1
|
De Paula GC, Simões RF, Garcia-Serrano AM, Duarte JMN. High-fat and High-sucrose Diet-induced Hypothalamic Inflammation Shows Sex Specific Features in Mice. Neurochem Res 2024; 49:3356-3366. [PMID: 39302596 PMCID: PMC11502605 DOI: 10.1007/s11064-024-04243-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Hypothalamic inflammation underlies diet-induced obesity and diabetes in rodent models. While diet normalization largely allows for recovery from metabolic impairment, it remains unknown whether long-term hypothalamic inflammation induced by obesogenic diets is a reversible process. In this study, we aimed at determining sex specificity of hypothalamic neuroinflammation and gliosis in mice fed a fat- and sugar-rich diet, and their reversibility upon diet normalization. Mice were fed a 60%-fat diet complemented by a 20% sucrose drink (HFHSD) for 3 days or 24 weeks, followed by a third group that had their diet normalized for the last 8 weeks of the study (reverse diet group, RevD). We determined the expression of pro- and anti-inflammatory cytokines, and of the inflammatory cell markers IBA1, CD68, GFAP and EMR1 in the hypothalamus, and analyzed morphology of microglia (IBA-1+ cells) and astrocytes (GFAP+ cells) in the arcuate nucleus. After 3 days of HFHSD feeding, male mice showed over-expression of IL-13, IL-18, IFN-γ, CD68 and EMR1 and reduced expression of IL-10, while females showed increased IL-6 and IBA1 and reduced IL-13, compared to controls. After 24 weeks of HFHSD exposure, male mice showed a general depression in the expression of cytokines, with prominent reduction of TNF-α, IL-6 and IL-13, but increased TGF-β, while female mice showed over-expression of IFN-γ and IL-18. Furthermore, both female and male mice showed some degree of gliosis after HFHSD feeding for 24 weeks. In mice of both sexes, diet normalization after prolonged HFHSD feeding resulted in partial neuroinflammation recovery in the hypothalamus, but gliosis was only recovered in females. In sum, HFHSD-fed mice display sex-specific inflammatory processes in the hypothalamus that are not fully reversible after diet normalization.
Collapse
Affiliation(s)
- Gabriela C De Paula
- Diabetes and Brain Function Unit, Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.
- Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden.
- Institute for Research in Biomedicine, Bellinzona, Switzerland.
| | - Rui F Simões
- Diabetes and Brain Function Unit, Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Alba M Garcia-Serrano
- Diabetes and Brain Function Unit, Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - João M N Duarte
- Diabetes and Brain Function Unit, Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
2
|
Cendra-Duarte E, Canals J, Becerra-Tomás N, Jardí C, Martín-Luján F, Arija V. Maternal dietary patterns and offspring behavioral problems. Pediatr Res 2024:10.1038/s41390-024-03462-3. [PMID: 39266631 DOI: 10.1038/s41390-024-03462-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND Mental health problems often begin in early childhood and could predict psychiatric and behavioral outcomes. Prenatal factors such as maternal nutrition have an impact on neurodevelopment. This study aims to investigate the association between maternal dietary patterns and emotional and behavioral problems in 4-year-old children. METHODS Within a cohort of 205 mother-child pairs, three maternal dietary patterns were identified: 'Sweet and Superfluous', 'Fish and Vegetables' and 'Meat and Cereals'. Child behavior was evaluated by means of the Child Behavior Checklist 1.5-5 (CBCL 1.5-5), the Teacher's Report Form 1.5-5 (TRF 1.5-5), and the Behavior Rating Inventory of Executive Function - Preschool Version (BRIEF-P). Multivariable analysis determined associations between maternal dietary patterns and their children's behavior. RESULTS Maternal adherence to the 'Sweet and Superfluous' pattern was positively associated with externalizing and depressive problems in children. The 'Meat and Cereals' pattern was linked to a higher risk for attention, hyperactivity and depressive problems as somatic complaints. Conversely, the 'Fish and Vegetables' pattern was associated with a reduced risk of hyperactivity problems. All these associations were more pronounced in girls than in boys. CONCLUSIONS Maternal diet during pregnancy is associated with the emotional and behavioral development of children at 4 years of age. IMPACT Previous research on prenatal dietary patterns and children's behavior is inconclusive. In our study, children of mothers who had higher intakes of sugar and processed foods during pregnancy were more likely to have emotional and behavioral problems at age 4, especially girls. A high-quality diet characterized by fish and vegetable consumption during pregnancy was associated with reduced anxiety and hyperactivity problems in girls. Our findings highlight the importance of prenatal nutrition for child neurodevelopment.
