502190 DOI: 10.1007/s00404-023-07349-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 03/21/2024]
Abstract
PURPOSE Obesity`s prevalence is rising in women of reproductive age worldwide and has become the most common medical condition at this age group. Besides, its occurrence is also rising during pregnancy. This condition not only increases the risk of noncommunicable diseases on the mother, such as cardiovascular disease and diabetes, but also transfers this risk to the offspring. METHODS This is a narrative review based on scientific and review articles on the matter. RESULTS Obesity is associated with an increased risk of gestational diabetes mellitus, gestational hypertension and preeclampsia, venous thromboembolism, infection, and mental health problems. Furthermore, it has an impact on the progress of labor and induction matters. Regarding offspring outcomes, it is related to higher incidence of congenital anomalies, perinatal mortality, and the occurrence of large for gestational age newborns. Still, it has implications on cardiometabolic risk and neurodevelopment in offspring. CONCLUSION It is, therefore, imperative to encourage the adoption of healthy lifestyles, especially in the peri-conception and interpregnancy periods. Likewise, there must be support in the multidisciplinary monitoring of these pregnant women to minimize associated complication rates.
Collapse
Affiliation(s)
- Fernanda Cristina Ribeiro Alves
- Obstetrics and Gynecology Department, Centro Hospitalar de Trás-os-Montes e Alto Douro, Avenida da Noruega, Lordelo, Vila Real, Portugal.
| | - Ana Moreira
- Obstetrics and Gynecology Department, Centro Hospitalar de Trás-os-Montes e Alto Douro, Avenida da Noruega, Lordelo, Vila Real, Portugal
| | - Osvaldo Moutinho
- Obstetrics and Gynecology Department, Centro Hospitalar de Trás-os-Montes e Alto Douro, Avenida da Noruega, Lordelo, Vila Real, Portugal
| |
Collapse
|
23
|
Kacperska M, Mizera J, Pilecki M, Pomierny-Chamioło L. The impact of excessive maternal weight on the risk of neuropsychiatric disorders in offspring-a narrative review of clinical studies. Pharmacol Rep 2024; 76:452-462. [PMID: 38649593 PMCID: PMC11126479 DOI: 10.1007/s43440-024-00598-1] [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: 11/14/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024]
Abstract
The global prevalence of overweight and obesity is a significant public health concern that also largely affects women of childbearing age. Human epidemiological studies indicate that prenatal exposure to excessive maternal weight or excessive gestational weight gain is linked to various neurodevelopmental disorders in children, including attention deficit hyperactivity disorder, autism spectrum disorder, internalizing and externalizing problems, schizophrenia, and cognitive/intellectual impairment. Considering that inadequate maternal body mass can induce serious disorders in offspring, it is important to increase efforts to prevent such outcomes. In this paper, we review human studies linking excessive maternal weight and the occurrence of mental disorders in children.
Collapse
Affiliation(s)
- Magdalena Kacperska
- Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Józef Mizera
- Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Maciej Pilecki
- Department of Psychiatry, Jagiellonian University Medical College, Kopernika 21a, 31-500, Kraków, Poland
| | - Lucyna Pomierny-Chamioło
- Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland.
| |
Collapse
|
24
|
Holthaus E, O'Neill M, Jeske W, DeChristopher P, Goodman J, Glynn L, Levin S, Muraskas J. Endocan: A biomarker for endothelial dysfunction and inflammation, linking maternal obesity and pediatric obesity in a cohort of preterm neonates. Eur J Obstet Gynecol Reprod Biol 2024; 297:132-137. [PMID: 38626514 DOI: 10.1016/j.ejogrb.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/18/2024]
Abstract
OBJECTIVES Numerous animal and epidemiologic studies have demonstrated a positive association between maternal obesity in pregnancy and obesity in offspring. The biologic mechanisms of this association remain under investigation. One proposed mechanism includes fetoplacental endothelial dysfunction secondary to inflammation. Endocan is a relatively new biomarker for endothelial dysfunction and inflammation. Our objectives were to examine (1) the association between maternal obesity and neonatal serum endocan at birth, and (2) the association between neonatal serum endocan at birth and pediatric obesity at 24-36 months of age. STUDY DESIGN This was a secondary analysis of a prospective cohort of neonates born < 33 weeks gestation. Serum endocan was collected within 48 hours of birth. Serum endocan levels were compared in neonates born to obese mothers vs. those born to non-obese mothers. BMI data were retrospectively collected from cohort neonates between 24 and 36 months of age. RESULTS The analysis included 120 mother/neonate dyads. Neonates born to obese mothers had higher median serum endocan at birth compared to neonates born to non-obese mothers (299 ng/L [205-586] vs. 251 ng/L [164-339], p = 0.045). In a linear regression modeled on neonatal serum endocan level, maternal obesity had a statistically significant positive association (p = 0.021). Higher mean serum endocan level at birth was associated with pediatric obesity between 24 and 36 months (obese vs. non-obese offspring; 574 ng/L (222) vs. 321 ng/L (166), p = 0.005). CONCLUSIONS In our cohort of preterm neonates, elevated serum endocan at birth was associated with both maternal obesity and downstream pediatric obesity. More research is needed to understand intergenerational transmission of obesity. A large focus has been on epigenetic modification. Endothelial dysfunction and inflammation may play important roles in these pathways. Effective biomarkers, including endocan, may also serve as intermediate outcomes in future pregnancy research.
Collapse
Affiliation(s)
- E Holthaus
- Maternal Fetal Medicine, Loyola University Medical Center, 2160 S. 1(st) Ave, Maywood, IL 60153, USA.
| | - M O'Neill
- Loyola University Stritch School of Medicine, 2160 S. 1(st) Ave, Maywood, IL 60153, USA
| | - W Jeske
- Thoracic and Cardiovascular Surgery, Cell and Molecular Physiology, Loyola University Chicago, 2160 S. 1(st) Ave, Maywood, IL 60153, USA
| | - P DeChristopher
- Pathology and Laboratory Medicine, Transfusion Medicine. Loyola University Medical Center, 2160 S. 1(st) Ave, Maywood, IL 60153, USA
| | - J Goodman
- Maternal Fetal Medicine, University of Missouri School of Medicine, MU Women's Hospital, 404 N Keene St, Columbia, MO 65201, USA
| | - L Glynn
- Pediatric Surgery, NYU Langone Hospital, 120 Mineola Blvd, Suite 210, Mineola, NY 11501, USA
| | - S Levin
- Neonatal Perinatal. University of Oklahoma College of Medicine, 1200 North Everett Drive, ETNP 7504, Oklahoma City, OK, 73104, USA
| | - J Muraskas
- Neonatal-Perinatal Research, Neonatology, Loyola University Medical Center, 2160 S. 1(st) Ave, Maywood, IL 60153, USA
| |
Collapse
|
25
|
Lee YJ, Kim J, Kwon YH. Long-Term Effects of Maternal Fat Consumption on the Brain Transcriptome of Obesogenic Diet-Fed Young Adult Mice Offspring. J Nutr 2024; 154:1532-1539. [PMID: 38484978 DOI: 10.1016/j.tjnut.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/17/2024] [Accepted: 03/11/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Substantial evidence has demonstrated that maternal high-fat (HF) consumption during gestation and lactation plays as a risk factor for neurodevelopmental alterations and subsequent neurological disorders. OBJECTIVE We investigated the regulatory mechanisms of maternal fat consumption on brain development and function in offspring at different ages. METHODS Mouse dams were fed either a control diet [low-fat (LF)] or an HF diet for 3 wk before mating and throughout pregnancy and lactation. Offspring were killed at postnatal day (PD) 21 (LF21 and HF21), and the rest were fed an HF diet for 12 wk until the killing at PD 105 (LF105 and HF105). The expression levels of genes and proteins in the brains of offspring were analyzed by microarray and immunoblotting, respectively. RESULTS Maternal dietary fat content, offspring age, and their interaction affected the expression levels of 1215, 10,453, and 2105 genes, respectively. The 67 differentially expressed genes (DEGs) between the HF21 and LF21 groups were enriched in several Gene Ontology terms related to nervous system development. Among 45 DEGs of the HF105/LF105 comparison, several genes associated with neurotransmitter action are detected. In addition, we observed increased activation of the AMP-dependent protein kinase-cAMP response element binding protein signaling pathway in HF105/LF105 comparison. However, maternal fat content did not change the protein levels of amyloid-β and tau hyperphosphorylation, the markers of neuropathogenesis. CONCLUSIONS Maternal HF feeding altered the expression of genes involved in the development and neurotransmitter system in the brains of PD 21 and HF diet-fed PD 105 offspring, respectively. Especially, the absence of overlap between DEGs at each comparison highlights the dynamic nature of alterations in gene expression in offspring of dams fed an HF diet. Further investigation on older adult offspring is necessary to elucidate the effects of maternal fat intake on the brain pathophysiology of offspring.
Collapse
Affiliation(s)
- Youn Ji Lee
- Department of Food and Nutrition, Seoul National University, Seoul, Korea
| | - Juhae Kim
- Department of Food and Nutrition, Seoul National University, Seoul, Korea; Research Institute of Human Ecology, Seoul National University, Seoul, Korea
| | - Young Hye Kwon
- Department of Food and Nutrition, Seoul National University, Seoul, Korea; Research Institute of Human Ecology, Seoul National University, Seoul, Korea.
| |
Collapse
|
26
|
Lubrano C, Parisi F, Cetin I. Impact of Maternal Environment and Inflammation on Fetal Neurodevelopment. Antioxidants (Basel) 2024; 13:453. [PMID: 38671901 PMCID: PMC11047368 DOI: 10.3390/antiox13040453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
During intrauterine life, external stimuli including maternal nutrition, lifestyle, socioeconomic conditions, anxiety, stress, and air pollution can significantly impact fetal development. The human brain structures begin to form in the early weeks of gestation and continue to grow and mature throughout pregnancy. This review aims to assess, based on the latest research, the impact of environmental factors on fetal and neonatal brain development, showing that oxidative stress and inflammation are implied as a common factor for most of the stressors. Environmental insults can induce a maternal inflammatory state and modify nutrient supply to the fetus, possibly through epigenetic mechanisms, leading to significant consequences for brain morphogenesis and neurological outcomes. These risk factors are often synergic and mutually reinforcing. Fetal growth restriction and preterm birth represent paradigms of intrauterine reduced nutrient supply and inflammation, respectively. These mechanisms can lead to an increase in free radicals and, consequently, oxidative stress, with well-known adverse effects on the offspring's neurodevelopment. Therefore, a healthy intrauterine environment is a critical factor in supporting normal fetal brain development. Hence, healthcare professionals and clinicians should implement effective interventions to prevent and reduce modifiable risk factors associated with an increased inflammatory state and decreased nutrient supply during pregnancy.
