1
|
Huang R, Zhou G, Cai J, Cao C, Zhu Z, Wu Q, Zhang F, Ding Y. Maternal consumption of urbanized diet compromises early-life health in association with gut microbiota. Gut Microbes 2025; 17:2483783. [PMID: 40176259 PMCID: PMC11988223 DOI: 10.1080/19490976.2025.2483783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 04/04/2025] Open
Abstract
Urbanization has significantly transformed dietary habits worldwide, contributing to a globally increased burden of non-communicable diseases and altered gut microbiota landscape. However, it is often overlooked that the adverse effects of these dietary changes can be transmitted from the mother to offspring during early developmental stages, subsequently influencing the predisposition to various diseases later in life. This review aims to delineate the detrimental effects of maternal urban-lifestyle diet (urbanized diet) on early-life health and gut microbiota assembly, provide mechanistic insights on how urbanized diet mediates mother-to-offspring transfer of bioactive substances in both intrauterine and extrauterine and thus affects fetal and neonatal development. Moreover, we also further propose a framework for developing microbiome-targeted precision nutrition and diet strategies specifically for pregnant and lactating women. The establishment of such knowledge can help develop proactive preventive measures from the beginning of life, ultimately reducing the long-term risk of disease and improving public health outcomes.
Collapse
Affiliation(s)
- Rong Huang
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Guicheng Zhou
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jie Cai
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Cha Cao
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhenjun Zhu
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Fen Zhang
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yu Ding
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
2
|
Ding L, Liu J, Zhou L, Zhang Q, Liu J, Xiao X. Maternal high-fat diet alters the transcriptional rhythm in white adipose tissue of adult offspring. J Nutr Biochem 2025; 138:109843. [PMID: 39826765 DOI: 10.1016/j.jnutbio.2025.109843] [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: 06/12/2024] [Revised: 01/11/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
A maternal high-fat diet (HFD) deteriorates the long-term metabolic health of offspring. Circadian rhythms are crucial for regulating metabolism. However, the impact of maternal HFD on the circadian clock in white adipose tissue (WAT) remains unexplored. This study aimed to identify transcriptional rhythmic alterations in inguinal WAT of adult male offspring induced by maternal HFD. To this end, female mice were fed an HFD and their male offspring were raised on a standard chow diet until 16 weeks of age. Transcriptome was performed and the data was analyzed using CircaCompare. The results showed that maternal HFD before and throughout pregnancy significantly altered the circadian rhythm of inguinal WAT while slightly modifying the WAT clock in adult male offspring. Specifically, maternal HFD contributed to gaining rhythmicity of Cry2, resulted in the elevated amplitude of Nr1d2, and led to increased midline estimating statistic of rhythm (MESOR) of Clock and Nr1d2. Furthermore, maternal HFD changed the rhythmic pattern of metabolic genes, such as Pparγ, Hacd2, and Acsl1, which are significantly enriched in metabolic regulation pathways. In conclusion, a maternal HFD before and throughout pregnancy altered the circadian rhythm of inguinal WAT in adult offspring. These alterations may play a significant role in disturbing metabolic homeostasis, potentially leading to metabolic dysfunction in adult male offspring.
Collapse
Affiliation(s)
- Lu Ding
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jing Liu
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Liyuan Zhou
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| | - Qian Zhang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jieying Liu
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH), Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Xinhua Xiao
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
3
|
Horner D, Jepsen JRM, Chawes B, Aagaard K, Rosenberg JB, Mohammadzadeh P, Sevelsted A, Vahman N, Vinding R, Fagerlund B, Pantelis C, Bilenberg N, Pedersen CET, Eliasen A, Brandt S, Chen Y, Prince N, Chu SH, Kelly RS, Lasky-Su J, Halldorsson TI, Strøm M, Strandberg-Larsen K, Olsen SF, Glenthøj BY, Bønnelykke K, Ebdrup BH, Stokholm J, Rasmussen MA. A western dietary pattern during pregnancy is associated with neurodevelopmental disorders in childhood and adolescence. Nat Metab 2025; 7:586-601. [PMID: 40033007 PMCID: PMC12022897 DOI: 10.1038/s42255-025-01230-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 02/06/2025] [Indexed: 03/05/2025]
Abstract
Despite the high prevalence of neurodevelopmental disorders, the influence of maternal diet during pregnancy on child neurodevelopment remains understudied. Here we show that a western dietary pattern during pregnancy is associated with child neurodevelopmental disorders. We analyse self-reported maternal dietary patterns at 24 weeks of pregnancy and clinically evaluated neurodevelopmental disorders at 10 years of age in the COPSAC2010 cohort (n = 508). We find significant associations with attention-deficit hyperactivity disorder (ADHD) and autism diagnoses. We validate the ADHD findings in three large, independent mother-child cohorts (n = 59,725, n = 656 and n = 348) through self-reported dietary modelling, maternal blood metabolomics and foetal blood metabolomics. Metabolome analyses identify 15 mediating metabolites in pregnancy that improve ADHD prediction. Longitudinal blood metabolome analyses, incorporating five time points per cohort in two independent cohorts, reveal that associations between western dietary pattern metabolite scores and neurodevelopmental outcomes are consistently significant in early-mid-pregnancy. These findings highlight the potential for targeted prenatal dietary interventions to prevent neurodevelopmental disorders and emphasise the importance of early intervention.
Collapse
Affiliation(s)
- David Horner
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark.
| | - Jens Richardt M Jepsen
- Center for Neuropsychiatric Schizophrenia Research (CNSR) and Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
- Mental Health Centre for Child and Adolescent Psychiatry - Research unit, Mental Health Services in the Capital Region of Denmark, Copenhagen, Denmark
| | - Bo Chawes
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Kristina Aagaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Julie B Rosenberg
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Center for Neuropsychiatric Schizophrenia Research (CNSR) and Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Parisa Mohammadzadeh
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Center for Neuropsychiatric Schizophrenia Research (CNSR) and Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Astrid Sevelsted
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Nilo Vahman
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Rebecca Vinding
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Fagerlund
- Center for Neuropsychiatric Schizophrenia Research (CNSR) and Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
- Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Niels Bilenberg
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Casper-Emil T Pedersen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Anders Eliasen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Sarah Brandt
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Yulu Chen
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicole Prince
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Su H Chu
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Thorhallur I Halldorsson
- Faculty of Food Science and Nutrition, School of Health Science, University of Iceland, Health Science Institute, Unit for Nutrition Research, Reykjavík, Iceland
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Marin Strøm
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Faculty of Health Sciences, University of the Faroe Islands, Tórshavn, Faroe Islands
| | | | - Sjurdur F Olsen
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Faculty of Health Sciences, University of the Faroe Islands, Tórshavn, Faroe Islands
- Section of Epidemiology, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Birte Y Glenthøj
- Center for Neuropsychiatric Schizophrenia Research (CNSR) and Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Bjørn H Ebdrup
- Center for Neuropsychiatric Schizophrenia Research (CNSR) and Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Stokholm
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Section of Food Microbiology, Gut Health and Fermentation, Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Morten Arendt Rasmussen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark.
- Section of Food Microbiology, Gut Health and Fermentation, Department of Food Science, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
4
|
Orchanian SB, Hsiao EY. The microbiome as a modulator of neurological health across the maternal-offspring interface. J Clin Invest 2025; 135:e184314. [PMID: 39959974 PMCID: PMC11827852 DOI: 10.1172/jci184314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025] Open
Abstract
The maternal microbiome is emerging as an important factor that influences the neurological health of mothers and their children. Recent studies highlight how microbial communities in the maternal gut can shape early-life development in ways that inform long-term health trajectories. Research on the neurodevelopmental effects of maternal microbiomes is expanding our understanding of the microbiome-gut-brain axis to include signaling across the maternal-offspring unit during the perinatal period. In this Review, we synthesize existing literature on how the maternal microbiome modulates brain function and behavior in both mothers and their developing offspring. We present evidence from human and animal studies showing that the maternal microbiome interacts with environmental factors to impact risk for neurodevelopmental abnormalities. We further discuss molecular and cellular mechanisms that facilitate maternal-offspring crosstalk for neuromodulation. Finally, we consider how advancing understanding of these complex interactions could lead to microbiome-based interventions for promoting maternal and offspring health.
Collapse
Affiliation(s)
| | - Elaine Y. Hsiao
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, California, USA
- UCLA Goodman-Luskin Microbiome Center, Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California, USA
| |
Collapse
|
5
|
Mou Y, Jansen PW, Sun H, White T, Voortman T. Diet quality during pregnancy, adolescent brain morphology, and cognitive performance in a population-based cohort. Am J Clin Nutr 2024; 120:1125-1133. [PMID: 39510724 DOI: 10.1016/j.ajcnut.2024.08.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/29/2024] [Accepted: 08/20/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Diet quality during pregnancy may affect offspring's neurobiology and cognitive performance in childhood. However, little is known about underlying mechanisms and potential long-term effects. OBJECTIVES To examine associations of diet quality during pregnancy with offspring pre- and early-adolescent brain morphology and to investigate whether brain morphology mediates associations of diet quality during pregnancy with full-scale intelligence quotient (IQ) in early adolescence. METHODS We studied 2223 and 1582 mother-child dyads with brain scans collected using magnetic resonance imaging at ages 10 and 14 y in the population-based Generation R Study in The Netherlands. We assessed dietary intake during pregnancy with 293-item food-frequency questionnaires and calculated predefined diet quality scores (total score 0-15), reflecting adherence to dietary guidelines. Cognitive performance was assessed using Wechsler Intelligence Scale for Children-V at age 14 y. We examined associations using multiple regression models, corrected for multiple testing. RESULTS After adjustment for child age, sex, socioeconomic factors, maternal age, smoking, and psychopathological symptoms during pregnancy, we found that higher diet quality during pregnancy was associated with a larger total brain (B: 4.54, 95% confidence interval [CI]: 1.80, 7.28), cerebral white matter (1.83, 95% CI: 0.56, 3.10), cerebral gray matter (1.99, 95% CI: 0.63, 3.35), and subcortical volumes (0.16, 95% CI: 0.04, 0.28) of children at age 10 y. Similar results were found for age 14 y. Widespread differences in cortical thickness, gyrification, and surface area in both hemispheres were also observed. Better diet quality during pregnancy was associated with higher full-scale IQ scores of adolescents, particularly on verbal comprehension and matrix reasoning. The associations between diet quality during pregnancy and full-scale IQ in early adolescence were partially mediated by brain volumetric markers in pre-adolescence. CONCLUSIONS Diet quality during pregnancy was associated with structural brain alterations in the offspring, which partly explained the relation between prenatal dietary patterns and cognitive outcomes in children.