Collapse
Affiliation(s)
- Esther Cendra-Duarte
- Universitat Rovira i Virgili, Nutrition and Mental Health (NUTRISAM) Research Group, Reus, Spain
- Institut Català de la Salut (ICS), Collaborative Group on Lifestyles, Nutrition, and Tobacco (CENIT), Institut d´Investigació en Atenció Primària IDIAP Jordi Gol, Reus, Spain
| | - Josefa Canals
- Universitat Rovira i Virgili, Nutrition and Mental Health (NUTRISAM) Research Group, Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Universitat Rovira i Virgili, Department of Psychology, Centre de Recerca en Avaluació i Mesura de la Conducta (CRAMC), Tarragona, Spain
| | - Nerea Becerra-Tomás
- Universitat Rovira i Virgili, Nutrition and Mental Health (NUTRISAM) Research Group, Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Cristina Jardí
- Universitat Rovira i Virgili, Nutrition and Mental Health (NUTRISAM) Research Group, Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Francisco Martín-Luján
- Institut Català de la Salut (ICS), Collaborative Group on Lifestyles, Nutrition, and Tobacco (CENIT), Institut d´Investigació en Atenció Primària IDIAP Jordi Gol, Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Institut Català de la Salut (ICS), Institut d'Investigació en Atenció Primària IDIAP Jordi Gol, Barcelona, Spain
| | - Victoria Arija
- Universitat Rovira i Virgili, Nutrition and Mental Health (NUTRISAM) Research Group, Reus, Spain.
- Institut Català de la Salut (ICS), Collaborative Group on Lifestyles, Nutrition, and Tobacco (CENIT), Institut d´Investigació en Atenció Primària IDIAP Jordi Gol, Reus, Spain.
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain.
- Institut Català de la Salut (ICS), Institut d'Investigació en Atenció Primària IDIAP Jordi Gol, Barcelona, Spain.
| |
Collapse
|
3
|
Valdearcos M, McGrath ER, Brown Mayfield SM, Folick A, Cheang RT, Li L, Bachor TP, Lippert RN, Xu AW, Koliwad SK. Microglia mediate the early-life programming of adult glucose control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601752. [PMID: 39005380 PMCID: PMC11244970 DOI: 10.1101/2024.07.02.601752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Mammalian glucose homeostasis is, in part, nutritionally programmed during early neonatal life, a critical window for the formation of synapses between hypothalamic glucoregulatory centers. Although microglia are known to prune synapses throughout the brain, their specific role in refining hypothalamic glucoregulatory circuits remains unknown. Here, we show that microglia in the mediobasal hypothalamus (MBH) of mice actively engage in synaptic pruning during early life. Microglial phagocytic activity is induced following birth, regresses upon weaning from maternal milk, and is exacerbated by feeding dams a high-fat diet while lactating. In particular, we show that microglia refine perineuronal nets (PNNs) within the neonatal MBH. Indeed, transiently depleting microglia before weaning (P6-16), but not afterward (P21-31), remarkably increased PNN abundance in the MBH. Furthermore, mice lacking microglia only from P6-16 had glucose intolerance due to impaired glucose-responsive pancreatic insulin secretion in adulthood, a phenotype not seen if microglial depletion occurred after weaning. Viral retrograde tracing revealed that this impairment is linked to a reduction in the number of neurons in specific hypothalamic glucoregulatory centers that synaptically connect to the pancreatic β-cell compartment. These findings show that microglia facilitate synaptic plasticity in the MBH during early life through a process that includes PNN refinement, to establish hypothalamic circuits that regulate adult glucose homeostasis.