Collapse
Affiliation(s)
- Chiara Lubrano
- Nutritional Sciences, Doctoral Programme (PhD), Università degli Studi di Milano, 20157 Milan, Italy;
- Department of Mother, Child and Neonate, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Francesca Parisi
- Department of Mother, Child and Neonate, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy;
| | - Irene Cetin
- Department of Mother, Child and Neonate, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy;
| |
Collapse
|
27
|
Carter SA, Lin JC, Chow T, Martinez MP, Alves JM, Feldman KR, Qiu C, Page KA, McConnell R, Xiang AH. Maternal obesity and diabetes during pregnancy and early autism screening score at well-child visits in standard clinical practice. AUTISM : THE INTERNATIONAL JOURNAL OF RESEARCH AND PRACTICE 2024; 28:975-984. [PMID: 37646431 PMCID: PMC10902177 DOI: 10.1177/13623613231188876] [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] [Indexed: 09/01/2023]
Abstract
LAY ABSTRACT Early intervention and treatment can help reduce disability in children diagnosed with autism spectrum disorder. Screening for autism spectrum disorder in young children identifies those at increased likelihood of diagnosis who may need further support. Previous research has reported that exposure to maternal obesity and diabetes during pregnancy is associated with higher likelihood of autism spectrum disorder diagnosis in children. However, little is known about whether these maternal conditions are associated with how very young children score on autism spectrum disorder screening tools. This study examined associations between exposure to maternal obesity and diabetes during pregnancy and offspring scores on the Quantitative Checklist for Autism in Toddlers, an autism spectrum disorder screening questionnaire administered between 18-24 months at well-child visits. A higher score on the Quantitative Checklist for Autism in Toddlers suggests a higher likelihood of autism spectrum disorder; children with scores 3 or greater are referred to developmental pediatricians for evaluation. Our study found that children of mothers with obesity or diabetes during pregnancy had higher scores than children whose mothers did not have these conditions. Associations with maternal obesity and gestational diabetes diagnosed at or before 26 weeks of pregnancy were also present in children who did not have later autism spectrum disorder diagnoses, suggesting that exposure to these conditions during early pregnancy may be associated with a broad range of social and behavioral abilities. Identifying associations between maternal health conditions and early Quantitative Checklist for Autism in Toddlers screening scores could influence future screening and provision of support for children of mothers with these conditions.
Collapse
Affiliation(s)
- Sarah A. Carter
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Jane C. Lin
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Ting Chow
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Mayra P. Martinez
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Jasmin M. Alves
- Division of Endocrinology, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Diabetes and Obesity Research Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Klara R. Feldman
- Department of Anesthesiology & Perioperative Medicine, Kaiser Permanente Southern California, Baldwin Park, CA
| | - Chunyuan Qiu
- Department of Anesthesiology & Perioperative Medicine, Kaiser Permanente Southern California, Baldwin Park, CA
| | - Kathleen A. Page
- Division of Endocrinology, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Rob McConnell
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anny H. Xiang
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| |
Collapse
|
28
|
Candia AA, Lean SC, Zhang CXW, McKeating DR, Cochrane A, Gulacsi E, Herrera EA, Krause BJ, Sferruzzi-Perri AN. Obesogenic Diet in Mice Leads to Inflammation and Oxidative Stress in the Mother in Association with Sex-Specific Changes in Fetal Development, Inflammatory Markers and Placental Transcriptome. Antioxidants (Basel) 2024; 13:411. [PMID: 38671859 PMCID: PMC11047652 DOI: 10.3390/antiox13040411] [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: 02/07/2024] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Obesity during pregnancy is related to adverse maternal and neonatal outcomes. Factors involved in these outcomes may include increased maternal insulin resistance, inflammation, oxidative stress, and nutrient mishandling. The placenta is the primary determinant of fetal outcomes, and its function can be impacted by maternal obesity. The aim of this study on mice was to determine the effect of obesity on maternal lipid handling, inflammatory and redox state, and placental oxidative stress, inflammatory signaling, and gene expression relative to female and male fetal growth. METHODS Female mice were fed control or obesogenic high-fat/high-sugar diet (HFHS) from 9 weeks prior to, and during, pregnancy. On day 18.5 of pregnancy, maternal plasma, and liver, placenta, and fetal serum were collected to examine the immune and redox states. The placental labyrinth zone (Lz) was dissected for RNA-sequencing analysis of gene expression changes. RESULTS the HFHS diet induced, in the dams, hepatic steatosis, oxidative stress (reduced catalase, elevated protein oxidation) and the activation of pro-inflammatory pathways (p38-MAPK), along with imbalanced circulating cytokine concentrations (increased IL-6 and decreased IL-5 and IL-17A). HFHS fetuses were asymmetrically growth-restricted, showing sex-specific changes in circulating cytokines (GM-CSF, TNF-α, IL-6 and IFN-γ). The morphology of the placenta Lz was modified by an HFHS diet, in association with sex-specific alterations in the expression of genes and proteins implicated in oxidative stress, inflammation, and stress signaling. Placental gene expression changes were comparable to that seen in models of intrauterine inflammation and were related to a transcriptional network involving transcription factors, LYL1 and PLAG1. CONCLUSION This study shows that fetal growth restriction with maternal obesity is related to elevated oxidative stress, inflammatory pathways, and sex-specific placental changes. Our data are important, given the marked consequences and the rising rates of obesity worldwide.
Collapse
Affiliation(s)
- Alejandro A. Candia
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
- Institute of Health Sciences, University of O’Higgins, Rancagua 2841959, Chile;
- Pathophysiology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile;
- Department for the Woman and Newborn Health Promotion, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Samantha C. Lean
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Cindy X. W. Zhang
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Daniel R. McKeating
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Anna Cochrane
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Edina Gulacsi
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Emilio A. Herrera
- Pathophysiology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile;
| | - Bernardo J. Krause
- Institute of Health Sciences, University of O’Higgins, Rancagua 2841959, Chile;
| | - Amanda N. Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| |
Collapse
|
29
|
Tamayo JM, Osman HC, Schwartzer JJ, Ashwood P. The influence of asthma on neuroinflammation and neurodevelopment: From epidemiology to basic models. Brain Behav Immun 2024; 116:218-228. [PMID: 38070621 DOI: 10.1016/j.bbi.2023.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 11/08/2023] [Accepted: 12/04/2023] [Indexed: 12/18/2023] Open
Abstract
Asthma is a highly heterogeneous inflammatory disease that can have a significant effect on both the respiratory system and central nervous system. Population based studies and animal models have found asthma to be comorbid with a number of neurological conditions, including depression, anxiety, and neurodevelopmental disorders. In addition, maternal asthma during pregnancy has been associated with neurodevelopmental disorders in the offspring, such as autism spectrum disorders and attention deficit hyperactivity disorder. In this article, we review the most current epidemiological studies of asthma that identify links to neurological conditions, both as it relates to individuals that suffer from asthma and the impacts asthma during pregnancy may have on offspring neurodevelopment. We also discuss the relevant animal models investigating these links, address the gaps in knowledge, and explore the potential future directions in this field.
Collapse
Affiliation(s)
- Juan M Tamayo
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, CA 95817, USA
| | - Hadley C Osman
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, CA 95817, USA
| | - Jared J Schwartzer
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, CA 95817, USA.
| |
Collapse
|
30
|
Adibi JJ, Zhao Y, Koistinen H, Mitchell RT, Barrett ES, Miller R, O'Connor TG, Xun X, Liang HW, Birru R, Smith M, Moog NK. Molecular pathways in placental-fetal development and disruption. Mol Cell Endocrinol 2024; 581:112075. [PMID: 37852527 PMCID: PMC10958409 DOI: 10.1016/j.mce.2023.112075] [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: 07/10/2023] [Revised: 09/11/2023] [Accepted: 09/24/2023] [Indexed: 10/20/2023]
Abstract
The first trimester of pregnancy ranks high in priority when minimizing harmful exposures, given the wide-ranging types of organogenesis occurring between 4- and 12-weeks' gestation. One way to quantify potential harm to the fetus in the first trimester is to measure a corollary effect on the placenta. Placental biomarkers are widely present in maternal circulation, cord blood, and placental tissue biopsied at birth or at the time of pregnancy termination. Here we evaluate ten diverse pathways involving molecules expressed in the first trimester human placenta based on their relevance to normal fetal development and to the hypothesis of placental-fetal endocrine disruption (perturbation in development that results in abnormal endocrine function in the offspring), namely: human chorionic gonadotropin (hCG), thyroid hormone regulation, peroxisome proliferator activated receptor protein gamma (PPARγ), leptin, transforming growth factor beta, epiregulin, growth differentiation factor 15, small nucleolar RNAs, serotonin, and vitamin D. Some of these are well-established as biomarkers of placental-fetal endocrine disruption, while others are not well studied and were selected based on discovery analyses of the placental transcriptome. A literature search on these biomarkers summarizes evidence of placenta-specific production and regulation of each biomarker, and their role in fetal reproductive tract, brain, and other specific domains of fetal development. In this review, we extend the theory of fetal programming to placental-fetal programming.