Collapse
Affiliation(s)
- Yuchan Mou
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, The Netherlands; The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, The Netherlands; Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Pauline W Jansen
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, The Netherlands; Department of Psychology, Education and Child Studies, Erasmus University Rotterdam, The Netherlands
| | - Hong Sun
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, The Netherlands; The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Tonya White
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, The Netherlands; Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Trudy Voortman
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, The Netherlands; Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, United States.
| |
Collapse
|
6
|
Bucknor MC, Gururajan A, Dale RC, Hofer MJ. High fat diet consumption and social instability stress impair stress adaptation and maternal care in C57Bl/6 dams. Psychoneuroendocrinology 2024; 169:107168. [PMID: 39146876 DOI: 10.1016/j.psyneuen.2024.107168] [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: 05/09/2024] [Revised: 07/16/2024] [Accepted: 08/06/2024] [Indexed: 08/17/2024]
Abstract
Poor maternal diet and psychosocial stress represent two environmental factors that can significantly impact maternal health during pregnancy. While various mouse models have been developed to study the relationship between maternal and offspring health and behaviour, few incorporate multiple sources of stress that mirror the complexity of human experiences. Maternal high-fat diet (HF) models in rodents are well-established, whereas use of psychosocial stress interventions in female mice are still emerging. The social instability stress (SIS) paradigm, serves as a chronic and unpredictable form of social stress. To evaluate the combined effects of a poor maternal diet and intermittent social stress on maternal health and behaviour, we developed a novel maternal stress model using adult female C57Bl/6 mice. We observed that all HF+ mice demonstrated rapid weight gain, elevated fasting blood glucose levels and impaired glucose tolerance independent of the presence (+) or absence (-) of SIS. Behavioural testing output revealed anxiety-like behaviours remained similar across all groups prior to pregnancy. However, integrated anxiety z-scores revealed a mixed anxious profile amongst HF+/SIS+ females prior to pregnancy. HF+/SIS+ females also did not show reduced plasma ACTH and corticosterone levels that were observed in our other HF+ and HF- stress groups after SIS exposure. Further, HF+/SIS+ females demonstrated significant postpartum maternal neglect, resulting in fewer numbers of live offspring. These findings suggest that prolonged maternal HF diet consumption, coupled with previous exposure to SIS, places a significant burden on the maternal stress response system, resulting in reduced parental investment and negative postpartum behaviour towards offspring.
Collapse
Affiliation(s)
- Morgan C Bucknor
- School of Life and Environmental Sciences, Faculty of Science, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
| | - Anand Gururajan
- Centenary Institute of Cancer Medicine and Cell Biology, Sydney, NSW, Australia.
| | - Russell C Dale
- The Children's Hospital at Westmead, Kids Neuroscience Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; The Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| | - Markus J Hofer
- School of Life and Environmental Sciences, Faculty of Science, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
7
|
Yang Y, Rivera L, Fang S, Cavalier M, Suris A, Zhou Y, Huang Y. Maternal high-fat diet alters Tet-mediated epigenetic regulation during heart development. iScience 2024; 27:110631. [PMID: 39262804 PMCID: PMC11388159 DOI: 10.1016/j.isci.2024.110631] [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: 01/10/2024] [Revised: 05/24/2024] [Accepted: 07/29/2024] [Indexed: 09/13/2024] Open
Abstract
Imbalanced dietary intake, such as a high-fat diet (HFD) during pregnancy, has been associated with adverse offspring outcomes. Metabolic stress from imbalanced food intake alters the function of epigenetic regulators, resulting in abnormal transcriptional outputs in embryos to cause congenital disorders. We report herein that maternal HFD exposure causes metabolic changes in pregnant mice and non-compaction cardiomyopathy (NCC) in E15.5 embryos, accompanied by decreased 5-hydroxymethylcytosine (5hmC) levels and altered chromatin accessibility in embryonic heart tissues. Remarkably, maternal vitamin C supplementation mitigates these detrimental effects, likely by restoring iron, a cofactor for Tet enzymes, in a reduced state. Using a genetic approach, we further demonstrated that the cardioprotective benefits of vitamin C under HFD conditions are attributable to enhanced Tet activity. Our results highlight an interaction between maternal diet, specifically HFD or vitamin C, and epigenetic modifications during early heart development, emphasizing the importance of balanced maternal nutrition for healthy embryonic development.
Collapse
Affiliation(s)
- Yuhan Yang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Logan Rivera
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Shaohai Fang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Maryn Cavalier
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Ashley Suris
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
- Department of Translational Medical Sciences, School of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Yun Huang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
- Department of Translational Medical Sciences, School of Medicine, Texas A&M University, Houston, TX 77030, USA
| |
Collapse
|
8
|
Zilkha N, Chuartzman SG, Fishman R, Ben-Dor S, Kimchi T. Maternal high-fat or low-protein diets promote autism-related behavior and altered social behavior within groups in offspring male mice. Sci Rep 2024; 14:19227. [PMID: 39164365 PMCID: PMC11336096 DOI: 10.1038/s41598-024-70062-w] [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: 04/19/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024] Open
Abstract
Maternal malnutrition has been associated with neurodevelopmental deficits and long-term implications on the offspring's health and behavior. Here, we investigated the effects of maternal low-protein diet (LPD) or obesity-inducing maternal high-fat diet (HFD) on dyadic social interactions, group organization and autism-related behaviors in mice. We found that maternal HFD induced an autism-related behavioral phenotype in the male offspring, including a robust decrease in sociability, increased aggression, cognitive rigidity and repetitive behaviors. Maternal LPD led to a milder yet significant effect on autism-related symptoms, with no effects on olfactory-mediated social behavior. Under naturalistic conditions in a group setting, this manifested in altered behavioral repertoires, increased magnitude in dominance relations, and reduced interactions with novel social stimuli in the HFD male offspring, but not in the LPD offspring. Finally, we found HFD-induced transcriptomic changes in the olfactory bulbs of the male offspring. Together, our findings show that maternal malnutrition induces long-lasting effects on aggression and autism-related behaviors in male offspring, and potential impairments in brain regions processing chemosensory signals.
Collapse
Affiliation(s)
- Noga Zilkha
- Department of Brain Sciences, Weizmann Institute of Science, 76100, Rehovot, Israel
| | | | - Ruth Fishman
- Department of Brain Sciences, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Shifra Ben-Dor
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Tali Kimchi
- Department of Brain Sciences, Weizmann Institute of Science, 76100, Rehovot, Israel.
| |
Collapse
|
9
|
Murru E, Carta G, Manca C, Verce M, Everard A, Serra V, Aroni S, Melis M, Banni S. Impact of prenatal THC exposure on lipid metabolism and microbiota composition in rat offspring. Heliyon 2024; 10:e35637. [PMID: 39170117 PMCID: PMC11336829 DOI: 10.1016/j.heliyon.2024.e35637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
Objective Recent studies have demonstrated that prenatal exposure to the psychoactive ingredient of cannabis that is tetrahydrocannabinol (THC) disrupts fatty acid (FA) signaling pathways in the developing brain, potentially linking to psychopathologic consequences. Our research aims to investigate whether changes in midbrain FA metabolism are linked to modifications in peripheral metabolism of FAs and shifts in microbiota composition. Methods In order to model prenatal exposure to THC (PTE) in rats, Sprague Dawley dams were systemically administered with THC (2 mg/kg, s.c.) or vehicle once daily from gestational day 5-20. To evaluate the metabolic impact of PTE in the offspring during preadolescence (postnatal day, PND, 25-28), we analyzed FA profiles and their bioactive metabolites in liver and midbrain tissues, and microbiota alterations. Results Our findings indicate that PTE leads to sex-specific metabolic changes. In both sexes, PTE resulted in increased liver de novo lipogenesis (DNL) and alterations in FA profiles, as well as changes in N-acylethanolamines (NAEs), ligands of peroxisome proliferator-activated receptor alpha (PPAR-α). In females only, PTE influenced gene expression of PPAR-α and fibroblast growth factor 21 (Fgf21). In male offspring only, PTE was associated with significantly reduced fasting glycaemia and with alterations in the levels of midbrain NAEs. Our analysis of the progeny gut microbiota revealed sex-dependent effects of PTE, notably an increased abundance of Ileibacterium in PTE-exposed male offspring, a change previously associated with the long-term effects of a maternal unbalanced diet. Conclusions Our data suggest that in male PTE offspring a reduced fasting glycaemia, resulting from increased liver DNL and the absence of a compensatory effect by Ppar-α and FGF21 on glycemic homeostasis, are associated to alterations in midbrain NAEs ligands of PPAR-α. These metabolic changes within the midbrain, along with Ileibacterium abundance, may partly elucidate the heightened susceptibility to psychopathologic conditions previously observed in male offspring following PTE.