Collapse
Affiliation(s)
- M Valdearcos
- Diabetes Center, University of California, San Francisco, CA, USA
- Equal contribution
| | - ER McGrath
- Diabetes Center, University of California, San Francisco, CA, USA
| | | | - A Folick
- Diabetes Center, University of California, San Francisco, CA, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA
| | - RT Cheang
- Diabetes Center, University of California, San Francisco, CA, USA
| | - L Li
- Diabetes Center, University of California, San Francisco, CA, USA
| | - TP Bachor
- Diabetes Center, University of California, San Francisco, CA, USA
| | - RN Lippert
- German Institute of Human Nutrition Potsdam Rehbrücke, Potsdam, Germany; German Center for Diabetes Research, Neuherberg, Germany; Max Planck Institute for Metabolism Research, Cologne, Germany
| | - AW Xu
- Diabetes Center, University of California, San Francisco, CA, USA
| | - SK Koliwad
- Diabetes Center, University of California, San Francisco, CA, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA
- Equal contribution
| |
Collapse
|
4
|
Heinecke F, Fornes D, Capobianco E, Flores Quiroga JP, Labiano M, Faletti AG, Jawerbaum A, White V. Intestinal alterations and mild glucose homeostasis impairments in the offspring born to overweight rats. Mol Cell Endocrinol 2024; 587:112201. [PMID: 38494045 DOI: 10.1016/j.mce.2024.112201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024]
Abstract
The gut plays a crucial role in metabolism by regulating the passage of nutrients, water and microbial-derived substances to the portal circulation. Additionally, it produces incretins, such as glucose-insulinotropic releasing peptide (GIP) and glucagon-like derived peptide 1 (GLP1, encoded by gcg gene) in response to nutrient uptake. We aimed to investigate whether offspring from overweight rats develop anomalies in the barrier function and incretin transcription. We observed pro-inflammatory related changes along with a reduction in Claudin-3 levels resulting in increased gut-permeability in fetuses and offspring from overweight rats. Importantly, we found decreased gip mRNA levels in both fetuses and offspring from overweight rats. Differently, gcg mRNA levels were upregulated in fetuses, downregulated in female offspring and unchanged in male offspring from overweight rats. When cultured with high glucose, intestinal explants showed an increase in gip and gcg mRNA levels in control offspring. In contrast, offspring from overweight rats did not exhibit any response in gip mRNA levels. Additionally, while females showed no response, male offspring from overweight rats did exhibit an upregulation in gcg mRNA levels. Furthermore, female and male offspring from overweight rats showed sex-dependent anomalies when orally challenged with a glucose overload, returning to baseline glucose levels after 120 min. These results open new research questions about the role of the adverse maternal metabolic condition in the programming of impairments in glucose homeostasis, enteroendocrine function and gut barrier function in the offspring from overweight mothers and highlight the importance of a perinatal maternal healthy metabolism.
Collapse
Affiliation(s)
- Florencia Heinecke
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Daiana Fornes
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Evangelina Capobianco
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Jeremias Pablo Flores Quiroga
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Marina Labiano
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Alicia G Faletti
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Alicia Jawerbaum
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Verónica White
- Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
5
|
Dearden L, Furigo IC, Pantaleão LC, Wong LWP, Fernandez-Twinn DS, de Almeida-Faria J, Kentistou KA, Carreira MV, Bidault G, Vidal-Puig A, Ong KK, Perry JRB, Donato J, Ozanne SE. Maternal obesity increases hypothalamic miR-505-5p expression in mouse offspring leading to altered fatty acid sensing and increased intake of high-fat food. PLoS Biol 2024; 22:e3002641. [PMID: 38833481 PMCID: PMC11149872 DOI: 10.1371/journal.pbio.3002641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 04/25/2024] [Indexed: 06/06/2024] Open
Abstract
In utero exposure to maternal obesity programs increased obesity risk. Animal models show that programmed offspring obesity is preceded by hyperphagia, but the mechanisms that mediate these changes are unknown. Using a mouse model of maternal obesity, we observed increased intake of a high-fat diet (HFD) in offspring of obese mothers that precedes the development of obesity. Through small RNA sequencing, we identified programmed overexpression of hypothalamic miR-505-5p that is established in the fetus, lasts to adulthood and is maintained in hypothalamic neural progenitor cells cultured in vitro. Metabolic hormones and long-chain fatty acids associated with obesity increase miR-505-5p expression in hypothalamic neurons in vitro. We demonstrate that targets of miR-505-5p are enriched in fatty acid metabolism pathways and overexpression of miR-505-5p decreased neuronal fatty acid metabolism in vitro. miR-505-5p targets are associated with increased BMI in human genetic studies. Intra-cerebroventricular injection of miR-505-5p in wild-type mice increased HFD intake, mimicking the phenotype observed in offspring exposed to maternal obesity. Conversely, maternal exercise intervention in an obese mouse pregnancy rescued the programmed increase of hypothalamic miR-505-5p in offspring of obese dams and reduced HFD intake to control offspring levels. This study identifies a novel mechanism by which maternal obesity programs obesity in offspring via increased intake of high-fat foods.