Collapse
Affiliation(s)
- Jennifer J Adibi
- Department of Epidemiology, University of Pittsburgh School of Public Health, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Yaqi Zhao
- St. Jude's Research Hospital, Memphis, TN, USA
| | - Hannu Koistinen
- Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland
| | - Rod T Mitchell
- Department of Paediatric Endocrinology, Royal Hospital for Children and Young People, Edinburgh BioQuarter, Edinburgh, UK
| | - Emily S Barrett
- Environmental and Population Health Bio-Sciences, Rutgers University School of Public Health, Piscataway, NJ, USA
| | - Richard Miller
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - Thomas G O'Connor
- Department of Psychiatry, University of Rochester Medical Center, Rochester, NY, USA
| | - Xiaoshuang Xun
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Hai-Wei Liang
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Rahel Birru
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Megan Smith
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nora K Moog
- Department of Medical Psychology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
31
|
Trujillo-Villarreal LA, Cruz-Carrillo G, Angeles-Valdez D, Garza-Villarreal EA, Camacho-Morales A. Paternal Prenatal and Lactation Exposure to a High-Calorie Diet Shapes Transgenerational Brain Macro- and Microstructure Defects, Impacting Anxiety-Like Behavior in Male Offspring Rats. eNeuro 2024; 11:ENEURO.0194-23.2023. [PMID: 38212114 PMCID: PMC10863632 DOI: 10.1523/eneuro.0194-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 11/21/2023] [Accepted: 12/11/2023] [Indexed: 01/13/2024] Open
Abstract
Prenatal exposure to high-energy diets (HED) increases the susceptibility to behavioral alterations in the male offspring. We addressed whether prenatal HED primes the transgenerational inheritance of structural brain changes impacting anxiety/depression-like behavior in the offspring. For this, we used female Wistar rats exposed to a HED [cafeteria (CAF) diet, n = 6] or chow [control (CON) n = 6] during development. Anxiety and depression-like behavior were evaluated in filial 1 (F1), filial 2 (F2), and filial 3 (F3) male offspring using the open field (OFT), elevated plus maze, novelty suppressed feeding (NSFT), tail suspension (TST), and forced swimming tests. Structural brain changes were identified by deformation-based morphometry (DBM) and diffusion tensor imaging using ex vivo MRI. We found that the F1, F2, and F3 offspring exposed to CAF diet displayed higher anxious scores including longer feeding latency during the NSFT, and in the closed arms, only F1 offspring showed longer stay on edges during the OFT versus control offspring. DBM analysis revealed that CAF offspring exhibited altered volume in the cerebellum, hypothalamus, amygdala, and hippocampus preserved up to the F3 generation of anxious individuals. Also, F3 CAF anxious exhibited greater fractional anisotropy and axial diffusivity (AD) in the amygdala, greater apparent diffusion coefficient in the corpus callosum, and greater AD in the hippocampus with respect to the control. Our results suggest that prenatal and lactation exposure to HED programs the transgenerational inheritance of structural brain changes related to anxiety-like behavior in the male offspring.
Collapse
Affiliation(s)
- Luis A Trujillo-Villarreal
- Department of Biochemistry, College of Medicine, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo Leon 64460, Mexico
- Neurometabolism Unit, Center for Research and Development in Health Sciences, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo Leon 64460, Mexico
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Queretaro 76230, Mexico
| | - Gabriela Cruz-Carrillo
- Department of Biochemistry, College of Medicine, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo Leon 64460, Mexico
- Neurometabolism Unit, Center for Research and Development in Health Sciences, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo Leon 64460, Mexico
| | - Diego Angeles-Valdez
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Queretaro 76230, Mexico
| | - Eduardo A Garza-Villarreal
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Queretaro 76230, Mexico
| | - Alberto Camacho-Morales
- Department of Biochemistry, College of Medicine, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo Leon 64460, Mexico
- Neurometabolism Unit, Center for Research and Development in Health Sciences, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo Leon 64460, Mexico
| |
Collapse
|
32
|
Croen LA, Ames JL, Qian Y, Alexeeff S, Ashwood P, Gunderson EP, Wu YW, Boghossian AS, Yolken R, Van de Water J, Weiss LA. Inflammatory Conditions During Pregnancy and Risk of Autism and Other Neurodevelopmental Disorders. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:39-50. [PMID: 38045769 PMCID: PMC10689278 DOI: 10.1016/j.bpsgos.2023.09.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 12/05/2023] Open
Abstract
Background Maternal inflammation can result from immune dysregulation and metabolic perturbations during pregnancy. Whether conditions associated with inflammation during pregnancy increase the likelihood of autism spectrum disorder (ASD) or other neurodevelopmental disorders (DDs) is not well understood. Methods We conducted a case-control study among children born in California from 2011 to 2016 to investigate maternal immune-mediated and cardiometabolic conditions during pregnancy and risk of ASD (n = 311) and DDs (n = 1291) compared with children from the general population (n = 967). Data on maternal conditions and covariates were retrieved from electronic health records. Maternal genetic data were used to assess a causal relationship. Results Using multivariable logistic regression, we found that mothers with asthma were more likely to deliver infants later diagnosed with ASD (odds ratio [OR] = 1.62, 95% CI: 1.15-2.29) or DDs (OR = 1.30, 95% CI: 1.02-1.64). Maternal obesity was also associated with child ASD (OR = 1.51, 95% CI: 1.07-2.13). Mothers with both asthma and extreme obesity had the greatest odds of delivering an infant later diagnosed with ASD (OR = 16.9, 95% CI: 5.13-55.71). These increased ASD odds were observed among female children only. Polygenic risk scores for obesity, asthma, and their combination showed no association with ASD risk. Mendelian randomization did not support a causal relationship between maternal conditions and ASD. Conclusions Inflammatory conditions during pregnancy are associated with risk for neurodevelopmental disorders in children. These risks do not seem to be due to shared genetic risk; rather, inflammatory conditions may share nongenetic risk factors with neurodevelopmental disorders. Children whose mothers have both asthma and obesity during pregnancy may benefit from earlier screening and intervention.
Collapse
Affiliation(s)
- Lisa A. Croen
- Division of Research, Kaiser Permanente Northern California, Oakland, California
- Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, California
| | - Jennifer L. Ames
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Yinge Qian
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Stacey Alexeeff
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology and the MIND Institute, University of California, Davis, Davis, California
| | - Erica P. Gunderson
- Division of Research, Kaiser Permanente Northern California, Oakland, California
- Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, California
| | - Yvonne W. Wu
- Departments of Neurology and Pediatrics, University of California San Francisco, San Francisco, California
| | - Andrew S. Boghossian
- Institute for Human Genetics, Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California
| | - Robert Yolken
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Judy Van de Water
- Division of Rheumatology/Allergy/Clinical Immunology, Department of Internal Medicine, University of California at Davis, Davis, California
| | - Lauren A. Weiss
- Institute for Human Genetics, Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California
| |
Collapse
|
33
|
Melo GM, Capucho AM, Sacramento JF, Ponce-de-Leão J, Fernandes MV, Almeida IF, Martins FO, Conde SV. Overnutrition during Pregnancy and Lactation Induces Gender-Dependent Dysmetabolism in the Offspring Accompanied by Heightened Stress and Anxiety. Nutrients 2023; 16:67. [PMID: 38201896 PMCID: PMC10781034 DOI: 10.3390/nu16010067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Maternal obesity and gestational diabetes predispose the next generation to metabolic disturbances. Moreover, the lactation phase also stands as a critical phase for metabolic programming. Nevertheless, the precise mechanisms originating these changes remain unclear. Here, we investigate the consequences of a maternal lipid-rich diet during gestation and lactation and its impact on metabolism and behavior in the offspring. Two experimental groups of Wistar female rats were used: a control group (NC) that was fed a standard diet during the gestation and lactation periods and an overnutrition group that was fed a high-fat diet (HF, 60% lipid-rich) during the same phases. The offspring were analyzed at postnatal days 21 and 28 and at 2 months old (PD21, PD28, and PD60) for their metabolic profiles (weight, fasting glycemia insulin sensitivity, and glucose tolerance) and euthanized for brain collection to evaluate metabolism and inflammation in the hypothalamus, hippocampus, and prefrontal cortex using Western blot markers of synaptic dynamics. At 2 months old, behavioral tests for anxiety, stress, cognition, and food habits were conducted. We observed that the female offspring born from HF mothers exhibited increased weight gain and decreased glucose tolerance that attenuated with age. In the offspring males, weight gain increased at P21 and worsened with age, while glucose tolerance remained unchanged. The offspring of the HF mothers exhibited elevated levels of anxiety and stress during behavioral tests, displaying decreased predisposition for curiosity compared to the NC group. In addition, the offspring from mothers with HF showed increased food consumption and a lower tendency towards food-related aggression. We conclude that exposure to an HF diet during pregnancy and lactation induces dysmetabolism in the offspring and is accompanied by heightened stress and anxiety. There was sexual dimorphism in the metabolic traits but not behavioral phenotypes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Silvia V. Conde
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Rua Camara Pestana, 6, Edificio 2, 1150-082 Lisboa, Portugal; (G.M.M.); (A.M.C.); (J.F.S.); (J.P.-d.-L.); (M.V.F.); (I.F.A.); (F.O.M.)
| |
Collapse
|
34
|
Batorsky R, Ceasrine AM, Shook LL, Kislal S, Bordt EA, Devlin BA, Perlis RH, Slonim DK, Bilbo SD, Edlow AG. Hofbauer cells and fetal brain microglia share transcriptional profiles and responses to maternal diet-induced obesity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.16.571680. [PMID: 38187648 PMCID: PMC10769274 DOI: 10.1101/2023.12.16.571680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Maternal immune activation is associated with adverse offspring neurodevelopmental outcomes, many mediated by in utero microglial programming. As microglia remain inaccessible throughout development, identification of noninvasive biomarkers reflecting fetal brain microglial programming could permit screening and intervention. We used lineage tracing to demonstrate the shared ontogeny between fetal brain macrophages (microglia) and fetal placental macrophages (Hofbauer cells) in a mouse model of maternal diet-induced obesity, and single-cell RNA-seq to demonstrate shared transcriptional programs. Comparison with human datasets demonstrated conservation of placental resident macrophage signatures between mice and humans. Single-cell RNA-seq identified common alterations in fetal microglial and Hofbauer cell gene expression induced by maternal obesity, as well as sex differences in these alterations. We propose that Hofbauer cells, which are easily accessible at birth, provide novel insights into fetal brain microglial programs, and may facilitate the early identification of offspring vulnerable to neurodevelopmental disorders in the setting of maternal exposures.