Collapse
Affiliation(s)
- Elisabetta Murru
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Gianfranca Carta
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Claudia Manca
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Marko Verce
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute (WELRI), avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Amandine Everard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute (WELRI), avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Valeria Serra
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Sonia Aroni
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Sebastiano Banni
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| |
Collapse
|
10
|
Horner D, Jepsen JRM, Chawes B, Aagaard K, Rosenberg JB, Mohammadzadeh P, Sevelsted A, Følsgaard N, Vinding R, Fagerlund B, Pantelis C, Bilenberg N, Pedersen CET, Eliasen A, Chen Y, Prince N, Chu SH, Kelly RS, Lasky-Su J, Halldorsson TI, Strøm M, Strandberg-Larsen K, Olsen SF, Glenthøj BY, Bønnelykke K, Ebdrup BH, Stokholm J, Rasmussen MA. A Western Dietary Pattern during Pregnancy is Associated with Neurodevelopmental Disorders in Childhood and Adolescence. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.07.24303907. [PMID: 38496582 PMCID: PMC10942528 DOI: 10.1101/2024.03.07.24303907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Despite the high prevalence of neurodevelopmental disorders, there is a notable gap in clinical studies exploring the impact of maternal diet during pregnancy on child neurodevelopment. This observational clinical study examined the association between pregnancy dietary patterns and neurodevelopmental disorders, as well as their symptoms, in a prospective cohort of 10-year-old children (n=508). Data-driven dietary patterns were derived from self-reported food frequency questionnaires. A Western dietary pattern in pregnancy (per SD change) was significantly associated with attention-deficit / hyperactivity disorder (ADHD) (OR 1.66 [1.21 - 2.27], p=0.002) and autism diagnosis (OR 2.22 [1.33 - 3.74], p=0.002) and associated symptoms (p<0.001). Findings for ADHD were validated in three large (n=59725, n=656, n=348), independent mother-child cohorts. Objective blood metabolome modelling at 24 weeks gestation identified 15 causally mediating metabolites which significantly improved ADHD prediction in external validation. Temporal analyses across five blood metabolome timepoints in two independent mother-child cohorts revealed that the association of Western dietary pattern metabolite scores with neurodevelopmental outcomes was consistently significant in early to mid-pregnancy, independent of later child timepoints. These findings underscore the importance of early intervention and provide robust evidence for targeted prenatal dietary interventions to prevent neurodevelopmental disorders in children.
Collapse
|
11
|
Chen B, de Launoit E, Renier N, Schneeberger M. Maternal nutritional programming shapes the cerebral landscape. Trends Endocrinol Metab 2024; 35:367-370. [PMID: 37949733 DOI: 10.1016/j.tem.2023.10.008] [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: 09/16/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
The escalating prevalence of maternal obesity raises concerns about its influence on offspring health. Exposure to obesogenic environments during early development leads to persistent alterations in brain function contributing to neurological disorders. Nutritional programming emerges as a promising avenue to counteract the deleterious effects of maternal obesity on offspring neurodevelopment.
Collapse
Affiliation(s)
- Bandy Chen
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Elisa de Launoit
- Laboratoire de Plasticité Structurale Sorbonne Université, ICM Paris Brain Institute, INSERM U1127, CNRS UMR7225, 75013 Paris, France
| | - Nicolas Renier
- Laboratoire de Plasticité Structurale Sorbonne Université, ICM Paris Brain Institute, INSERM U1127, CNRS UMR7225, 75013 Paris, France
| | - Marc Schneeberger
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA; Wu Tsai Institute for Mind and Brain, Yale University, New Haven, CT, USA.
| |
Collapse
|
12
|
Elgazzaz M, Berdasco C, Garai J, Baddoo M, Lu S, Daoud H, Zabaleta J, Mauvais-Jarvis F, Lazartigues E. Maternal Western diet programs cardiometabolic dysfunction and hypothalamic inflammation via epigenetic mechanisms predominantly in the male offspring. Mol Metab 2024; 80:101864. [PMID: 38159883 PMCID: PMC10806294 DOI: 10.1016/j.molmet.2023.101864] [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: 10/05/2023] [Revised: 12/04/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024] Open
Abstract
OBJECTIVE Maternal exposure during pregnancy is a strong determinant of offspring health outcomes. Such exposure induces changes in the offspring epigenome resulting in gene expression and functional changes. In this study, we investigated the effect of maternal Western hypercaloric diet (HCD) programming during the perinatal period on neuronal plasticity and cardiometabolic health in adult offspring. METHODS C57BL/6J dams were fed HCD for 1 month prior to mating with regular diet (RD) sires and kept on the same diet throughout pregnancy and lactation. At weaning, offspring were maintained on either HCD or RD for 3 months resulting in 4 treatment groups that underwent cardiometabolic assessments. DNA and RNA were extracted from the hypothalamus to perform whole genome methylation, mRNA, and miRNA sequencing followed by bioinformatic analyses. RESULTS Maternal programming resulted in male-specific hypertension and hyperglycemia, with both males and females showing increased sympathetic tone to the vasculature. Surprisingly, programmed male offspring fed HCD in adulthood exhibited lower glucose levels, less insulin resistance, and leptin levels compared to non-programmed HCD-fed male mice. Hypothalamic genes involved in inflammation and type 2 diabetes were targeted by differentially expressed miRNA, while genes involved in glial and astrocytic differentiation were differentially methylated in programmed male offspring. These data were supported by our findings of astrogliosis, microgliosis and increased microglial activation in programmed males in the paraventricular nucleus (PVN). Programming induced a protective effect in male mice fed HCD in adulthood, resulting in lower protein levels of hypothalamic TGFβ2, NF-κB2, NF-κBp65, Ser-pIRS1, and GLP1R compared to non-programmed HCD-fed males. Although TGFβ2 was upregulated in male mice exposed to HCD pre- or post-natally, only blockade of the brain TGFβ receptor in RD-HCD mice improved glucose tolerance and a trend to weight loss. CONCLUSIONS Our study shows that maternal HCD programs neuronal plasticity in the offspring and results in male-specific hypertension and hyperglycemia associated with hypothalamic inflammation in mechanisms and pathways distinct from post-natal HCD exposure. Together, our data unmask a compensatory role of HCD programming, likely via priming of metabolic pathways to handle excess nutrients in a more efficient way.
Collapse
Affiliation(s)
- Mona Elgazzaz
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA; Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Clara Berdasco
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA
| | - Jone Garai
- Department of Interdisciplinary Oncology and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Melody Baddoo
- Department of Pathology and Laboratory Medicine/Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Shiping Lu
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Hisham Daoud
- School of Computer and Cyber Sciences, Augusta University, Augusta, GA 30901, USA
| | - Jovanny Zabaleta
- Department of Interdisciplinary Oncology and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Franck Mauvais-Jarvis
- Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA; Department of Medicine, Section of Endocrinology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Eric Lazartigues
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA; Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| |
Collapse
|
13
|
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
|
14
|
Filho AMC, Gomes NS, Lós DB, Leite IB, Tremblay MÈ, Macêdo DS. Microglia and Microbiome-Gut-Brain Axis. ADVANCES IN NEUROBIOLOGY 2024; 37:303-331. [PMID: 39207699 DOI: 10.1007/978-3-031-55529-9_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The mammalian gut contains a community of microorganisms called gut microbiome. The gut microbiome is integrated into mammalian physiology, contributing to metabolism, production of metabolites, and promoting immunomodulatory actions. Microglia, the brain's resident innate immune cells, play an essential role in homeostatic neurogenesis, synaptic remodeling, and glial maturation. Microglial dysfunction has been implicated in the pathogenesis of several neuropsychiatric disorders. Recent findings indicate that microglia are influenced by the gut microbiome and their derived metabolites throughout life. The pathways by which microbiota regulate microglia have only started to be understood, but this discovery has the potential to provide valuable insights into the pathogenesis of brain disorders associated with an altered microbiome. Here, we discuss the recent literature on the role of the gut microbiome in modulating microglia during development and adulthood and summarize the key findings on this bidirectional crosstalk in selected examples of neuropsychiatric and neurodegenerative disorders. We also highlight some current caveats and perspectives for the field.
Collapse
Affiliation(s)
- Adriano Maia Chaves Filho
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Nayana Soares Gomes
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Deniele Bezerra Lós
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Isabel Bessa Leite
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Department of Molecular Medicine, Université de Laval, Québec City, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montréal, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| | - Danielle S Macêdo
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, SP, Brazil.
| |
Collapse
|
15
|
Gauvrit T, Benderradji H, Pelletier A, Aboulouard S, Faivre E, Carvalho K, Deleau A, Vallez E, Launay A, Bogdanova A, Besegher M, Le Gras S, Tailleux A, Salzet M, Buée L, Delahaye F, Blum D, Vieau D. Multi-Omics Data Integration Reveals Sex-Dependent Hippocampal Programming by Maternal High-Fat Diet during Lactation in Adult Mouse Offspring. Nutrients 2023; 15:4691. [PMID: 37960344 PMCID: PMC10649590 DOI: 10.3390/nu15214691] [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: 10/05/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
Early-life exposure to high-fat diets (HF) can program metabolic and cognitive alterations in adult offspring. Although the hippocampus plays a crucial role in memory and metabolic homeostasis, few studies have reported the impact of maternal HF on this structure. We assessed the effects of maternal HF during lactation on physiological, metabolic, and cognitive parameters in young adult offspring mice. To identify early-programming mechanisms in the hippocampus, we developed a multi-omics strategy in male and female offspring. Maternal HF induced a transient increased body weight at weaning, and a mild glucose intolerance only in 3-month-old male mice with no change in plasma metabolic parameters in adult male and female offspring. Behavioral alterations revealed by a Barnes maze test were observed both in 6-month-old male and female mice. The multi-omics strategy unveiled sex-specific transcriptomic and proteomic modifications in the hippocampus of adult offspring. These studies that were confirmed by regulon analysis show that, although genes whose expression was modified by maternal HF were different between sexes, the main pathways affected were similar with mitochondria and synapses as main hippocampal targets of maternal HF. The effects of maternal HF reported here may help to better characterize sex-dependent molecular pathways involved in cognitive disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Thibaut Gauvrit
- UMR-S1172, Lille Neurosciences & Cognition, University of Lille, INSERM, CHU Lille, 59000 Lille, France; (T.G.); (H.B.); (E.F.); (K.C.); (A.D.); (A.L.); (A.B.); (L.B.); (D.B.)