Collapse
Affiliation(s)
- Laura Dearden
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Isadora C. Furigo
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo, Brazil
- Centre for Health and Life Sciences, Coventry University, Coventry, United Kingdom
| | - Lucas C. Pantaleão
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - L W. P. Wong
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Denise S. Fernandez-Twinn
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Juliana de Almeida-Faria
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
- University of Campinas, Faculty of Medical Sciences, Department of Pharmacology, Campinas, Brazil
| | | | - Maria V. Carreira
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Guillaume Bidault
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Ken K. Ong
- MRC Epidemiology Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - John R. B. Perry
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Epidemiology Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Jose Donato
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo, Brazil
| | - Susan E. Ozanne
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, United Kingdom
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| |
Collapse
|
6
|
Kearns ML, Reynolds CM. Developmentally programmed obesity: Is there a role for anti-inflammatory nutritional strategies? Exp Physiol 2024; 109:633-646. [PMID: 38031876 PMCID: PMC11061634 DOI: 10.1113/ep091209] [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: 09/07/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023]
Abstract
Pregnancy represents a period of immense maternal physiological adaptation, with progressive increases in lipid storage potential and insulin resistance to support fetal/placental growth. This requires significant change in the adipose tissue. Women living with obesity/overweight are more susceptible to these changes causing complications such as gestational diabetes. This is particularly worrying as up to 60% of European women are living with overweight/obesity at the onset of pregnancy. Furthermore, less than 1% meet all nutrition guidelines. There is now evidence that these deep metabolic changes can result in a predisposition to metabolic disease in both the mother and child in later life. Health and nutrition status during this period therefore represents a window to future health. This period offers a valuable opportunity for intervention to prevent the negative consequences of poor in utero environments and increases the long-term quality of life for mother and offspring. This review will examine a range of in utero factors which determine adipose tissue development, the impact of these factors on later-life obesity and metabolic health and the therapeutic value of dietary anti-inflammatory nutritional interventions during pregnancy and early life. When it comes to early life nutrition, a 'one size fits all' approach is not always appropriate. Understanding the mechanisms of adipose tissue development in response to differing nutritional strategies may be important in the context of complicated or adverse in utero environments and represents a substantial step towards a more personalised nutritional approach for the prevention of obesity, metabolic syndrome and related non-communicable diseases in future generations.
Collapse
Affiliation(s)
- Michelle L. Kearns
- Conway Institute/School of Public Health Physiotherapy and Sports Science/Institute of Food and Health/Diabetes Complications Research CentreUniversity College DublinDublin 4Ireland
| | - Clare M. Reynolds
- Conway Institute/School of Public Health Physiotherapy and Sports Science/Institute of Food and Health/Diabetes Complications Research CentreUniversity College DublinDublin 4Ireland
| |
Collapse
|
7
|
Guma E, Chakravarty MM. Immune Alterations in the Intrauterine Environment Shape Offspring Brain Development in a Sex-Specific Manner. Biol Psychiatry 2024:S0006-3223(24)01260-5. [PMID: 38679357 PMCID: PMC11511788 DOI: 10.1016/j.biopsych.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Exposure to immune dysregulation in utero or in early life has been shown to increase risk for neuropsychiatric illness. The sources of inflammation can be varied, including acute exposures due to maternal infection or acute stress, or persistent exposures due to chronic stress, obesity, malnutrition, or autoimmune diseases. These exposures may cause subtle alteration in brain development, structure, and function that can become progressively magnified across the life span, potentially increasing the likelihood of developing a neuropsychiatric conditions. There is some evidence that males are more susceptible to early-life inflammatory challenges than females. In this review, we discuss the various sources of in utero or early-life immune alteration and the known effects on fetal development with a sex-specific lens. To do so, we leveraged neuroimaging, behavioral, cellular, and neurochemical findings. Gaining clarity about how the intrauterine environment affects offspring development is critically important for informing preventive and early intervention measures that may buffer against the effects of these early-life risk factors.
Collapse
Affiliation(s)
- Elisa Guma
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, Maryland; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.
| | - M Mallar Chakravarty
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
8
|
Huang Y, Wang A, Zhou W, Li B, Zhang L, Rudolf AM, Jin Z, Hambly C, Wang G, Speakman JR. Maternal dietary fat during lactation shapes single nucleus transcriptomic profile of postnatal offspring hypothalamus in a sexually dimorphic manner in mice. Nat Commun 2024; 15:2382. [PMID: 38493217 PMCID: PMC10944494 DOI: 10.1038/s41467-024-46589-x] [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: 09/28/2023] [Accepted: 03/01/2024] [Indexed: 03/18/2024] Open
Abstract
Maternal overnutrition during lactation predisposes offspring to develop metabolic diseases and exacerbates the relevant syndromes in males more than females in later life. The hypothalamus is a heterogenous brain region that regulates energy balance. Here we combined metabolic trait quantification of mother and offspring mice under low and high fat diet (HFD) feeding during lactation, with single nucleus transcriptomic profiling of their offspring hypothalamus at peak lacation to understand the cellular and molecular alterations in response to maternal dietary pertubation. We found significant expansion in neuronal subpopulations including histaminergic (Hdc), arginine vasopressin/retinoic acid receptor-related orphan receptor β (Avp/Rorb) and agouti-related peptide/neuropeptide Y (AgRP/Npy) in male offspring when their mothers were fed HFD, and increased Npy-astrocyte interactions in offspring responding to maternal overnutrition. Our study provides a comprehensive offspring hypothalamus map at the peak lactation and reveals how the cellular subpopulations respond to maternal dietary fat in a sex-specific manner during development.