Collapse
Affiliation(s)
| | - Alexis M. Ceasrine
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Lydia L. Shook
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sezen Kislal
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Evan A. Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin A. Devlin
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Roy H. Perlis
- Department of Psychiatry and Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Donna K. Slonim
- Department of Computer Science, Tufts University, Medford, MA
| | - Staci D. Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
- Lurie Center for Autism, Massachusetts General Hospital, Boston, MA
| | - Andrea G. Edlow
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Sheng JA, Handa RJ, Tobet SA. Evaluating different models of maternal stress on stress-responsive systems in prepubertal mice. Front Neurosci 2023; 17:1292642. [PMID: 38130695 PMCID: PMC10733493 DOI: 10.3389/fnins.2023.1292642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction Maternal adversity during pregnancy influences neurodevelopment in human and model animal offspring. Adversity can result from stressors coming from many different directions ranging from environmental to nutritional and physiological to immune (e.g., infection). Most stressors result in fetal overexposure to glucocorticoids that have been directly linked to long- and short-term negative impacts on neurological health of offspring. Neuropsychiatric diseases postulated to have fetal origins are diverse and include such things cardiovascular disease, obesity, affective disorders, and metabolic and immune disorders. Methods The experiments in the current study compare 3 stressors: prenatal exposure to dexamethasone (DEX), maternal high fat diet (HFD), and maternal caloric restriction (CR). Offspring of mothers with these treatments were examined prepubertally to evaluate stress responsiveness and stress-related behaviors in in male and female mice. Results Prenatal exposure to synthetic glucocorticoid, DEX, resulted in decreased neonatal body weights, reduced social interaction behavior, and hypoactive stress response offspring exposed to maternal DEX. Maternal CR resulted in decreased body weights and social interaction behavior in males and females and increased anxiety-like behavior and acute stress response only in males. HFD resulted in altered body weight gain in both sex offspring with decreased anxiety-like behavior in a female-biased manner. Discussion The idea that glucocorticoid responses to different stressors might serve as a common stimulus across stress paradigms is insufficient, given that different modes of prenatal stress produced differential effects. Opposite nutritional stressors produced similar outcomes for anxiety-like behavior in both sexes, social-like behavior in females, and a hyperactive adrenal stress response in males. One common theme among the three models of maternal stress (DEX, CR, and HFD) was consistent data showing their role in activating the maternal and fetal immune response. By tuning in on the more immediate immunological aspect on the developing fetus (e.g., hormones, cytokines), additional studies may tease out more direct outcomes of maternal stress in rodents and increase their translational value to human studies.
Collapse
Affiliation(s)
- Julietta A. Sheng
- Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Robert J. Handa
- Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Stuart A. Tobet
- Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Psychiatry, Mass General Hospital, Harvard Medical School, Boston, MA, United States
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
- Innovation Center on Sex Differences in Medicine, Mass General Hospital, Cambridge, MA, United States
| |
Collapse
|
36
|
Nakaki A, Crovetto F, Urru A, Piella G, Borras R, Comte V, Vellvé K, Paules C, Segalés L, Dacal M, Gomez Y, Youssef L, Casas R, Castro-Barquero S, Martín-Asuero A, Oller Guzmán T, Morilla I, Martínez-Àran A, Camacho A, Pascual Tutusaus M, Arranz A, Rebollo-Polo M, Gomez-Chiari M, Bargallo N, Pozo ÓJ, Gomez-Gomez A, Izquierdo Renau M, Eixarch E, Vieta E, Estruch R, Crispi F, Gonzalez-Ballester MA, Gratacós E. Effects of Mediterranean diet or mindfulness-based stress reduction on fetal and neonatal brain development: a secondary analysis of a randomized clinical trial. Am J Obstet Gynecol MFM 2023; 5:101188. [PMID: 37839546 DOI: 10.1016/j.ajogmf.2023.101188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/13/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND Maternal suboptimal nutrition and high stress levels are associated with adverse fetal and infant neurodevelopment. OBJECTIVE This study aimed to investigate if structured lifestyle interventions involving a Mediterranean diet or mindfulness-based stress reduction during pregnancy are associated with differences in fetal and neonatal brain development. STUDY DESIGN This was a secondary analysis of the randomized clinical trial Improving Mothers for a Better Prenatal Care Trial Barcelona that was conducted in Barcelona, Spain, from 2017 to 2020. Participants with singleton pregnancies were randomly allocated into 3 groups, namely Mediterranean diet intervention, stress reduction program, or usual care. Participants in the Mediterranean diet group received monthly individual sessions and free provision of extra-virgin olive oil and walnuts. Pregnant women in the stress reduction group underwent an 8-week mindfulness-based stress reduction program adapted for pregnancy. Magnetic resonance imaging of 90 fetal brains was performed at 36 to 39 weeks of gestation and the Neonatal Neurobehavioral Assessment Scale was completed for 692 newborns at 1 to 3 months. Fetal outcomes were the total brain volume and lobular or regional volumes obtained from a 3-dimensional reconstruction and semiautomatic segmentation of magnetic resonance images. Neonatal outcomes were the 6 clusters scores of the Neonatal Neurobehavioral Assessment Scale. Multiple regression analyses were conducted to assess the association between the interventions and the fetal and neonatal outcomes. RESULTS When compared with the usual care group, the offspring exposed to a maternal Mediterranean diet had a larger total fetal brain volume (mean, 284.11 cm3; standard deviation, 23.92 cm3 vs 294.01 cm3; standard deviation, 26.29 cm3; P=.04), corpus callosum (mean, 1.16 cm3; standard deviation, 0.19 cm3 vs 1.26 cm3; standard deviation, 0.22 cm3; P=.03), and right frontal lobe (44.20; standard deviation, 4.09 cm3 vs 46.60; standard deviation, 4.69 cm3; P=.02) volumes based on magnetic resonance imaging measures and higher scores in the Neonatal Neurobehavioral Assessment Scale clusters of autonomic stability (mean, 7.4; standard deviation, 0.9 vs 7.6; standard deviation, 0.7; P=.04), social interaction (mean, 7.5; standard deviation, 1.5 vs 7.8; standard deviation, 1.3; P=.03), and range of state (mean, 4.3; standard deviation, 1.3 vs 4.5; standard deviation, 1.0; P=.04). When compared with the usual care group, offspring from the stress reduction group had larger fetal left anterior cingulate gyri volume (1.63; standard deviation, 0.32 m3 vs 1.79; standard deviation, 0.30 cm3; P=.03) based on magnetic resonance imaging and higher scores in the Neonatal Neurobehavioral Assessment Scale for regulation of state (mean, 6.0; standard deviation, 1.8 vs 6.5; standard deviation, 1.5; P<.01). CONCLUSION Maternal structured lifestyle interventions involving the promotion of a Mediterranean diet or stress reduction during pregnancy were associated with changes in fetal and neonatal brain development.
Collapse
Affiliation(s)
- Ayako Nakaki
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (Drs Nakaki, Bargallo, Eixarch, Crispi and Gratacos); Department of Surgery and Surgical specializations, Faculty of Medicine and Helath Sciences, University of Barcelona, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Castro-Barquero, Arranz, Eixarch, Crispi and Gratacos)
| | - Francesca Crovetto
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos); Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain (Drs Crovetto, Izquierdo Renau, and Gratacos)
| | - Andrea Urru
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain (Drs Urru and Piella, Mr Comte, and Dr Gonzalez-Ballester)
| | - Gemma Piella
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain (Drs Urru and Piella, Mr Comte, and Dr Gonzalez-Ballester)
| | - Roger Borras
- Cardiovascular Institute, Hospital Clínic, IDIBAPS, Universitat Autònoma de Barcelona, Barcelona, Spain (Mr Borras); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain (Mr Borras and Dr Bargallo)
| | - Valentin Comte
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain (Drs Urru and Piella, Mr Comte, and Dr Gonzalez-Ballester)
| | - Kilian Vellvé
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos)
| | - Cristina Paules
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos)
| | - Laura Segalés
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos)
| | - Marta Dacal
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos)
| | - Yvan Gomez
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos)
| | - Lina Youssef
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos); Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona Campus, Barcelona, Spain (Dr Youssef)
| | - Rosa Casas
- Department of Internal Medicine Hospital Clinic, IDIBAPS, University of Barcelona, Barcelona, Spain (Drs Casas, Castro-Barquero, and Estruch); Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERON), Madrid, Spain (Drs Casas, Castro-Barquero, and Estruch)
| | - Sara Castro-Barquero
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos); Department of Internal Medicine Hospital Clinic, IDIBAPS, University of Barcelona, Barcelona, Spain (Drs Casas, Castro-Barquero, and Estruch); Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERON), Madrid, Spain (Drs Casas, Castro-Barquero, and Estruch)
| | - Andrés Martín-Asuero
- Instituto esMindfulness, Barcelona, Spain (Dr Martín-Asuero and Ms Oller Guzmán)
| | - Teresa Oller Guzmán
- Instituto esMindfulness, Barcelona, Spain (Dr Martín-Asuero and Ms Oller Guzmán)
| | - Ivette Morilla
- Department of Psychiatry and Psychology, Hospital Clinic, Neuroscience Institute, IDIBAPS, University of Barcelona, CIBERSAM, Barcelona, Spain (Drs Morilla, Martínez-Àran, and Vieta)
| | - Anabel Martínez-Àran
- Department of Psychiatry and Psychology, Hospital Clinic, Neuroscience Institute, IDIBAPS, University of Barcelona, CIBERSAM, Barcelona, Spain (Drs Morilla, Martínez-Àran, and Vieta)
| | - Alba Camacho
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos)
| | - Mireia Pascual Tutusaus
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos)
| | - Angela Arranz
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos)
| | - Monica Rebollo-Polo
- Diagnostic Imaging and Image Guided Therapy, Institut de Recerca Sant Joan de Dèu, Esplugues de Llobregat, Spain (Drs Rebollo-Polo and Gomez-Chiari); Radiology Department, Hôpitaux Universitaires de Genève, Geneva, Switzerland (Dr Rebollo-Polo)
| | - Marta Gomez-Chiari
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos); Diagnostic Imaging and Image Guided Therapy, Institut de Recerca Sant Joan de Dèu, Esplugues de Llobregat, Spain (Drs Rebollo-Polo and Gomez-Chiari); Diagnostic Imaging Department, Hospital Sant Joan de Dèu, Esplugues de Llobregat, Spain (Dr Gomez-Chiari)
| | - Nuria Bargallo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (Drs Nakaki, Bargallo, Eixarch, Crispi and Gratacos); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain (Mr Borras and Dr Bargallo); Radiology Department, Center of Image Diagnostic, Hospital Clínic. Facultad de Medicina, Universidad de Barcelona, Barcelona, Spain (Dr Bargallo)
| | - Óscar J Pozo
- Applied Metabolomics Research Group, IMIM-Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain (Drs Pozo and Gomez-Gomez)
| | - Alex Gomez-Gomez
- Applied Metabolomics Research Group, IMIM-Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain (Drs Pozo and Gomez-Gomez)
| | - Montserrat Izquierdo Renau
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain (Drs Crovetto, Izquierdo Renau, and Gratacos); Neonatology Department, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain (Dr Izquierdo Renau)
| | - Elisenda Eixarch
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (Drs Nakaki, Bargallo, Eixarch, Crispi and Gratacos); Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain (Drs Eixarch, Crispi, and Gratacos)
| | - Eduard Vieta
- Department of Psychiatry and Psychology, Hospital Clinic, Neuroscience Institute, IDIBAPS, University of Barcelona, CIBERSAM, Barcelona, Spain (Drs Morilla, Martínez-Àran, and Vieta)
| | - Ramon Estruch
- Department of Internal Medicine Hospital Clinic, IDIBAPS, University of Barcelona, Barcelona, Spain (Drs Casas, Castro-Barquero, and Estruch); Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERON), Madrid, Spain (Drs Casas, Castro-Barquero, and Estruch)
| | - Fàtima Crispi
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (Drs Nakaki, Bargallo, Eixarch, Crispi and Gratacos); Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain (Drs Eixarch, Crispi, and Gratacos).