- Alzheimer & Tauopathies, LabEX DISTALZ, 59045 Lille, France
| | - Hamza Benderradji
- UMR-S1172, Lille Neurosciences & Cognition, University of Lille, INSERM, CHU Lille, 59000 Lille, France; (T.G.); (H.B.); (E.F.); (K.C.); (A.D.); (A.L.); (A.B.); (L.B.); (D.B.)
- Alzheimer & Tauopathies, LabEX DISTALZ, 59045 Lille, France
| | - Alexandre Pelletier
- The Department of Pharmacology & Biophysics, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA;
| | - Soulaimane Aboulouard
- U1192—Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), University of Lille, INSERM, 59000 Lille, France; (S.A.); (M.S.)
| | - Emilie Faivre
- UMR-S1172, Lille Neurosciences & Cognition, University of Lille, INSERM, CHU Lille, 59000 Lille, France; (T.G.); (H.B.); (E.F.); (K.C.); (A.D.); (A.L.); (A.B.); (L.B.); (D.B.)
- Alzheimer & Tauopathies, LabEX DISTALZ, 59045 Lille, France
| | - Kévin Carvalho
- UMR-S1172, Lille Neurosciences & Cognition, University of Lille, INSERM, CHU Lille, 59000 Lille, France; (T.G.); (H.B.); (E.F.); (K.C.); (A.D.); (A.L.); (A.B.); (L.B.); (D.B.)
- Alzheimer & Tauopathies, LabEX DISTALZ, 59045 Lille, France
| | - Aude Deleau
- UMR-S1172, Lille Neurosciences & Cognition, University of Lille, INSERM, CHU Lille, 59000 Lille, France; (T.G.); (H.B.); (E.F.); (K.C.); (A.D.); (A.L.); (A.B.); (L.B.); (D.B.)
- Alzheimer & Tauopathies, LabEX DISTALZ, 59045 Lille, France
| | - Emmanuelle Vallez
- Institut Pasteur de Lille, U1011-EGID, University of Lille, INSERM, CHU Lille, 59000 Lille, France; (E.V.); (A.T.)
| | - Agathe Launay
- UMR-S1172, Lille Neurosciences & Cognition, University of Lille, INSERM, CHU Lille, 59000 Lille, France; (T.G.); (H.B.); (E.F.); (K.C.); (A.D.); (A.L.); (A.B.); (L.B.); (D.B.)
- Alzheimer & Tauopathies, LabEX DISTALZ, 59045 Lille, France
| | - Anna Bogdanova
- UMR-S1172, Lille Neurosciences & Cognition, University of Lille, INSERM, CHU Lille, 59000 Lille, France; (T.G.); (H.B.); (E.F.); (K.C.); (A.D.); (A.L.); (A.B.); (L.B.); (D.B.)
- Alzheimer & Tauopathies, LabEX DISTALZ, 59045 Lille, France
| | - Mélanie Besegher
- US 41-UMS 2014-PLBS, Animal Facility, University of Lille, CNRS, INSERM, CHU Lille, 59000 Lille, France;
| | - Stéphanie Le Gras
- CNRS U7104, INSERM U1258, GenomEast Platform, IGBMC, University of Strasbourg, 67412 Illkirch, France;
| | - Anne Tailleux
- Institut Pasteur de Lille, U1011-EGID, University of Lille, INSERM, CHU Lille, 59000 Lille, France; (E.V.); (A.T.)
| | - Michel Salzet
- U1192—Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), University of Lille, INSERM, 59000 Lille, France; (S.A.); (M.S.)
| | - Luc Buée
- UMR-S1172, Lille Neurosciences & Cognition, University of Lille, INSERM, CHU Lille, 59000 Lille, France; (T.G.); (H.B.); (E.F.); (K.C.); (A.D.); (A.L.); (A.B.); (L.B.); (D.B.)
- Alzheimer & Tauopathies, LabEX DISTALZ, 59045 Lille, France
| | - Fabien Delahaye
- Sanofi Precision Medicine and Computational Biology, 94081 Vitry-sur-Seine, France;
| | - David Blum
- UMR-S1172, Lille Neurosciences & Cognition, University of Lille, INSERM, CHU Lille, 59000 Lille, France; (T.G.); (H.B.); (E.F.); (K.C.); (A.D.); (A.L.); (A.B.); (L.B.); (D.B.)
- Alzheimer & Tauopathies, LabEX DISTALZ, 59045 Lille, France
| | - Didier Vieau
- UMR-S1172, Lille Neurosciences & Cognition, University of Lille, INSERM, CHU Lille, 59000 Lille, France; (T.G.); (H.B.); (E.F.); (K.C.); (A.D.); (A.L.); (A.B.); (L.B.); (D.B.)
- Alzheimer & Tauopathies, LabEX DISTALZ, 59045 Lille, France
| |
Collapse
|
16
|
Bobotis BC, Braniff O, Gargus M, Akinluyi ET, Awogbindin IO, Tremblay MÈ. Sex differences of microglia in the healthy brain from embryonic development to adulthood and across lifestyle influences. Brain Res Bull 2023; 202:110752. [PMID: 37652267 DOI: 10.1016/j.brainresbull.2023.110752] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/15/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
Microglia, the central nervous system innate immune cells, play a critical role in maintaining a homeostatic environment in the brain throughout life. These cells exhibit an impressive range of functions and characteristics that help to ensure proper functioning of the brain. Notably, microglia can present differences in their genetic and physical traits, which can be influenced by a range of factors, including age, environmental exposures, disease, and sex. Remarkably, microglia have been found to express receptors for sex hormones, suggesting that these hormones may play a role in modulating microglial behavior and potentially contribute to sex differences. Additionally, sex-chromosomal factors were shown to impact microglial genetics and functioning. In this review, we will examine how microglial responses in homeostasis are impacted by their interaction with sex hormones and sex chromosomes. Specifically, our investigation will focus on examining this interaction from embryonic development to adulthood, and the influence of lifestyle elements on various microglial features, including density and distribution, morphology, transcriptome, and proteome.
Collapse
Affiliation(s)
| | - Olivia Braniff
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Makenna Gargus
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Elizabeth Toyin Akinluyi
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Pharmacology and Therapeutics, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Ifeoluwa Oluleke Awogbindin
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Neuroimmunology Group, Molecular Drug Metabolism and Toxicology Laboratory, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Neurosciences Axis, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada; Department of Molecular Medicine, Université Laval, Québec, QC, Canada; Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada.
| |
Collapse
|
17
|
Qi C, Wang L, Lu Y, Jin C. Editorial: Molecules, environments, and neurological disorders. Front Endocrinol (Lausanne) 2023; 14:1244800. [PMID: 37701895 PMCID: PMC10494539 DOI: 10.3389/fendo.2023.1244800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/07/2023] [Indexed: 09/14/2023] Open
Affiliation(s)
- Cai Qi
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
| | - Lu Wang
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Yong Lu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunyu Jin
- Division of Endocrinology and Metabolism, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
18
|
Douglas A, Stevens B, Lynch L. Interleukin-17 as a key player in neuroimmunometabolism. Nat Metab 2023; 5:1088-1100. [PMID: 37488456 PMCID: PMC10440016 DOI: 10.1038/s42255-023-00846-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/14/2023] [Indexed: 07/26/2023]
Abstract
In mammals, interleukin (IL)-17 cytokines are produced by innate and adaptive lymphocytes. However, the IL-17 family has widespread expression throughout evolution, dating as far back as cnidaria, molluscs and worms, which predate lymphocytes. The evolutionary conservation of IL-17 suggests that it is involved in innate defence strategies, but also that this cytokine family has a fundamental role beyond typical host defence. Throughout evolution, IL-17 seems to have a major function in homeostatic maintenance at barrier sites. Most recently, a pivotal role has been identified for IL-17 in regulating cellular metabolism, neuroimmunology and tissue physiology, particularly in adipose tissue. Here we review the emerging role of IL-17 signalling in regulating metabolic processes, which may shine a light on the evolutionary role of IL-17 beyond typical immune responses. We propose that IL-17 helps to coordinate the cross-talk among the nervous, endocrine and immune systems for whole-body energy homeostasis as a key player in neuroimmunometabolism.
Collapse
Affiliation(s)
- Aaron Douglas
- School of Biochemistry and Immunology, TBSI, Trinity College Dublin, Dublin, Ireland
| | - Brenneth Stevens
- School of Biochemistry and Immunology, TBSI, Trinity College Dublin, Dublin, Ireland
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lydia Lynch
- School of Biochemistry and Immunology, TBSI, Trinity College Dublin, Dublin, Ireland.