Collapse
Affiliation(s)
- Yi Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- Broad Institute of MIT and Harvard, Metabolism Program, Cambridge, MA, 02142, USA
| | - Anyongqi Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Wenjiang Zhou
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Centre for Evolutionary Biology, Fudan University, Shanghai, 200438, China
| | - Baoguo Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Linshan Zhang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Centre for Evolutionary Biology, Fudan University, Shanghai, 200438, China
| | - Agata M Rudolf
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zengguang Jin
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Catherine Hambly
- School of Biological Sciences, University of Aberdeen, Aberdeen, AB24 3FX, UK
| | - Guanlin Wang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Centre for Evolutionary Biology, Fudan University, Shanghai, 200438, China.
| | - John R Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- School of Biological Sciences, University of Aberdeen, Aberdeen, AB24 3FX, UK.
- China Medical University, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
9
|
Skowronski AA, Leibel RL, LeDuc CA. Neurodevelopmental Programming of Adiposity: Contributions to Obesity Risk. Endocr Rev 2024; 45:253-280. [PMID: 37971140 PMCID: PMC10911958 DOI: 10.1210/endrev/bnad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 11/19/2023]
Abstract
This review analyzes the published evidence regarding maternal factors that influence the developmental programming of long-term adiposity in humans and animals via the central nervous system (CNS). We describe the physiological outcomes of perinatal underfeeding and overfeeding and explore potential mechanisms that may mediate the impact of such exposures on the development of feeding circuits within the CNS-including the influences of metabolic hormones and epigenetic changes. The perinatal environment, reflective of maternal nutritional status, contributes to the programming of offspring adiposity. The in utero and early postnatal periods represent critically sensitive developmental windows during which the hormonal and metabolic milieu affects the maturation of the hypothalamus. Maternal hyperglycemia is associated with increased transfer of glucose to the fetus driving fetal hyperinsulinemia. Elevated fetal insulin causes increased adiposity and consequently higher fetal circulating leptin concentration. Mechanistic studies in animal models indicate important roles of leptin and insulin in central and peripheral programming of adiposity, and suggest that optimal concentrations of these hormones are critical during early life. Additionally, the environmental milieu during development may be conveyed to progeny through epigenetic marks and these can potentially be vertically transmitted to subsequent generations. Thus, nutritional and metabolic/endocrine signals during perinatal development can have lifelong (and possibly multigenerational) impacts on offspring body weight regulation.
Collapse
Affiliation(s)
- Alicja A Skowronski
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rudolph L Leibel
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Charles A LeDuc
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
10
|
Arslan S, Yıldıran H, Seymen CM. The Effect of Maternal High-Fat Diet on Adipose Tissue Histology and Lipid Metabolism-Related Genes Expression in Offspring Rats. Nutrients 2024; 16:150. [PMID: 38201978 PMCID: PMC10780511 DOI: 10.3390/nu16010150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
The developing fetus is dependent on the maternal nutritional environment. This study was conducted to determine the effects of a maternal high-fat diet (HFD) applied during pregnancy and/or lactation on the expression levels of some lipid-related genes in rat models. Half of the pregnant rats (n: 6) were fed an HFD (energy from fat: 45%), while the other half (n: 6) were fed a control diet (CD) (energy from fat, 7.7%) during the pregnancy period. During lactation, dams in both groups were divided into two subgroups, with half fed the CD and the other half fed the HFD. Thus, four groups were obtained: CD-CD, CD-HFD, HFD-CD, and HFD-HFD. At the end of lactation, all mothers and half of the offspring were sacrificed. The remaining offspring were fed a CD for five weeks. The average birth weight of the CD group offspring was found to be lower than that of the HFD group (p < 0.05). The amount of adipose tissue was highest in CD-HFD (p < 0.05), while gene expression levels were similar between groups (p > 0.05), and the most degenerative histological changes were observed in the eight-week HFD-HFD (p < 0.05). This study suggests that maternal HFD during pregnancy and lactation may increase adiposity in offspring rats, especially during the weaning period.