| | - Miguel Angel Gonzalez-Ballester
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain (Drs Urru and Piella, Mr Comte, and Dr Gonzalez-Ballester); ICREA, Barcelona, Spain (Dr Gonzalez-Ballester)
| | - Eduard Gratacós
- BCNatal Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain (Drs Nakaki, Crovetto, Vellvé, Paules, Segalés, Ms Dacal, Drs Gomez, Youssef, Castro-Barquero, Mses Camacho and Pascual Tutsaus, and Drs Arranz, Gomez-Chairi, Eixarch, Crispi and Gratacos); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (Drs Nakaki, Bargallo, Eixarch, Crispi and Gratacos); Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain (Drs Crovetto, Izquierdo Renau, and Gratacos); Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain (Drs Eixarch, Crispi, and Gratacos)
| |
Collapse
|
37
|
Pulli EP, Nolvi S, Eskola E, Nordenswan E, Holmberg E, Copeland A, Kumpulainen V, Silver E, Merisaari H, Saunavaara J, Parkkola R, Lähdesmäki T, Saukko E, Kataja E, Korja R, Karlsson L, Karlsson H, Tuulari JJ. Structural brain correlates of non-verbal cognitive ability in 5-year-old children: Findings from the FinnBrain birth cohort study. Hum Brain Mapp 2023; 44:5582-5601. [PMID: 37606608 PMCID: PMC10619410 DOI: 10.1002/hbm.26463] [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: 03/28/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/23/2023] Open
Abstract
Non-verbal cognitive ability predicts multiple important life outcomes, for example, school and job performance. It has been associated with parieto-frontal cortical anatomy in prior studies in adult and adolescent populations, while young children have received relatively little attention. We explored the associations between cortical anatomy and non-verbal cognitive ability in 165 5-year-old participants (mean scan age 5.40 years, SD 0.13; 90 males) from the FinnBrain Birth Cohort study. T1-weighted brain magnetic resonance images were processed using FreeSurfer. Non-verbal cognitive ability was measured using the Performance Intelligence Quotient (PIQ) estimated from the Block Design and Matrix Reasoning subtests from the Wechsler Preschool and Primary Scale of Intelligence (WPPSI-III). In vertex-wise general linear models, PIQ scores associated positively with volumes in the left caudal middle frontal and right pericalcarine regions, as well as surface area in left the caudal middle frontal, left inferior temporal, and right lingual regions. There were no associations between PIQ and cortical thickness. To the best of our knowledge, this is the first study to examine structural correlates of non-verbal cognitive ability in a large sample of typically developing 5-year-olds. The findings are generally in line with prior findings from older age groups, with the important addition of the positive association between volume / surface area in the right medial occipital region and non-verbal cognitive ability. This finding adds to the literature by discovering a new brain region that should be considered in future studies exploring the role of cortical structure for cognitive development in young children.
Collapse
Affiliation(s)
- Elmo P. Pulli
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
| | - Saara Nolvi
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
- Turku Institute for Advanced Studies, Department of Psychology and Speech‐Language PathologyUniversity of TurkuTurkuFinland
| | - Eeva Eskola
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
- Department of PsychologyUniversity of TurkuTurkuFinland
| | - Elisabeth Nordenswan
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
| | - Eeva Holmberg
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
| | - Anni Copeland
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
| | - Venla Kumpulainen
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
| | - Eero Silver
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
| | - Harri Merisaari
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
- Department of RadiologyUniversity of TurkuTurkuFinland
| | - Jani Saunavaara
- Department of Medical PhysicsTurku University Hospital and University of TurkuTurkuFinland
| | - Riitta Parkkola
- Department of RadiologyUniversity of TurkuTurkuFinland
- Department of RadiologyTurku University HospitalTurkuFinland
| | - Tuire Lähdesmäki
- Pediatric Neurology, Department of Pediatrics and Adolescent MedicineTurku University Hospital and University of TurkuTurkuFinland
| | | | - Eeva‐Leena Kataja
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
| | - Riikka Korja
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
- Department of PsychologyUniversity of TurkuTurkuFinland
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
- Department of Pediatrics and Adolescent MedicineTurku University Hospital and University of TurkuTurkuFinland
| | - Hasse Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
- Department of PsychiatryTurku University Hospital and University of TurkuTurkuFinland
| | - Jetro J. Tuulari
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
- Department of PsychiatryTurku University Hospital and University of TurkuTurkuFinland
- Turku Collegium for Science, Medicine and TechnologyUniversity of TurkuTurkuFinland
- Department of PsychiatryUniversity of OxfordOxfordUK
| |
Collapse
|
38
|
Marzola P, Melzer T, Pavesi E, Gil-Mohapel J, Brocardo PS. Exploring the Role of Neuroplasticity in Development, Aging, and Neurodegeneration. Brain Sci 2023; 13:1610. [PMID: 38137058 PMCID: PMC10741468 DOI: 10.3390/brainsci13121610] [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: 10/23/2023] [Revised: 11/16/2023] [Accepted: 11/18/2023] [Indexed: 12/24/2023] Open
Abstract
Neuroplasticity refers to the ability of the brain to reorganize and modify its neural connections in response to environmental stimuli, experience, learning, injury, and disease processes. It encompasses a range of mechanisms, including changes in synaptic strength and connectivity, the formation of new synapses, alterations in the structure and function of neurons, and the generation of new neurons. Neuroplasticity plays a crucial role in developing and maintaining brain function, including learning and memory, as well as in recovery from brain injury and adaptation to environmental changes. In this review, we explore the vast potential of neuroplasticity in various aspects of brain function across the lifespan and in the context of disease. Changes in the aging brain and the significance of neuroplasticity in maintaining cognitive function later in life will also be reviewed. Finally, we will discuss common mechanisms associated with age-related neurodegenerative processes (including protein aggregation and accumulation, mitochondrial dysfunction, oxidative stress, and neuroinflammation) and how these processes can be mitigated, at least partially, by non-invasive and non-pharmacologic lifestyle interventions aimed at promoting and harnessing neuroplasticity.
Collapse
Affiliation(s)
- Patrícia Marzola
- Department of Morphological Sciences and Graduate Neuroscience Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil; (P.M.); (T.M.); (E.P.)
| | - Thayza Melzer
- Department of Morphological Sciences and Graduate Neuroscience Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil; (P.M.); (T.M.); (E.P.)
| | - Eloisa Pavesi
- Department of Morphological Sciences and Graduate Neuroscience Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil; (P.M.); (T.M.); (E.P.)
| | - Joana Gil-Mohapel
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
- Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada
| | - Patricia S. Brocardo
- Department of Morphological Sciences and Graduate Neuroscience Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil; (P.M.); (T.M.); (E.P.)
| |
Collapse
|
39
|
Lee JY, Lee HJ, Jang YH, Kim H, Im K, Yang S, Hoh JK, Ahn JH. Maternal pre-pregnancy obesity affects the uncinate fasciculus white matter tract in preterm infants. Front Pediatr 2023; 11:1225960. [PMID: 38034827 PMCID: PMC10684693 DOI: 10.3389/fped.2023.1225960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
Background A growing body of evidence suggests an association between a higher maternal pre-pregnancy body mass index (BMI) and adverse long-term neurodevelopmental outcomes for their offspring. Despite recent attention to the effects of maternal obesity on fetal and neonatal brain development, changes in the brain microstructure of preterm infants born to mothers with pre-pregnancy obesity are still not well understood. This study aimed to detect the changes in the brain microstructure of obese mothers in pre-pregnancy and their offspring born as preterm infants using diffusion tensor imaging (DTI). Methods A total of 32 preterm infants (born to 16 mothers with normal BMI and 16 mothers with a high BMI) at <32 weeks of gestation without brain injury underwent brain magnetic resonance imaging at term-equivalent age (TEA). The BMI of all pregnant women was measured within approximately 12 weeks before pregnancy or the first 2 weeks of gestation. We analyzed the brain volume using a morphologically adaptive neonatal tissue segmentation toolbox and calculated the major white matter (WM) tracts using probabilistic maps of the Johns Hopkins University neonatal atlas. We investigated the differences in brain volume and WM microstructure between preterm infants of mothers with normal and high BMI. The DTI parameters were compared among groups using analysis of covariance adjusted for postmenstrual age at scan and multiple comparisons. Results Preterm infants born to mothers with a high BMI showed significantly increased cortical gray matter volume (p = 0.001) and decreased WM volume (p = 0.003) after controlling for postmenstrual age and multiple comparisons. We found a significantly lower axial diffusivity in the uncinate fasciculus (UNC) in mothers with high BMI than that in mothers with normal BMI (1.690 ± 0.066 vs. 1.762 ± 0.101, respectively; p = 0.005). Conclusion Our study is the first to demonstrate that maternal obesity impacts perinatal brain development patterns in preterm infants at TEA, even in the absence of apparent brain injury. These findings provide evidence for the detrimental effects of maternal obesity on brain developmental trajectories in offspring and suggest potential neurodevelopmental outcomes based on an altered UNC WM microstructure, which is known to be critical for language and social-emotional functions.