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Fan P, Wang Y, Lu K, Hong Y, Xu M, Han X, Liu Y. Modeling maternal cholesterol exposure reveals a reduction of neural progenitor proliferation using human cerebral organoids. LIFE MEDICINE 2023; 2:lnac034. [PMID: 39872117 PMCID: PMC11749704 DOI: 10.1093/lifemedi/lnac034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/22/2022] [Indexed: 01/29/2025]
Abstract
Maternal obesity raises the risk of high-cholesterol exposure for their offspring. Studies in cohorts and animal models report that maternal obesity could increase the risk of neurodevelopmental disorders in offspring including intellectual disabilities and autism spectrum disorders (ASDs). However, whether exposure to high cholesterol is responsible for brain developmental defects, as well as its underlying mechanism, is still unclear. Here, we constructed a cholesterol exposure model utilizing human pluripotent stem cell (hPSC)-derived cerebral organoids by exogenously adding cholesterol into the culture system. We observed enlargement of endosomes, decreased neural progenitor proliferation, and premature neural differentiation in brain organoids with the treatment of cholesterol. Moreover, in comparison with published transcriptome data, we found that our single-cell sequencing results showed a high correlation with ASD, indicating that high cholesterol during maternal might mediate the increased risk of ASD in the offspring. Our results reveal a reduction of neural progenitor proliferation in a cholesterol exposure model, which might be a promising indicator for prenatal diagnosis and offer a dynamic human model for maternal environment exposure.
Collapse
Affiliation(s)
- Pan Fan
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yuanhao Wang
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Kaiqin Lu
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yuan Hong
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Min Xu
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiao Han
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yan Liu
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
20
|
Barreto Meichtry L, Silva da Silva G, Londero L, Munir Mustafa Dahleh M, Cardoso Bortolotto V, Machado Araujo S, Aparecida Musachio E, Trivisiol da Silva D, Emanuelli T, Ricardo Sigal Carriço M, Roehrs R, Petri Guerra G, Prigol M. Exposure to trans fat during the developmental period ofDrosophila melanogasteralters the composition of fatty acids in the head and induces depression-like behavior. Neuroscience 2023; 519:10-22. [PMID: 36933760 DOI: 10.1016/j.neuroscience.2023.03.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Given the importance of understanding the disorders caused by trans fatty acids (TFAs), this study sought to add different concentrations hydrogenated vegetable fat (HVF) to the diet of Drosophila melanogaster during the developmental period and evaluate the effects on neurobehavioral parameters. Longevity, hatching rate, and behavioral functions were assessed, such as negative geotaxis, forced swimming, light/dark, mating, and aggressiveness. The fatty acids (FAs) present in the heads of the flies were quantified as well as serotonin (5HT) and dopamine (DA) levels. Our findings showed that flies that received HVF at all concentrations during development showed reduced longevity and hatching rates, in addition to increased depression-like, anxious-like, anhedonia-like, and aggressive behaviors. As for the biochemical parameters, there was a more significant presence of TFA in flies exposed to HVF at all concentrations evaluated and lower 5HT and DA levels. This study shows that HVF during the developmental phase can cause neurological changes and consequently induce behavioral disorders, thereby highlighting the importance of the type of FA offered in the early stages of life.
Collapse
Affiliation(s)
- Luana Barreto Meichtry
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio Pampa - Universidade Federal do Pampa - Campus Itaqui - Rua Luiz Joaquim de Sá Britto, Promorar, Itaqui, Rio Grande do Sul, Brazil, CEP 97650-000
| | - Guilherme Silva da Silva
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio Pampa - Universidade Federal do Pampa - Campus Itaqui - Rua Luiz Joaquim de Sá Britto, Promorar, Itaqui, Rio Grande do Sul, Brazil, CEP 97650-000
| | - Larissa Londero
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio Pampa - Universidade Federal do Pampa - Campus Itaqui - Rua Luiz Joaquim de Sá Britto, Promorar, Itaqui, Rio Grande do Sul, Brazil, CEP 97650-000
| | - Mustafa Munir Mustafa Dahleh
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio Pampa - Universidade Federal do Pampa - Campus Itaqui - Rua Luiz Joaquim de Sá Britto, Promorar, Itaqui, Rio Grande do Sul, Brazil, CEP 97650-000
| | - Vandreza Cardoso Bortolotto
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio Pampa - Universidade Federal do Pampa - Campus Itaqui - Rua Luiz Joaquim de Sá Britto, Promorar, Itaqui, Rio Grande do Sul, Brazil, CEP 97650-000
| | - Stífani Machado Araujo
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio Pampa - Universidade Federal do Pampa - Campus Itaqui - Rua Luiz Joaquim de Sá Britto, Promorar, Itaqui, Rio Grande do Sul, Brazil, CEP 97650-000
| | - Elize Aparecida Musachio
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio Pampa - Universidade Federal do Pampa - Campus Itaqui - Rua Luiz Joaquim de Sá Britto, Promorar, Itaqui, Rio Grande do Sul, Brazil, CEP 97650-000
| | - Dariane Trivisiol da Silva
- Departamento de Tecnologia e Ciência dos Alimentos, Centro de Ciências Rurais Universidade Federal de Santa Maria, Santa Maria, RS, Brazil, CEP 97105-900
| | - Tatiana Emanuelli
- Departamento de Tecnologia e Ciência dos Alimentos, Centro de Ciências Rurais Universidade Federal de Santa Maria, Santa Maria, RS, Brazil, CEP 97105-900
| | - Murilo Ricardo Sigal Carriço
- Universidade Federal do Pampa - Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), BR-472 Km 7, Uruguaiana, Brazil, CEP 97501-970
| | - Rafael Roehrs
- Universidade Federal do Pampa - Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), BR-472 Km 7, Uruguaiana, Brazil, CEP 97501-970
| | - Gustavo Petri Guerra
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio Pampa - Universidade Federal do Pampa - Campus Itaqui - Rua Luiz Joaquim de Sá Britto, Promorar, Itaqui, Rio Grande do Sul, Brazil, CEP 97650-000
| | - Marina Prigol
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio Pampa - Universidade Federal do Pampa - Campus Itaqui - Rua Luiz Joaquim de Sá Britto, Promorar, Itaqui, Rio Grande do Sul, Brazil, CEP 97650-000.
| |
Collapse
|
21
|
Cruz-Carrillo G, Trujillo-Villarreal LA, Ángeles-Valdez D, Concha L, Garza-Villarreal EA, Camacho-Morales A. Prenatal Cafeteria Diet Primes Anxiety-like Behavior Associated to Defects in Volume and Diffusion in the Fimbria-fornix of Mice Offspring. Neuroscience 2023; 511:70-85. [PMID: 36592924 DOI: 10.1016/j.neuroscience.2022.12.021] [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: 09/03/2022] [Revised: 11/25/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022]
Abstract
Prenatal exposure to high-energy diets primes brain alterations that increase the risk of developing behavioral and cognitive failures. Alterations in the structure and connectivity of brain involved in learning and memory performance are found in adult obese murine models and in humans. However, the role of prenatal exposure to high-energy diets in the modulation of the brain's structure and function during cognitive decline remains unknown. We used female C57BL6 mice (n = 10) exposed to a high-energy diets (Cafeteria diet (CAF)) or Chow diet for 9 weeks (before, during and after pregnancy) to characterize their effect on brain structural organization and learning and memory performance in the offspring at two-month-old (n = 17). Memory and learning performance were evaluated using the Y-maze test including forced and spontaneous alternation, novel object recognition (NORT), open field and Barnes maze tests. We found no alterations in the short- or long-time spatial memory performance in male offspring prenatally exposed to CAF diet when compared to the control, but they increased time spent in the edges resembling anxiety-like behavior. By using deformation-based morphometry and diffusion tensor imaging analysis we found that male offspring exposed to CAF diet showed increased volume in primary somatosensory cortex and a reduced volume of fimbria-fornix, which correlate with alterations in its white matter integrity. Biological modeling revealed that prenatal exposure to CAF diet predicts low volume in the fimbria-fornix, which was associated with anxiety in the offspring. The findings suggest that prenatal exposure to high-energy diets prime brain structural alterations related to anxiety in the offspring.
Collapse
Affiliation(s)
- Gabriela Cruz-Carrillo
- Universidad Autónoma de Nuevo Leon, College of Medicine, Department of Biochemistry, Monterrey, NL, Mexico; Universidad Autónoma de Nuevo Leon, Center for Research and Development in Health Sciences, Neurometabolism Unit, San Nicolás de los Garza, NL, Mexico
| | - Luis Angel Trujillo-Villarreal
- Universidad Autónoma de Nuevo Leon, College of Medicine, Department of Biochemistry, Monterrey, NL, Mexico; Universidad Autónoma de Nuevo Leon, Center for Research and Development in Health Sciences, Neurometabolism Unit, San Nicolás de los Garza, NL, Mexico
| | - Diego Ángeles-Valdez
- Universidad Nacional Autónoma de México, Instituto de Neurobiología, Departamento de Neurobiología Conductual y Cognitiva, Campus UNAM-Juriquilla, 76230 Queretaro, Mexico
| | - Luis Concha
- Universidad Nacional Autónoma de México, Instituto de Neurobiología, Departamento de Neurobiología Conductual y Cognitiva, Campus UNAM-Juriquilla, 76230 Queretaro, Mexico
| | - Eduardo A Garza-Villarreal
- Universidad Nacional Autónoma de México, Instituto de Neurobiología, Departamento de Neurobiología Conductual y Cognitiva, Campus UNAM-Juriquilla, 76230 Queretaro, Mexico
| | - Alberto Camacho-Morales
- Universidad Autónoma de Nuevo Leon, College of Medicine, Department of Biochemistry, Monterrey, NL, Mexico; Universidad Autónoma de Nuevo Leon, Center for Research and Development in Health Sciences, Neurometabolism Unit, San Nicolás de los Garza, NL, Mexico.