Collapse
Affiliation(s)
- Sabriye Arslan
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara 06490, Turkey;
| | - Hilal Yıldıran
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara 06490, Turkey;
| | - Cemile Merve Seymen
- Department of Histology and Embryology, Faculty of Medicine, Gazi University, Ankara 06500, Turkey;
| |
Collapse
|
11
|
Xu Y, Yang D, Wang L, Król E, Mazidi M, Li L, Huang Y, Niu C, Liu X, Lam SM, Shui G, Douglas A, Speakman JR. Maternal High Fat Diet in Lactation Impacts Hypothalamic Neurogenesis and Neurotrophic Development, Leading to Later Life Susceptibility to Obesity in Male but Not Female Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2305472. [PMID: 37867217 PMCID: PMC10724448 DOI: 10.1002/advs.202305472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Indexed: 10/24/2023]
Abstract
Early life nutrition can reprogram development and exert long-term consequences on body weight regulation. In mice, maternal high-fat diet (HFD) during lactation predisposed male but not female offspring to diet-induced obesity when adult. Molecular and cellular changes in the hypothalamus at important time points are examined in the early postnatal life in relation to maternal diet and demonstrated sex-differential hypothalamic reprogramming. Maternal HFD in lactation decreased the neurotropic development of neurons formed at the embryo stage (e12.5) and impaired early postnatal neurogenesis in the hypothalamic regions of both males and females. Males show a larger increased ratio of Neuropeptide Y (NPY) to Pro-opiomelanocortin (POMC) neurons in early postnatal neurogenesis, in response to maternal HFD, setting an obese tone for male offspring. These data provide insights into the mechanisms by which hypothalamic reprograming by early life overnutrition contributes to the sex-dependent susceptibility to obesity in adult life in mice.
Collapse
Affiliation(s)
- Yanchao Xu
- Shenzhen key laboratory for metabolic healthCenter for Energy Metabolism and ReproductionShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Dengbao Yang
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Lu Wang
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
- Institute of Biological and Environmental SciencesUniversity of AberdeenAberdeenScotlandAB24 2TZUK
- University of Chinese Academy of SciencesShijingshanBeijing100049P. R. China
- School of PharmacyKey Laboratory of Molecular Pharmacology and Drug EvaluationMinistry of EducationYantai UniversityYantai264005P. R. China
| | - Elżbieta Król
- Institute of Biological and Environmental SciencesUniversity of AberdeenAberdeenScotlandAB24 2TZUK
| | - Mohsen Mazidi
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
- University of Chinese Academy of SciencesShijingshanBeijing100049P. R. China
| | - Li Li
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
- University of Chinese Academy of SciencesShijingshanBeijing100049P. R. China
| | - Yi Huang
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Chaoqun Niu
- Shenzhen key laboratory for metabolic healthCenter for Energy Metabolism and ReproductionShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Xue Liu
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Alex Douglas
- Institute of Biological and Environmental SciencesUniversity of AberdeenAberdeenScotlandAB24 2TZUK
| | - John R. Speakman
- Shenzhen key laboratory for metabolic healthCenter for Energy Metabolism and ReproductionShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
- Institute of Biological and Environmental SciencesUniversity of AberdeenAberdeenScotlandAB24 2TZUK
- China medical universityShenyang110000P. R. China
| |
Collapse
|
12
|
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
|
13
|
Martins MG, Silver Z, Ayoub K, Hyland L, Woodside B, Kiss ACI, Abizaid A. Maternal glucose intolerance during pregnancy affects offspring POMC expression and results in adult metabolic alterations in a sex-dependent manner. Front Endocrinol (Lausanne) 2023; 14:1189207. [PMID: 37396180 PMCID: PMC10311085 DOI: 10.3389/fendo.2023.1189207] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/23/2023] [Indexed: 07/04/2023] Open
Abstract
Introduction Gestational diabetes (GDM) is associated with negative outcomes in mothers and their offspring, including greater risks of macrosomia at birth and the development of metabolic disorders. While these outcomes are well-established, the mechanisms by which this increased metabolic vulnerability is conferred on the offspring are comparatively lacking. One proposed mechanism is that maternal glycemic dysregulation alters the development of the hypothalamic regions related to metabolism and energy balance. Methods To investigate this possibility, in this study, we first examined the effects of STZ-induced maternal glucose intolerance on the offspring on pregnancy day (PD) 19, and, in a second experiment, in early adulthood (postnatal day (PND) 60). Whether effects would be influenced by sex, or exposure of offspring to a high-fat diet was also investigated. The impact of maternal STZ treatment on POMC neuron number in the ARC of offspring at both time points was also examined. Results As expected, STZ administration on PD 7 decreased maternal glucose tolerance, and increased risk for macrosomia, and loss of pups at birth. Offspring of STZ-treated mothers were also more vulnerable to developing metabolic impairments in adulthood. These were accompanied by sex-specific effects of maternal STZ treatment in the offspring, including fewer POMC neurons in the ARC of female but not male infants in late pregnancy and a higher number of POMC neurons in the ARC of both male and female adult offspring of STZ-treated dams, which was exacerbated in females exposed to a high-fat diet after weaning. Discussion This work suggests that maternal hyperglycemia induced by STZ treatment, in combination with early-life exposure to an obesogenic diet, leads to adult metabolic alterations that correlate with the increased hypothalamic expression of POMC, showing that maternal glycemic dysregulation can impact the development of hypothalamic circuits regulating energy state with a stronger impact on female offspring.