Collapse
Affiliation(s)
- Joo Young Lee
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science and Engineering, Seoul, Republic of Korea
| | - Hyun Ju Lee
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
- Division of Neonatology and Development Medicine, Hanyang University Hospital, Seoul, Republic of Korea
| | - Yong Hun Jang
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science and Engineering, Seoul, Republic of Korea
| | - Hyuna Kim
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science and Engineering, Seoul, Republic of Korea
| | - Kiho Im
- Fetal Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
- Division of Newborn Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Seung Yang
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
- Department of Pediatrics, Hanyang University Hospital, Seoul, Republic of Korea
| | - Jeong-Kyu Hoh
- Department of Obstetrics and Gynecology, Hanyang University College of Medicine, Seoul, Republic of Korea
- Department of Obstetrics and Gynecology, Hanyang University Hospital, Seoul, Republic of Korea
| | - Ja-Hye Ahn
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
- Division of Neonatology and Development Medicine, Hanyang University Hospital, Seoul, Republic of Korea
| |
Collapse
|
40
|
Chen S, Wang X, Lee BK, Gardner RM. Associations between maternal metabolic conditions and neurodevelopmental conditions in offspring: the mediating effects of obstetric and neonatal complications. BMC Med 2023; 21:422. [PMID: 37936224 PMCID: PMC10631144 DOI: 10.1186/s12916-023-03116-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/16/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Maternal pre-gestational diabetes (PGDM), gestational diabetes mellitus (GDM), and overweight/obesity have been associated with increased risks of offspring neurodevelopmental conditions (NDCs) including autism, intellectual disability (ID), and attention deficit/hyperactivity disorder (ADHD). Less is known about whether and how obstetric and neonatal complications (e.g., preterm birth, neonatal asphyxia) could mediate these associations. METHODS In this Swedish register-based cohort study, we examined complications during pregnancy, delivery, and the neonatal period as potential mediators of the relationships between maternal metabolic conditions and offspring NDCs. We quantified the extent to which these obstetric and neonatal factors could mediate the associations of maternal metabolic conditions with offspring NDCs by applying parametric regression models for single mediation analyses and weighting-based methods for multiple mediation analyses under counterfactual frameworks. RESULTS The study sample included 2,352,969 singleton children born to 1,299,692 mothers from 1987-2010 who were followed up until December 31, 2016, of whom 135,832 children (5.8%) were diagnosed with at least one NDC. A substantial portion of the association between maternal PGDM and children's odds of NDCs could be explained by the combined group of obstetric and neonatal complications in the multiple mediation analysis. For instance, these complications explained 44.4% of the relationship between maternal PGDM and offspring ID risk. The proportion of the relationship between maternal overweight/obesity and children's risk of NDCs that could be explained by obstetric and neonatal complications was considerably smaller, ranging from 1.5 to 8.1%. Some complications considered on their own, including pregnancy hypertensive diseases, preterm birth, neonatal asphyxia, and hematological comorbidities, could explain at least 10% of the associations between maternal PGDM and offspring NDCs. Complications during the neonatal period showed a stronger joint mediating effect for the relationship between PGDM and offspring NDCs than those during pregnancy or delivery. CONCLUSIONS Obstetric and neonatal complications could explain nearly half of the association between maternal PGDM and offspring risk of NDCs. The mediating effects were more pronounced for complications during the neonatal period and for specific complications such as pregnancy hypertensive diseases, preterm birth, neonatal asphyxia, and hematological comorbidities. Effective preventive strategies for offspring NDCs should holistically address both the primary metabolic issues related to PGDM and the wide array of potential complications, especially those in the neonatal period.
Collapse
Affiliation(s)
- Shuyun Chen
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden.
| | - Xi Wang
- PolicyLab, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Brian K Lee
- Department of Epidemiology and Biostatistics, Drexel University School of Public Health, Philadelphia, PA, USA
- A.J. Drexel Autism Institute, Philadelphia, PA, USA
| | - Renee M Gardner
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
41
|
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
|
42
|
Wu D, Li Y, Chen L, Klein M, Franke B, Chen J, Buitelaar J. Maternal gestational weight gain and offspring's neurodevelopmental outcomes: A systematic review and meta-analysis. Neurosci Biobehav Rev 2023; 153:105360. [PMID: 37573899 DOI: 10.1016/j.neubiorev.2023.105360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023]
Abstract
Abnormal gestational weight gain (GWG) has been increasing globally, up to 47% of all pregnancies. Multiple studies have focused on the association between GWG and adverse neurodevelopmental outcomes in the offspring, however with inconsistent results. We performed a systematic review and meta-analysis to evaluate associations between excessive or insufficient GWG and offspring's neurodevelopmental outcomes. Meta-analysis of these 23 studies using a random-effects model revealed associations between excessive GWG and neurodevelopmental disorders (ASD & ID & ADHD together: OR=1.12 [95% CI 1.06-1.19]), ASD (OR=1.18 [95% CI 1.08-1.29]), ADHD (OR=1.08 [95% CI 1.02-1.14]), ASD with ID (OR=1.15 [95% CI 1.01-1.32]), and ASD without ID (OR=1.12 [95% CI 1.06-1.19]). Insufficient GWG was associated with higher risk for ID (OR=1.14 [95% CI 1.03-1.26]). These results emphasize the significant impact, though of small effect size, of GWG across multiple neurodevelopmental disorders. It is important to note that these results do not establish causality. Other factors such as genetic factors, gene-environment interactions may confound the relationship between GWG and neurodevelopmental outcomes. To better understand the role of GWG in neurodevelopmental disorders, future studies should consider using genetically sensitive designs that can account for these potential confounders.
Collapse
Affiliation(s)
- Dan Wu
- Department of Child Health Care, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200062, Shanghai, China; Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Yicheng Li
- Department of Child Health Care, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200062, Shanghai, China
| | - Lingyan Chen
- Department of Occupational Therapy Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki-city, 852-8520, Japan
| | - Marieke Klein
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Barbara Franke
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Department of Psychiatry, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Jinjin Chen
- Department of Child Health Care, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200062, Shanghai, China.
| | - Jan Buitelaar
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Karakter Child and Adolescent Psychiatry University Centre, 6525 GA Nijmegen, the Netherlands
| |
Collapse
|
43
|
Wang WC, Ding M, Strohmaier S, Schernhammer E, Sun Q, Chavarro JE, Tiemeier H. Maternal adherence to healthy lifestyle and risk of depressive symptoms in the offspring: mediation by offspring lifestyle. Psychol Med 2023; 53:6068-6076. [PMID: 36377496 DOI: 10.1017/s0033291722003257] [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] [Indexed: 11/16/2022]
Abstract
BACKGROUND Adherence to healthy lifestyles can be beneficial for depression among adults, but the intergenerational impact of maternal healthy lifestyles on offspring depressive symptoms is unknown. METHODS In total, 10 368 mothers in Nurses' Health Study II and 13 478 offspring in the Growing Up Today Study were paired. Maternal and offspring healthy lifestyles were defined as a composite score including a healthy diet, normal body mass index (BMI), never-smoking, light-to-moderate consumption of alcohol, and regular moderate-to-vigorous physical activity. Maternal lifestyles were assessed during their offspring's childhood. Offspring depressive symptoms were repeatedly assessed five times using the Center for Epidemiological Studies Depression Scale-10 (CESD-10); the offspring were between the ages of 14 and 30 when the first CESD-10 was assessed. Covariates included maternal variables (age at baseline, race/ethnicity, antidepressant use, pregnancy complications, etc.) and offspring age and sex. RESULTS Children of mothers with the healthiest lifestyle had significantly fewer depressive symptoms (a 0.30 lower CESD-10 score, 95% confidence interval (CI) 0.09-0.50) in comparison with children of mothers with the least healthy lifestyle. The association was only found significant in female offspring but not in males. For individual maternal lifestyle factors, a normal BMI, never-smoking, and adherence to regular physical activity were independently associated with fewer depressive symptoms among the offspring. The association between maternal healthy lifestyles and offspring depressive symptoms was mediated by offspring's healthy lifestyles (mediation effect: 53.2%, 95% CI 15.8-87.3). CONCLUSIONS Our finding indicates the potential mechanism of intergenerational transmission of healthy lifestyles to reduce the risk of depressive symptoms in offspring.