| |
Collapse
|
22
|
Ibeh S, Bakkar NMZ, Ahmad F, Nwaiwu J, Barsa C, Mekhjian S, Reslan MA, Eid AH, Harati H, Nabha S, Mechref Y, El-Yazbi AF, Kobeissy F. High fat diet exacerbates long-term metabolic, neuropathological, and behavioral derangements in an experimental mouse model of traumatic brain injury. Life Sci 2023; 314:121316. [PMID: 36565814 DOI: 10.1016/j.lfs.2022.121316] [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: 10/20/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
AIMS Traumatic brain injury (TBI) constitutes a serious public health concern. Although TBI targets the brain, it can exert several systemic effects which can worsen the complications observed in TBI subjects. Currently, there is no FDA-approved therapy available for its treatment. Thus, there has been an increasing need to understand other factors that could modulate TBI outcomes. Among the factors involved are diet and lifestyle. High-fat diets (HFD), rich in saturated fat, have been associated with adverse effects on brain health. MAIN METHODS To study this phenomenon, an experimental mouse model of open head injury, induced by the controlled cortical impact was used along with high-fat feeding to evaluate the impact of HFD on brain injury outcomes. Mice were fed HFD for a period of two months where several neurological, behavioral, and molecular outcomes were assessed to investigate the impact on chronic consequences of the injury 30 days post-TBI. KEY FINDINGS Two months of HFD feeding, together with TBI, led to a notable metabolic, neurological, and behavioral impairment. HFD was associated with increased blood glucose and fat-to-lean ratio. Spatial learning and memory, as well as motor coordination, were all significantly impaired. Notably, HFD aggravated neuroinflammation, oxidative stress, and neurodegeneration. Also, cell proliferation post-TBI was repressed by HFD, which was accompanied by an increased lesion volume. SIGNIFICANCE Our research indicated that chronic HFD feeding can worsen functional outcomes, predispose to neurodegeneration, and decrease brain recovery post-TBI. This sheds light on the clinical impact of HFD on TBI pathophysiology and rehabilitation as well.
Collapse
Affiliation(s)
- Stanley Ibeh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nour-Mounira Z Bakkar
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Fatima Ahmad
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Neuroscience Research Center, Lebanese University, Beirut, Lebanon
| | - Judith Nwaiwu
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Deparment of Chemistry, Texas Tech University, Lubbock, TX, USA
| | - Chloe Barsa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Sarine Mekhjian
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mohammad Amine Reslan
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Hayat Harati
- Neuroscience Research Center, Lebanese University, Beirut, Lebanon
| | - Sanaa Nabha
- Neuroscience Research Center, Lebanese University, Beirut, Lebanon
| | - Yehia Mechref
- Deparment of Chemistry, Texas Tech University, Lubbock, TX, USA
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Deparment of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Faculty of Pharmacy, Alamein International University, Al-Alamein, Egypt.
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Morehouse School of Medicine, Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), 720 Westview Dr. SW, Atlanta, GA 30310, USA.
| |
Collapse
|
23
|
Thomson S, Drummond K, O'Hely M, Symeonides C, Chandran C, Mansell T, Saffery R, Sly P, Mueller J, Vuillermin P, Ponsonby AL. Increased maternal non-oxidative energy metabolism mediates association between prenatal di-(2-ethylhexyl) phthalate (DEHP) exposure and offspring autism spectrum disorder symptoms in early life: A birth cohort study. ENVIRONMENT INTERNATIONAL 2023; 171:107678. [PMID: 36516674 DOI: 10.1016/j.envint.2022.107678] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 11/08/2022] [Accepted: 12/05/2022] [Indexed: 06/17/2023]
Abstract
Prenatal phthalate exposure has previously been linked to the development of autism spectrum disorder (ASD). However, the underlying biological mechanisms remain unclear. We investigated whether maternal and child central carbon metabolism is involved as part of the Barwon Infant Study (BIS), a population-based birth cohort of 1,074 Australian children. We estimated phthalate daily intakes using third-trimester urinary phthalate metabolite concentrations and other relevant indices. The metabolome of maternal serum in the third trimester, cord serum at birth and child plasma at 1 year were measured by nuclear magnetic resonance. We used the Small Molecule Pathway Database and principal component analysis to construct composite metabolite scores reflecting metabolic pathways. ASD symptoms at 2 and 4 years were measured in 596 and 674 children by subscales of the Child Behavior Checklist and the Strengths and Difficulties Questionnaire, respectively. Multivariable linear regression analyses demonstrated (i) prospective associations between higher prenatal di-(2-ethylhexyl) phthalate (DEHP) levels and upregulation of maternal non-oxidative energy metabolism pathways, and (ii) prospective associations between upregulation of these pathways and increased offspring ASD symptoms at 2 and 4 years of age. Counterfactual mediation analyses indicated that part of the mechanism by which higher prenatal DEHP exposure influences the development of ASD symptoms in early childhood is through a maternal metabolic shift in pregnancy towards non-oxidative energy pathways, which are inefficient compared to oxidative metabolism. These results highlight the importance of the prenatal period and suggest that further investigation of maternal energy metabolism as a molecular mediator of the adverse impact of prenatal environmental exposures such as phthalates is warranted.
Collapse
Affiliation(s)
- Sarah Thomson
- Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC 3052, Australia
| | - Katherine Drummond
- Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC 3052, Australia
| | - Martin O'Hely
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, 299 Ryrie Street, Geelong, VIC 3220, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, The University of Melbourne, 50 Flemington Rd, Parkville, VIC 3052, Australia
| | - Christos Symeonides
- Murdoch Children's Research Institute, Royal Children's Hospital, The University of Melbourne, 50 Flemington Rd, Parkville, VIC 3052, Australia
| | - Chitra Chandran
- Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC 3052, Australia
| | - Toby Mansell
- Murdoch Children's Research Institute, Royal Children's Hospital, The University of Melbourne, 50 Flemington Rd, Parkville, VIC 3052, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, Royal Children's Hospital, The University of Melbourne, 50 Flemington Rd, Parkville, VIC 3052, Australia
| | - Peter Sly
- Murdoch Children's Research Institute, Royal Children's Hospital, The University of Melbourne, 50 Flemington Rd, Parkville, VIC 3052, Australia; Child Health Research Centre, The University of Queensland, 62 Graham St, South Brisbane, QLD 4101, Australia
| | - Jochen Mueller
- Queensland Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Peter Vuillermin
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, 299 Ryrie Street, Geelong, VIC 3220, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, The University of Melbourne, 50 Flemington Rd, Parkville, VIC 3052, Australia
| | - Anne-Louise Ponsonby
- Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC 3052, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, The University of Melbourne, 50 Flemington Rd, Parkville, VIC 3052, Australia.
| |
Collapse
|
24
|
Bucknor MC, Gururajan A, Dale RC, Hofer MJ. A comprehensive approach to modeling maternal immune activation in rodents. Front Neurosci 2022; 16:1071976. [PMID: 36590294 PMCID: PMC9800799 DOI: 10.3389/fnins.2022.1071976] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Prenatal brain development is a highly orchestrated process, making it a very vulnerable window to perturbations. Maternal stress and subsequent inflammation during pregnancy leads to a state referred to as, maternal immune activation (MIA). If persistent, MIA can pose as a significant risk factor for the manifestation of neurodevelopmental disorders (NDDs) such as autism spectrum disorder and schizophrenia. To further elucidate this association between MIA and NDD risk, rodent models have been used extensively across laboratories for many years. However, there are few uniform approaches for rodent MIA models which make not only comparisons between studies difficult, but some established approaches come with limitations that can affect experimental outcomes. Here, we provide researchers with a comprehensive review of common experimental variables and potential limitations that should be considered when designing an MIA study based in a rodent model. Experimental variables discussed include: innate immune stimulation using poly I:C and LPS, environmental gestational stress paradigms, rodent diet composition and sterilization, rodent strain, neonatal handling, and the inclusion of sex-specific MIA offspring analyses. We discuss how some aspects of these variables have potential to make a profound impact on MIA data interpretation and reproducibility.
Collapse
Affiliation(s)
- Morgan C. Bucknor
- School of Life and Environmental Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Anand Gururajan
- The Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Russell C. Dale
- The Children’s Hospital at Westmead, Kids Neuroscience Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- The Children’s Hospital at Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Markus J. Hofer
- School of Life and Environmental Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
25
|
Jiang Q, Li T, Chen W, Huo Y, Mou X, Zhao W. Microbial regulation of offspring diseases mediated by maternal-associated microbial metabolites. Front Microbiol 2022; 13:955297. [PMID: 36406399 PMCID: PMC9672376 DOI: 10.3389/fmicb.2022.955297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
The microbiota plays a crucial role in individuals’ early and long-term health. Previous studies indicated that the microbial regulation of health may start before birth. As the in utero environment is (nearly) sterile, the regulation is probably be originated from maternal microbiota and mediated by their metabolites transferred across the placenta. After the birth, various metabolites are continuously delivered to offspring through human milk feeding. Meanwhile, some components, for example, human milk oligosaccharides, in human milk can only be fermented by microbes, which brings beneficial effects on offspring health. Hence, we speculated that human milk-derived metabolites may also play roles in microbial regulation. However, reports between maternal-associated microbial metabolites and offspring diseases are still lacking and sparsely distributed in several fields. Also, the definition of the maternal-associated microbial metabolite is still unclear. Thus, it would be beneficial to comb through the current knowledge of these metabolites related to diseases for assisting our goals of early prediction, early diagnosis, early prevention, or early treatment through actions only on mothers. Therefore, this review aims to present studies showing how researchers came to the path of investigating these metabolites and then to present studies linking them to the development of offspring asthma, type 1 diabetes mellitus, food allergy, neonatal necrotizing enterocolitis, or autism spectrum disorder. Potential English articles were collected from PubMed by searching terms of disease(s), maternal, and a list of microbial metabolites. Articles published within 5 years were preferred.