Collapse
Affiliation(s)
- Marina Galleazzo Martins
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
- Department of Physiology, Institute of Biosciences of the University of São Paulo (IB/USP), São Paulo, São Paulo, Brazil
| | - Zachary Silver
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Kiara Ayoub
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Lindsay Hyland
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Barbara Woodside
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Ana Carolina Inhasz Kiss
- Department of Physiology, Institute of Biosciences of the University of São Paulo (IB/USP), São Paulo, São Paulo, Brazil
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil
| | - Alfonso Abizaid
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
14
|
Shrivastava K, Swaminathan T, Barlotta A, Athreya V, Choudhry H, Rossi MA. Maternal overnutrition is associated with altered synaptic input to lateral hypothalamic area. Mol Metab 2023; 71:101702. [PMID: 36898526 PMCID: PMC10025284 DOI: 10.1016/j.molmet.2023.101702] [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: 01/04/2023] [Revised: 02/18/2023] [Accepted: 03/02/2023] [Indexed: 03/12/2023] Open
Abstract
OBJECTIVE Maternal overnutrition is associated with adverse outcomes in offspring, including increased risk for obesity and diabetes. Here, we aim to test the effects of maternal obesity on lateral hypothalamic feeding circuit function and determine the relationship with body weight regulation. METHODS Using a mouse model of maternal obesity, we assessed how perinatal overnutrition affected food intake and body weight regulation in adult offspring. We then used channelrhodopsin-assisted circuit mapping and electrophysiological recordings to assess the synaptic connectivity within an extended amygdala-lateral hypothalamic pathway. RESULTS We show that maternal overnutrition during gestation and throughout lactation produces offspring that are heavier than controls prior to weaning. When weaned onto chow, the body weights of over-nourished offspring normalize to control levels. However, when presented with highly palatable food as adults, both male and female maternally over-nourished offspring are highly susceptible to diet-induced obesity. This is associated with altered synaptic strength in an extended amygdala-lateral hypothalamic pathway, which is predicted by developmental growth rate. Additionally, lateral hypothalamic neurons receiving synaptic input from the bed nucleus of the stria terminalis have enhanced excitatory input following maternal overnutrition which is predicted by early life growth rate. CONCLUSIONS Together, these results demonstrate one way in which maternal obesity rewires hypothalamic feeding circuits to predispose offspring to metabolic dysfunction.
Collapse
Affiliation(s)
| | | | | | | | | | - Mark A Rossi
- Child Health Institute of New Jersey, USA; Department of Psychiatry, Robert Wood Johnson Medical School, USA; Brain Health Institute, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
15
|
Wongkitikamjorn W, Wada E, Hosomichi J, Maeda H, Satrawaha S, Hong H, Yoshida KI, Ono T, Hayashi YK. Metabolic dysregulation and decreased capillarization in skeletal muscles of male adolescent offspring rats exposed to gestational intermittent hypoxia. Front Physiol 2023; 14:1067683. [PMID: 36711021 PMCID: PMC9878705 DOI: 10.3389/fphys.2023.1067683] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023] Open
Abstract
Gestational intermittent hypoxia (IH) is a hallmark of obstructive sleep apnea that occurs frequently during pregnancy, and effects caused by this environmental change during pregnancy may be transmitted to the offspring. In this study, we aimed to clarify the effects of IH in pregnant rats on the skeletal muscle of adolescent offspring rats. Mother rats underwent IH from gestation day 7-21, and their 5-weeks-old male offspring were analyzed. All male offspring rats were born and raised under normoxia conditions. Although no general growth retardation was observed, we found that exposure to gestational IH reduces endurance running capacity of adolescent offspring rats. Both a respiratory muscle (diaphragm; DIA) and a limb muscle (tibialis anterior; TA) showed no histological abnormalities, including fiber size and fiber type distribution. To identify the possible mechanism underlying the reduced running capacity, regulatory factors associated with energy metabolism were analyzed in different parts of skeletal muscles. Compared with rats born under conditions of gestational normoxia, gestational IH offspring rats showed significantly lower expression of genes associated with glucose and lipid metabolism, and lower protein levels of phosphorylated AMPK and AKT. Furthermore, gene expression of adiponectin receptors one and two was significantly decreased in the DIA and TA muscles. In addition, the DIA muscle from adolescent rats had significantly decreased capillary density as a result of gestational IH. However, these changes were not observed in a sucking muscle (geniohyoid) and a masticating muscle (masseter) of these rats. These results suggest that respiratory and limb muscles are vulnerable to gestational IH, which induces altered energy metabolism with decreased aerobic motor function. These changes were partially owing to the decreased expression of adiponectin receptors and decreased capillary density in adolescent offspring rats.