Collapse
Affiliation(s)
- Wei-Chen Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ming Ding
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Susanne Strohmaier
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, Vienna, Austria
| | - Eva Schernhammer
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, Vienna, Austria
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Qi Sun
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Jorge E Chavarro
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Henning Tiemeier
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Social and Behavioral Science, Harvard T.H. Chan School of Public Health, 677 Huntington Ave., Boston, MA 02115, USA
| |
Collapse
|
44
|
Dow C, Galera C, Charles MA, Heude B. Maternal pre-pregnancy BMI and offspring hyperactivity-inattention trajectories from 3 to 8 years in the EDEN birth cohort study. Eur Child Adolesc Psychiatry 2023; 32:2057-2065. [PMID: 35851811 DOI: 10.1007/s00787-022-02047-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 07/06/2022] [Indexed: 12/15/2022]
Abstract
Evidence suggests obesity during pregnancy is associated with offspring attention-deficit hyperactivity disorder. However, studies have been limited to evaluating the association at a single age with inadequate data on important maternal lifestyle confounders and unmeasured familial confounding. The objective of this study was to examine the association between maternal pre-pregnancy body mass index (BMI) and child hyperactivity-inattention symptoms (HIS) at 3, 5 and 8 years. Data came from the EDEN mother-child cohort. Maternal pre-pregnancy BMI status (kg/m2) was calculated using pre-pregnancy weight and height (self-reported by mothers or measured by midwives). HIS were assessed by parental-report on the Strengths and Difficulties Questionnaire at 3, 5 and 8 years of age and used to derive developmental trajectories of HIS (n = 1428). Multivariate models were adjusted for confounders including socioeconomic status, maternal lifestyle behaviours (exercise, diet, smoking, alcohol), childcare and a stimulating home environment. Paternal BMI was used as a negative control. Compared to a normal pre-pregnancy BMI, pre-pregnancy maternal obesity was positively associated with increased odds of a high HIS trajectory between 3 and 8 years old in both unadjusted and adjusted logistic regression (adjusted odds ratio [aOR] 1.87 [95% CI 1.12, 3.12]). Pre-pregnancy overweight was not significantly associated after adjustment for confounders (aOR 1.32 [0.87, 2.01]). Maternal pre-pregnancy obesity, but not overweight, was associated with increased likelihood of a high HIS trajectory in children from 3 to 8 years old. This association persisted despite controlling for many important maternal lifestyle factors and paternal BMI. Further research is warranted to identify possible mediators involved.
Collapse
Affiliation(s)
- Courtney Dow
- INSERM, UMR1153 Epidemiology and Biostatistics Sorbonne Paris Cité Center (CRESS), ORCHAD Team, Paris, France.
| | - Cédric Galera
- Univ. Bordeaux, Inserm, Bordeaux Population Health Center, UMR 1219, 33000, Bordeaux, France
- Centre Hospitalier Perrens, Bordeaux, France
| | - Marie-Aline Charles
- INSERM, UMR1153 Epidemiology and Biostatistics Sorbonne Paris Cité Center (CRESS), ORCHAD Team, Paris, France
| | - Barbara Heude
- INSERM, UMR1153 Epidemiology and Biostatistics Sorbonne Paris Cité Center (CRESS), ORCHAD Team, Paris, France
| |
Collapse
|
45
|
Kwok J, Khanolainen DP, Speyer LG, Murray AL, Torppa MP, Auyeung B. Examining Maternal Cardiometabolic Markers in Pregnancy on Child Emotional and Behavior Trajectories: Using Growth Curve Models on a Cohort Study. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:614-622. [PMID: 37881536 PMCID: PMC10593919 DOI: 10.1016/j.bpsgos.2023.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 10/27/2023] Open
Abstract
Background Poor maternal cardiometabolic health in pregnancy is associated with negative effects on child health outcomes, but there is limited literature on child and adolescent socioemotional outcomes. The study aimed to investigate associations between maternal cardiometabolic markers during pregnancy with child and adolescent socioemotional trajectories. Methods Growth curve models were run to examine how maternal cardiometabolic markers in pregnancy affected child socioemotional trajectories from ages 4 to 16. Models were adjusted for all pregnancy trimesters and maternal, child, and socioeconomic covariates. This study used the Avon Longitudinal Study of Parents and Children (United Kingdom) cohort. Participants consisted of mother-child pairs (N = 15,133). Maternal predictors of fasting glucose, triglycerides, high-density lipoprotein, low-density lipoprotein, and body mass index were taken from each pregnancy trimester (T1, T2, T3). Child outcomes included emotional problems, conduct problems, and hyperactivity problems from the Strengths and Difficulties Questionnaire. Results Fully adjusted models showed significant associations between elevated T1 fasting glucose and increased conduct problems, higher T1 body mass index and increased hyperactivity problems, lowered T1 high-density lipoprotein and decreased hyperactivity problems, and elevated T2 triglycerides and increased hyperactivity problems. Conclusions Maternal cardiometabolic risk is associated with conduct and hyperactivity outcomes from ages 4 to 16. This study suggests that maternal markers of fasting glucose, low-density lipoprotein, high-density lipoprotein, and triglycerides during pregnancy could be added as supplements for clinical measures of risk when predicting child and adolescent socioemotional trajectories.
Collapse
Affiliation(s)
- Janell Kwok
- School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | | | - Lydia G. Speyer
- School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, United Kingdom
- Department of Psychology, Lancaster University, Lancaster, United Kingdom
| | - Aja L. Murray
- School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Minna P. Torppa
- Department of Teacher Education, University of Jyväskylä, Jyväskylä, Finland
| | - Bonnie Auyeung
- School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, United Kingdom
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
46
|
Powell TL, Uhlson C, Madi L, Berry KZ, Chassen SS, Jansson T, Ferchaud-Roucher V. Fetal sex differences in placental LCPUFA ether and plasmalogen phosphatidylethanolamine and phosphatidylcholine contents in pregnancies complicated by obesity. Biol Sex Differ 2023; 14:66. [PMID: 37770949 PMCID: PMC10540428 DOI: 10.1186/s13293-023-00548-1] [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: 05/15/2023] [Accepted: 09/13/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND We have previously reported that maternal obesity reduces placental transport capacity for lysophosphatidylcholine-docosahexaenoic acid (LPC-DHA), a preferred form for transfer of DHA (omega 3) to the fetal brain, but only in male fetuses. Phosphatidylethanolamine (PE) and phosphatidylcholine (PC), have either sn-1 ester, ether or vinyl ether (plasmalogen) linkages to primarily unsaturated and monounsaturated fatty acids and DHA or arachidonic acid (ARA, omega 6) in the sn-2 position. Whether ether and plasmalogen PC and PE metabolism in placenta impacts transfer to the fetus is unexplored. We hypothesized that ether and plasmalogen PC and PE containing DHA and ARA are reduced in maternal-fetal unit in pregnancies complicated by obesity and these differences are dependent on fetal sex. METHODS In maternal, umbilical cord plasma and placentas from obese women (11 female/5 male infants) and normal weight women (9 female/7 male infants), all PC and PE species containing DHA and ARA were analyzed by LC-MS/MS. Placental protein expression of enzymes involved in phospholipid synthesis, were determined by immunoblotting. All variables were compared between control vs obese groups and separated by fetal sex, in each sample using the Benjamini-Hochberg false discovery rate adjustment to account for multiple testing. RESULTS Levels of ester PC containing DHA and ARA were profoundly reduced by 60-92% in male placentas of obese mothers, while levels of ether and plasmalogen PE containing DHA and ARA were decreased by 51-84% in female placentas. PLA2G4C abundance was lower in male placentas and LPCAT4 abundance was lower solely in females in obesity. In umbilical cord, levels of ester, ether and plasmalogen PC and PE with DHA were reduced by 43-61% in male, but not female, fetuses of obese mothers. CONCLUSIONS We found a fetal sex effect in placental PE and PC ester, ether and plasmalogen PE and PC containing DHA in response to maternal obesity which appears to reflect an ability of female placentas to adapt to maintain optimal fetal DHA transfer in maternal obesity.
Collapse
Affiliation(s)
- Theresa L Powell
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Charis Uhlson
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lana Madi
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Karin Zemski Berry
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephanie S Chassen
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Veronique Ferchaud-Roucher
- Nantes Université, CHU Nantes, INRAE UMR 1280 PhAN, CRNH Ouest, 44000, Nantes, France.
- Nantes Université, INRAE, UMR 1280 PhAN, CHU Hôtel Dieu, HNB1, 1 place Alexis Ricordeau, 44093, Nantes, France.
| |
Collapse
|
47
|
Custodio RJP, Hobloss Z, Myllys M, Hassan R, González D, Reinders J, Bornhorst J, Weishaupt AK, Seddek AL, Abbas T, Friebel A, Hoehme S, Getzmann S, Hengstler JG, van Thriel C, Ghallab A. Cognitive Functions, Neurotransmitter Alterations, and Hippocampal Microstructural Changes in Mice Caused by Feeding on Western Diet. Cells 2023; 12:2331. [PMID: 37759553 PMCID: PMC10529844 DOI: 10.3390/cells12182331] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Metabolic Dysfunction Associated Steatotic Liver Disease (MASLD) is the most common chronic liver disease in Western countries. It is becoming increasingly evident that peripheral organ-centered inflammatory diseases, including liver diseases, are linked with brain dysfunctions. Therefore, this study aims to unravel the effect of MASLD on brain histology, cognitive functions, and neurotransmitters. For this purpose, mice fed for 48 weeks on standard (SD) or Western diet (WD) were evaluated by behavioral tests, followed by sacrifice and analysis of the liver-brain axis including histopathology, immunohistochemistry, and biochemical analyses. Histological analysis of the liver showed features of Metabolic Dysfunction-Associated Steatohepatitis (MASH) in the WD-fed mice including lipid droplet accumulation, inflammation, and fibrosis. This was accompanied by an elevation of transaminase and alkaline phosphatase activities, increase in inflammatory cytokine and bile acid concentrations, as well as altered amino acid concentrations in the blood. Interestingly, compromised blood capillary morphology coupled with astrogliosis and microgliosis were observed in brain hippocampus of the WD mice, indicating neuroinflammation or a disrupted neurovascular unit. Moreover, attention was impaired in WD-fed mice along with the observations of impaired motor activity and balance, enhanced anxiety, and stereotyped head-twitch response (HTR) behaviors. Analysis of neurotransmitters and modulators including dopamine, serotonin, GABA, glutamate, and acetylcholine showed region-specific dysregulation in the brain of the WD-fed mice. In conclusion, the induction of MASH in mice is accompanied by the alteration of cellular morphology and neurotransmitter expression in the brain, associated with compromised cognitive functions.