Collapse
Affiliation(s)
- Qingru Jiang
- Center for Infection and Immunity Studies, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Tian Li
- Department of Gynecology and Obstetrics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Wei Chen
- Center for Infection and Immunity Studies, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Yingfang Huo
- Center for Infection and Immunity Studies, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Xiangyu Mou
- Center for Infection and Immunity Studies, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Xiangyu Mou,
| | - Wenjing Zhao
- Center for Infection and Immunity Studies, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
- *Correspondence: Wenjing Zhao,
| |
Collapse
|
26
|
Hessami K, Norooznezhad AH, Monteiro S, Barrozo ER, Abdolmaleki AS, Arian SE, Zargarzadeh N, Shekerdemian LS, Aagaard KM, Shamshirsaz AA. COVID-19 Pandemic and Infant Neurodevelopmental Impairment: A Systematic Review and Meta-analysis. JAMA Netw Open 2022; 5:e2238941. [PMID: 36306133 PMCID: PMC9617178 DOI: 10.1001/jamanetworkopen.2022.38941] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
IMPORTANCE Primary studies proposed that aberrant maternal antiviral immunity and/or giving birth in quarantine, such as during the ongoing COVID-19 pandemic, may be associated with the risk of neurodevelopmental impairment (NDI) in offspring. OBJECTIVES To evaluate the associations of birth and being raised during the COVID-19 pandemic with risk of NDI among infants and to assess the association of gestational exposure to SARS-CoV-2 with risk of NDI. DATA SOURCES PubMed, Web of Science, Scopus, Embase, and preprint servers were systematically searched from inception to March 25, 2022. STUDY SELECTION Studies evaluating the neurodevelopment of infants born during the SARS-CoV-2 pandemic were included in this systematic review and meta-analysis. Studies using Ages and Stages Questionnaires, Third Edition (ASQ-3), were used for quantitative meta-analysis. DATA EXTRACTION AND SYNTHESIS Following the Preferred Reporting Items for Systematic Reviews and Meta-analyses, a random-effects model meta-analysis was used to pool the proportion and odds ratios (ORs) of overall NDI, as well as each developmental domain on ASQ-3 with the corresponding 95% CI. MAIN OUTCOMES AND MEASURES The primary outcome was the risk of overall NDI among infants screened during the pandemic vs prepandemic. The secondary outcome was the comparison of NDI by ASQ-3 domain among infants born to women with known gestational exposure to SARS-CoV-2 vs no exposure. RESULTS A total of 8 studies were included, including 21 419 infants (11 438 screened in pandemic and 9981 in prepandemic period). NDI was present in 330 of 8992 infants (7%; 95% CI, 4%-10%) screened during the COVID-19 pandemic from January 2020 to January 2021. Among the pandemic cohort, the prevalence of NDI among infants with gestational exposure to SARS-CoV-2 was 77 of 691 (12%; 95% CI, 6%-18%). Compared with the prepandemic cohort (2015-2019), the pandemic cohort was more likely to have communication impairment (OR, 1.70; 95% CI, 1.37-2.11; P < .001), without significant differences in other ASQ-3 domains (eg, gross motor, fine motor, personal-social, and problem-solving). In contrast, maternal SARS-CoV-2 infection was not associated with significant differences in any neurodevelopment domain in offspring, except for increasing the odds of fine motor impairment (OR, 3.46; 95% CI, 1.43-8.38; P < .001). CONCLUSIONS AND RELEVANCE In this systematic review and meta-analysis examining the association between COVID-19 pandemic and the risk of NDI, findings suggest that overall neurodevelopment in the first year of life was not changed by either being born or raised during the SARS-CoV-2 pandemic or by gestational exposure to SARS-CoV-2. Interestingly, the first year of life during the COVID-19 pandemic, regardless of maternal infection, was significantly associated with the risk of communication delay among the offspring.
Collapse
Affiliation(s)
- Kamran Hessami
- Maternal Fetal Care Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Amir Hossein Norooznezhad
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sonia Monteiro
- Meyer Center for Developmental Pediatrics and Autism, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston
| | - Enrico R. Barrozo
- Division of Maternal Fetal Medicine, Texas Children’s Hospital, Baylor College of Medicine, Houston
| | | | - Sara E. Arian
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas
| | - Nikan Zargarzadeh
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Lara S. Shekerdemian
- Division of Critical Care, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston
| | - Kjersti M. Aagaard
- Division of Maternal Fetal Medicine, Texas Children’s Hospital, Baylor College of Medicine, Houston
| | - Alireza A. Shamshirsaz
- Maternal Fetal Care Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
27
|
Laufer BI, Hasegawa Y, Zhang Z, Hogrefe CE, Del Rosso LA, Haapanen L, Hwang H, Bauman MD, Van de Water J, Taha AY, Slupsky CM, Golub MS, Capitanio JP, VandeVoort CA, Walker CK, LaSalle JM. Multi-omic brain and behavioral correlates of cell-free fetal DNA methylation in macaque maternal obesity models. Nat Commun 2022; 13:5538. [PMID: 36130949 PMCID: PMC9492781 DOI: 10.1038/s41467-022-33162-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/06/2022] [Indexed: 11/28/2022] Open
Abstract
Maternal obesity during pregnancy is associated with neurodevelopmental disorder (NDD) risk. We utilized integrative multi-omics to examine maternal obesity effects on offspring neurodevelopment in rhesus macaques by comparison to lean controls and two interventions. Differentially methylated regions (DMRs) from longitudinal maternal blood-derived cell-free fetal DNA (cffDNA) significantly overlapped with DMRs from infant brain. The DMRs were enriched for neurodevelopmental functions, methylation-sensitive developmental transcription factor motifs, and human NDD DMRs identified from brain and placenta. Brain and cffDNA methylation levels from a large region overlapping mir-663 correlated with maternal obesity, metabolic and immune markers, and infant behavior. A DUX4 hippocampal co-methylation network correlated with maternal obesity, infant behavior, infant hippocampal lipidomic and metabolomic profiles, and maternal blood measurements of DUX4 cffDNA methylation, cytokines, and metabolites. We conclude that in this model, maternal obesity was associated with changes in the infant brain and behavior, and these differences were detectable in pregnancy through integrative analyses of cffDNA methylation with immune and metabolic factors.
Collapse
Affiliation(s)
- Benjamin I Laufer
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, 95616, USA
- UC Davis Genome Center, University of California, Davis, CA, 95616, USA
- MIND Institute, School of Medicine, University of California Davis, Sacramento, CA, 95817, USA
- Department of OMNI Bioinformatics, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Yu Hasegawa
- Department of Food Science and Technology, University of California Davis, Davis, CA, 95616, USA
| | - Zhichao Zhang
- Department of Food Science and Technology, University of California Davis, Davis, CA, 95616, USA
| | - Casey E Hogrefe
- California National Primate Research Center, University of California Davis, Davis, CA, 95616, USA
| | - Laura A Del Rosso
- California National Primate Research Center, University of California Davis, Davis, CA, 95616, USA
| | - Lori Haapanen
- MIND Institute, School of Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Hyeyeon Hwang
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, 95616, USA
- UC Davis Genome Center, University of California, Davis, CA, 95616, USA
- MIND Institute, School of Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Melissa D Bauman
- MIND Institute, School of Medicine, University of California Davis, Sacramento, CA, 95817, USA
- California National Primate Research Center, University of California Davis, Davis, CA, 95616, USA
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Davis, CA, 95616, USA
- Perinatal Origins of Disparities Center, University of California Davis, Davis, CA, 95616, USA
| | - Judy Van de Water
- Perinatal Origins of Disparities Center, University of California Davis, Davis, CA, 95616, USA
- Department of Internal Medicine, University of California Davis, Davis, CA, 95616, USA
| | - Ameer Y Taha
- Department of Food Science and Technology, University of California Davis, Davis, CA, 95616, USA
| | - Carolyn M Slupsky
- Department of Food Science and Technology, University of California Davis, Davis, CA, 95616, USA
- Perinatal Origins of Disparities Center, University of California Davis, Davis, CA, 95616, USA
- Department of Nutrition, University of California Davis, Davis, CA, 95616, USA
| | - Mari S Golub
- California National Primate Research Center, University of California Davis, Davis, CA, 95616, USA
| | - John P Capitanio
- California National Primate Research Center, University of California Davis, Davis, CA, 95616, USA
- Department of Psychology, University of California Davis, Davis, CA, 95616, USA
| | - Catherine A VandeVoort
- California National Primate Research Center, University of California Davis, Davis, CA, 95616, USA
- Department of Obstetrics and Gynecology, School of Medicine, University of California Davis, Davis, CA, 95616, USA
| | - Cheryl K Walker
- MIND Institute, School of Medicine, University of California Davis, Sacramento, CA, 95817, USA
- California National Primate Research Center, University of California Davis, Davis, CA, 95616, USA
- Perinatal Origins of Disparities Center, University of California Davis, Davis, CA, 95616, USA
- Department of Obstetrics and Gynecology, School of Medicine, University of California Davis, Davis, CA, 95616, USA
| | - Janine M LaSalle
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, 95616, USA.
- UC Davis Genome Center, University of California, Davis, CA, 95616, USA.
- MIND Institute, School of Medicine, University of California Davis, Sacramento, CA, 95817, USA.