Collapse
Affiliation(s)
- Raly James Perez Custodio
- Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany; (R.J.P.C.); (Z.H.); (M.M.); (R.H.); (D.G.); (J.R.); (S.G.)
| | - Zaynab Hobloss
- Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany; (R.J.P.C.); (Z.H.); (M.M.); (R.H.); (D.G.); (J.R.); (S.G.)
| | - Maiju Myllys
- Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany; (R.J.P.C.); (Z.H.); (M.M.); (R.H.); (D.G.); (J.R.); (S.G.)
| | - Reham Hassan
- Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany; (R.J.P.C.); (Z.H.); (M.M.); (R.H.); (D.G.); (J.R.); (S.G.)
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt;
| | - Daniela González
- Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany; (R.J.P.C.); (Z.H.); (M.M.); (R.H.); (D.G.); (J.R.); (S.G.)
| | - Jörg Reinders
- Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany; (R.J.P.C.); (Z.H.); (M.M.); (R.H.); (D.G.); (J.R.); (S.G.)
| | - Julia Bornhorst
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstraße 20, 42119 Wuppertal, Germany; (J.B.); (A.-K.W.)
| | - Ann-Kathrin Weishaupt
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstraße 20, 42119 Wuppertal, Germany; (J.B.); (A.-K.W.)
| | - Abdel-latif Seddek
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt;
| | - Tahany Abbas
- Histology Department, Faculty of Medicine, South Valley University, Qena 83523, Egypt;
| | - Adrian Friebel
- Institute of Computer Science & Saxonian Incubator for Clinical Research (SIKT), University of Leipzig, Haertelstraße 16-18, 04107 Leipzig, Germany; (A.F.); (S.H.)
| | - Stefan Hoehme
- Institute of Computer Science & Saxonian Incubator for Clinical Research (SIKT), University of Leipzig, Haertelstraße 16-18, 04107 Leipzig, Germany; (A.F.); (S.H.)
| | - Stephan Getzmann
- Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany; (R.J.P.C.); (Z.H.); (M.M.); (R.H.); (D.G.); (J.R.); (S.G.)
| | - Jan G. Hengstler
- Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany; (R.J.P.C.); (Z.H.); (M.M.); (R.H.); (D.G.); (J.R.); (S.G.)
| | - Christoph van Thriel
- Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany; (R.J.P.C.); (Z.H.); (M.M.); (R.H.); (D.G.); (J.R.); (S.G.)
| | - Ahmed Ghallab
- Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany; (R.J.P.C.); (Z.H.); (M.M.); (R.H.); (D.G.); (J.R.); (S.G.)
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt;
| |
Collapse
|
48
|
Cerdó T, Nieto-Ruíz A, García-Santos JA, Rodríguez-Pöhnlein A, García-Ricobaraza M, Suárez A, Bermúdez MG, Campoy C. Current Knowledge About the Impact of Maternal and Infant Nutrition on the Development of the Microbiota-Gut-Brain Axis. Annu Rev Nutr 2023; 43:251-278. [PMID: 37603431 DOI: 10.1146/annurev-nutr-061021-025355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
The prenatal and early postnatal periods are stages during which dynamic changes and the development of the brain and gut microbiota occur, and nutrition is one of the most important modifiable factors that influences this process. Given the bidirectional cross talk between the gut microbiota and the brain through the microbiota-gut-brain axis (MGBA), there is growing interest in evaluating the potential effects of nutritional interventions administered during these critical developmental windows on gut microbiota composition and function and their association with neurodevelopmental outcomes. We review recent preclinical and clinical evidence from animal studies and infant/child populations. Although further research is needed, growing evidence suggests that different functional nutrients affect the establishment and development of the microbiota-gut-brain axis and could have preventive and therapeutic use in the treatment of neuropsychiatric disorders. Therefore, more in-depth knowledge regarding the effect of nutrition on the MGBA during critical developmental windows may enable the prevention of later neurocognitive and behavioral disorders and allow the establishment of individualized nutrition-based programs that can be used from the prenatal to the early and middle stages of life.
Collapse
Affiliation(s)
- Tomás Cerdó
- Maimonides Institute for Research in Biomedicine of Córdoba, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
| | - Ana Nieto-Ruíz
- Department of Paediatrics, Faculty of Medicine, University of Granada, Granada, Spain;
- Instituto de Investigación Biosanitaria (IBS-GRANADA), Granada, Spain
- Instituto de Neurociencias "Doctor Federico Olóriz," Biomedical Research Centre, University of Granada, Granada, Spain
| | - José Antonio García-Santos
- Department of Paediatrics, Faculty of Medicine, University of Granada, Granada, Spain;
- Instituto de Investigación Biosanitaria (IBS-GRANADA), Granada, Spain
- Instituto de Neurociencias "Doctor Federico Olóriz," Biomedical Research Centre, University of Granada, Granada, Spain
| | - Anna Rodríguez-Pöhnlein
- Department of Paediatrics, Faculty of Medicine, University of Granada, Granada, Spain;
- Instituto de Investigación Biosanitaria (IBS-GRANADA), Granada, Spain
- Instituto de Neurociencias "Doctor Federico Olóriz," Biomedical Research Centre, University of Granada, Granada, Spain
| | - María García-Ricobaraza
- Department of Paediatrics, Faculty of Medicine, University of Granada, Granada, Spain;
- Instituto de Investigación Biosanitaria (IBS-GRANADA), Granada, Spain
- Instituto de Neurociencias "Doctor Federico Olóriz," Biomedical Research Centre, University of Granada, Granada, Spain
| | - Antonio Suárez
- Department of Biochemistry and Molecular Biology 2, Faculty of Pharmacy, University of Granada, Granada, Spain
- Instituto de Nutrición y Tecnología de los Alimentos, Biomedical Research Centre, University of Granada, Granada, Spain
| | - Mercedes G Bermúdez
- Department of Paediatrics, Faculty of Medicine, University of Granada, Granada, Spain;
- Instituto de Investigación Biosanitaria (IBS-GRANADA), Granada, Spain
- Instituto de Neurociencias "Doctor Federico Olóriz," Biomedical Research Centre, University of Granada, Granada, Spain
| | - Cristina Campoy
- Department of Paediatrics, Faculty of Medicine, University of Granada, Granada, Spain;
- Instituto de Investigación Biosanitaria (IBS-GRANADA), Granada, Spain
- Instituto de Neurociencias "Doctor Federico Olóriz," Biomedical Research Centre, University of Granada, Granada, Spain
- Spanish Network of Biomedical Research in Epidemiology and Public Health, Granada Node, Carlos III Health Institute, Madrid, Spain
| |
Collapse
|
49
|
Mort EJ, Fordington S, Heritage S, Fowden AL, Jones S, Camm EJ. Age and an obesogenic diet affect mouse behaviour in a sex-dependent manner. Eur J Neurosci 2023; 58:2451-2468. [PMID: 37377042 PMCID: PMC10946847 DOI: 10.1111/ejn.16070] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/29/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023]
Abstract
Obesity is rising globally and is associated with neurodevelopmental and psychiatric disorders among children, adolescents and young adults. Whether obesity is the cause or the consequence of these disorders remains unclear. To examine the behavioural effects of obesity systematically, locomotion, anxiety and social behaviour were assessed in male and female C57Bl/6J mice using the open field, elevated plus maze and social preference task. First, the effects of age and sex were examined in control mice, before investigating post-weaning consumption of a high fat-high sugar diet commonly consumed in human populations with high rates of obesity. In the open field and elevated plus maze, locomotor activity and anxiety-related behaviours reduced with aging in both sexes, but with different sex-specific profiles. The high fat-high sugar diet reduced food and calorie intake and increased body mass and fat deposition in both sexes. In the open field, both male and female mice on the obesogenic diet showed reduced locomotion; whereas, in the elevated plus maze, only females fed with the obesogenic diet displayed reduced anxiety-related behaviours. Both male and female mice on the obesogenic diet had a significantly higher social preference index than the control group. In conclusion, the findings demonstrate that the behavioural effects of age and diet-induced obesity all depend on the sex of the mouse. This emphasises the importance of considering the age of the animal and including both sexes when assessing behavioural phenotypes arising from dietary manipulations.
Collapse
Affiliation(s)
- Emily J. Mort
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Surina Fordington
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Sophie Heritage
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Abigail L. Fowden
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Susan Jones
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Emily J. Camm
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
| |
Collapse
|
50
|
Witek K, Wydra K, Suder A, Filip M. Maternal monosaccharide diets evoke cognitive, locomotor, and emotional disturbances in adolescent and young adult offspring rats. Front Nutr 2023; 10:1176213. [PMID: 37229474 PMCID: PMC10203434 DOI: 10.3389/fnut.2023.1176213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/12/2023] [Indexed: 05/27/2023] Open
Abstract
Anxiety and depression are the most common mental disorders affecting people worldwide. Recent studies have highlighted that a maternal high-sugar diet (HSD) could be a risk factor for neurobehavioural dysregulations, including mood disorders. Increased consumption of added sugar in food such as refined fructose/glucose can increase the risk of metabolic disorders and impact susceptibility to mental disorders. Furthermore, a few papers have reported disabilities in learning and memory among offspring after maternal HSD, thus suggesting a relationship between maternal nutrition and offspring neurogenesis. In this study, we evaluated the impact of maternal monosaccharide consumption based on a glucose (GLU) or fructose (FRU) diet during pregnancy and lactation in adolescent and young adult offspring rats of both sexes on cognitive, locomotor, and emotional disturbances. Locomotor activity, short-term memory, anxiety-like and depressive-like behavior were evaluated in the offspring. We report for the first time that the maternal GLU or FRU diet is sufficient to evoke anxiety-like behavior among adolescent and young adult offspring. Moreover, we found that maternal monosaccharide diets lead to hyperactivity and depressive-like behavior in male adolescent rats. We also noticed that a maternal FRU diet significantly enhanced novelty-seeking behavior only in young adult male rats. Our novel findings indicated that the maternal monosaccharide diet, especially a diet enriched in FRU, resulted in strong behavioral alterations in offspring rats at early life stages. This study also revealed that male rats were more susceptible to hyperactivity and anxiety- and depressive-like phenotypes than female rats. These results suggest that maternal monosaccharide consumption during pregnancy and lactation is an important factor affecting the emotional status of offspring.
Collapse
|