- Perinatal Origins of Disparities Center, University of California Davis, Davis, CA, 95616, USA.
| |
Collapse
|
28
|
Manuello J, Verdejo-Román J, Torres Espínola F, Escudero-Marín M, Catena A, Cauda F, Campoy C. Influence of Gestational Diabetes and Pregestational Maternal BMI on the Brain of Six-Year-Old Offspring. Pediatr Neurol 2022; 133:55-62. [PMID: 35759804 DOI: 10.1016/j.pediatrneurol.2022.05.005] [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: 09/24/2021] [Revised: 03/02/2022] [Accepted: 05/10/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Gestational diabetes (GD) and maternal excess weight are common pregnancy conditions that increase the risk of future complications for both the mother and her offspring. Their consequences on neurodevelopment are widely described in the literature, but less is known concerning the potential transgenerational influence on the brain structure. METHODS We used a combination of support vectors machine and hierarchical clustering to investigate the potential presence of anatomical brain differences in a sample of 109 children aged six years, born to mothers with overweight or obesity, or to mothers diagnosed with GD during pregnancy. RESULTS Significant effects are visible in the brain of children born to mothers with GD associated with pregestational excess weight, especially overweight instead of obesity. No differences in children's brain were observed when considering those born to normal-weight mothers. CONCLUSIONS Our study highlights the need for clinical attention of pregnant women at risk to develop GD, and especially those with pregestational excess weight, since this status was found to be associated with detectable transgenerational brain changes. These effects may be due to the absence of specific and individualized intervention in these mothers during pregnancy.
Collapse
Affiliation(s)
- Jordi Manuello
- Gcs-Fmri, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy; Focus Lab, Department of Psychology, University of Turin, Turin, Italy
| | - Juan Verdejo-Román
- Mind, Brain and Behavior Research Centre, University of Granada, Granada, Spain
| | - Francisco Torres Espínola
- Euristikos Excellence Centre For Pediatric Research, University of Granada, Granada, Spain; Department of Pediatrics, School of Medicine, University of Granada, Granada, Spain; DR. Federico Oloriz Neurosciences Institute, University of Granada, Granada, Spain
| | - Mireia Escudero-Marín
- Euristikos Excellence Centre For Pediatric Research, University of Granada, Granada, Spain; Department of Pediatrics, School of Medicine, University of Granada, Granada, Spain; DR. Federico Oloriz Neurosciences Institute, University of Granada, Granada, Spain
| | - Andrés Catena
- Mind, Brain and Behavior Research Centre, University of Granada, Granada, Spain
| | - Franco Cauda
- Gcs-Fmri, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy; Focus Lab, Department of Psychology, University of Turin, Turin, Italy
| | - Cristina Campoy
- Euristikos Excellence Centre For Pediatric Research, University of Granada, Granada, Spain; Department of Pediatrics, School of Medicine, University of Granada, Granada, Spain; DR. Federico Oloriz Neurosciences Institute, University of Granada, Granada, Spain; Spanish Network of Biomedical Research In Epidemiology and Public Health (Ciberesp), Granada's Node, Institute of Health Carlos III, Madrid, Spain; Biohealth Research Institute (IBS), Granada, Health Sciences Technological Park, Granada, Spain.
| |
Collapse
|
29
|
Urbonaite G, Knyzeliene A, Bunn FS, Smalskys A, Neniskyte U. The impact of maternal high-fat diet on offspring neurodevelopment. Front Neurosci 2022; 16:909762. [PMID: 35937892 PMCID: PMC9354026 DOI: 10.3389/fnins.2022.909762] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/27/2022] [Indexed: 12/11/2022] Open
Abstract
A maternal high-fat diet affects offspring neurodevelopment with long-term consequences on their brain health and behavior. During the past three decades, obesity has rapidly increased in the whole human population worldwide, including women of reproductive age. It is known that maternal obesity caused by a high-fat diet may lead to neurodevelopmental disorders in their offspring, such as autism spectrum disorder, attention deficit hyperactivity disorder, anxiety, depression, and schizophrenia. A maternal high-fat diet can affect offspring neurodevelopment due to inflammatory activation of the maternal gut, adipose tissue, and placenta, mirrored by increased levels of pro-inflammatory cytokines in both maternal and fetal circulation. Furthermore, a maternal high fat diet causes gut microbial dysbiosis further contributing to increased inflammatory milieu during pregnancy and lactation, thus disturbing both prenatal and postnatal neurodevelopment of the offspring. In addition, global molecular and cellular changes in the offspring's brain may occur due to epigenetic modifications including the downregulation of brain-derived neurotrophic factor (BDNF) expression and the activation of the endocannabinoid system. These neurodevelopmental aberrations are reflected in behavioral deficits observed in animals, corresponding to behavioral phenotypes of certain neurodevelopmental disorders in humans. Here we reviewed recent findings from rodent models and from human studies to reveal potential mechanisms by which a maternal high-fat diet interferes with the neurodevelopment of the offspring.
Collapse
Affiliation(s)
- Gintare Urbonaite
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Agne Knyzeliene
- Centre for Cardiovascular Science, The Queen’s Medical Research Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - Fanny Sophia Bunn
- Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
| | - Adomas Smalskys
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Urte Neniskyte
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- VU LSC-EMBL Partnership for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
30
|
Kwon HK, Choi GB, Huh JR. Maternal inflammation and its ramifications on fetal neurodevelopment. Trends Immunol 2022; 43:230-244. [PMID: 35131181 PMCID: PMC9005201 DOI: 10.1016/j.it.2022.01.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/12/2022]
Abstract
Exposure to heightened inflammation in pregnancy caused by infections or other inflammatory insults has been associated with the onset of neurodevelopmental and psychiatric disorders in children. Rodent models have provided unique insights into how this maternal immune activation (MIA) disrupts brain development. Here, we discuss the key immune factors involved, highlight recent advances in determining the molecular and cellular pathways of MIA, and review how the maternal immune system affects fetal development. We also examine the roles of microbiomes in shaping maternal immune function and the development of autism-like phenotypes. A comprehensive understanding of the gut bacteria-immune-neuro interaction in MIA is essential for developing diagnostic and therapeutic measures for high-risk pregnant women and identifying targets for treating inflammation-induced neurodevelopmental disorders.
Collapse
Affiliation(s)
- Ho-Keun Kwon
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea; Pohang University of Science and Technology, Pohang, Korea.
| | - Gloria B Choi
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jun R Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
31
|
Mouse models of immune dysfunction: their neuroanatomical differences reflect their anxiety-behavioural phenotype. Mol Psychiatry 2022; 27:3047-3055. [PMID: 35422470 PMCID: PMC9205773 DOI: 10.1038/s41380-022-01535-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 02/18/2022] [Accepted: 03/17/2022] [Indexed: 11/08/2022]
Abstract
Extensive evidence supports the role of the immune system in modulating brain function and behaviour. However, past studies have revealed striking heterogeneity in behavioural phenotypes produced from immune system dysfunction. Using magnetic resonance imaging, we studied the neuroanatomical differences among 11 distinct genetically modified mouse lines (n = 371), each deficient in a different element of the immune system. We found a significant and heterogeneous effect of immune dysfunction on the brains of both male and female mice. However, by imaging the whole brain and using Bayesian hierarchical modelling, we were able to identify patterns within the heterogeneous phenotype. Certain structures-such as the corpus callosum, midbrain, and thalamus-were more likely to be affected by immune dysfunction. A notable brain-behaviour relationship was identified with neuroanatomy endophenotypes across mouse models clustering according to anxiety-like behaviour phenotypes reported in literature, such as altered volume in brains regions associated with promoting fear response (e.g., the lateral septum and cerebellum). Interestingly, genes with preferential spatial expression in the most commonly affected regions are also associated with multiple sclerosis and other immune-mediated diseases. In total, our data suggest that the immune system modulates anxiety behaviour through well-established brain networks.
Collapse
|
32
|
Gawlińska K, Gawliński D, Kowal-Wiśniewska E, Jarmuż-Szymczak M, Filip M. Alteration of the Early Development Environment by Maternal Diet and the Occurrence of Autistic-like Phenotypes in Rat Offspring. Int J Mol Sci 2021; 22:ijms22189662. [PMID: 34575826 PMCID: PMC8472469 DOI: 10.3390/ijms22189662] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/30/2021] [Accepted: 09/03/2021] [Indexed: 12/13/2022] Open
Abstract
Epidemiological and preclinical studies suggest that maternal obesity increases the risk of autism spectrum disorder (ASD) in offspring. Here, we assessed the effects of exposure to modified maternal diets limited to pregnancy and lactation on brain development and behavior in rat offspring of both sexes. Among the studied diets, a maternal high-fat diet (HFD) disturbed the expression of ASD-related genes (Cacna1d, Nlgn3, and Shank1) and proteins (SHANK1 and TAOK2) in the prefrontal cortex of male offspring during adolescence. In addition, a maternal high-fat diet induced epigenetic changes by increasing cortical global DNA methylation and the expression of miR-423 and miR-494. As well as the molecular changes, behavioral studies have shown male-specific disturbances in social interaction and an increase in repetitive behavior during adolescence. Most of the observed changes disappeared in adulthood. In conclusion, we demonstrated the contribution of a maternal HFD to the predisposition to an ASD-like phenotype in male adolescent offspring, while a protective effect occurred in females.
Collapse
Affiliation(s)
- Kinga Gawlińska
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343 Kraków, Poland; (D.G.); (M.F.)
- Correspondence:
| | - Dawid Gawliński
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343 Kraków, Poland; (D.G.); (M.F.)
| | - Ewelina Kowal-Wiśniewska
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland; (E.K.-W.); (M.J.-S.)
| | - Małgorzata Jarmuż-Szymczak
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland; (E.K.-W.); (M.J.-S.)
| | - Małgorzata Filip
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343 Kraków, Poland; (D.G.); (M.F.)
| |
Collapse
|