Collapse
Affiliation(s)
- Wirongrong Wongkitikamjorn
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan,Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Eiji Wada
- Department of Pathophysiology, Tokyo Medical University, Tokyo, Japan
| | - Jun Hosomichi
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hideyuki Maeda
- Department of Forensic Medicine, Tokyo Medical University, Tokyo, Japan
| | - Sirichom Satrawaha
- Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Haixin Hong
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan,Department of Stomatology, Shenzhen University General Hospital, Shenzhen, China
| | - Ken-ichi Yoshida
- Department of Forensic Medicine, Tokyo Medical University, Tokyo, Japan
| | - Takashi Ono
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yukiko K. Hayashi
- Department of Pathophysiology, Tokyo Medical University, Tokyo, Japan,*Correspondence: Yukiko K. Hayashi,
| |
Collapse
|
16
|
Anwer H, Mason D, Zajitschek S, Hesselson D, Noble DWA, Morris MJ, Lagisz M, Nakagawa S. Intergenerational effects of overfeeding on aversive learning in zebrafish ( Danio rerio). Ecol Evol 2022; 12:e9423. [PMID: 36311397 PMCID: PMC9596326 DOI: 10.1002/ece3.9423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
The obesity epidemic is concerning as obesity appears to negatively impact cognition and behavior. Furthermore, some studies suggest that this negative effect could be carried across generations from both mothers and fathers although evidence is not consistent. Here, we attempt to address how obesogenic diets in the parental generation (F0) can impact offspring's cognition and anxiety intergenerationally (F1) in a zebrafish model. We compare both mean trait values and their variances. Using a multifactorial design, we created a total of four groups: F1T (treatment mothers × treatment fathers); F1M (treatment mothers × control fathers); F1P (treatment fathers × control mothers); and F1C (control mothers × control fathers, F1C); and subjected them to anxiety tank tests and aversive learning assays. When both parents were exposed, offspring (F1T) displayed the poorest aversive learning, while offspring that only had one parent exposed (F1P and F1M) learnt the aversive learning task the best. Zebrafish in all groups displayed no statistically significant differences in anxiety-associated behaviors. Males and females also performed similarly in both anxiety and aversive learning assays. While all F1 groups had similar levels of fasting blood glucose, variance in glucose levels were reduced in F1P and F1T indicating the importance of investigating heteroskedasticity between groups. Furthermore, anxiety behaviors of these two groups appeared to be less repeatable. To our knowledge, this is the first study to test the intergenerational effects of an obesogenic diet on zebrafish cognition. Our multifactorial design as well as repeated tests also allowed us to disentangle maternal and paternal effects (as well as combined effects) and accurately detect subtle information such as between-individual variation.
Collapse
Affiliation(s)
- Hamza Anwer
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental SciencesUniversity of New South WalesSydneyNew South WalesAustralia
- Diabetes and Metabolism DivisionGarvan Institute of Medical ResearchDarlinghurst, SydneyNew South WalesAustralia
| | - Dominic Mason
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental SciencesUniversity of New South WalesSydneyNew South WalesAustralia
- Diabetes and Metabolism DivisionGarvan Institute of Medical ResearchDarlinghurst, SydneyNew South WalesAustralia
| | - Susanne Zajitschek
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental SciencesUniversity of New South WalesSydneyNew South WalesAustralia
- Diabetes and Metabolism DivisionGarvan Institute of Medical ResearchDarlinghurst, SydneyNew South WalesAustralia
- Liverpool John Moores UniversitySchool of Biological and Environmental SciencesLiverpoolUK
| | - Daniel Hesselson
- Diabetes and Metabolism DivisionGarvan Institute of Medical ResearchDarlinghurst, SydneyNew South WalesAustralia
- Centenary Institute and Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
| | - Daniel W. A. Noble
- Division of Ecology and Evolution, Research School of BiologyThe Australian National UniversityCanberraAustralian Capital TerritoryAustralia
| | - Margaret J. Morris
- Department of Pharmacology, School of Medical SciencesUniversity of New South WalesSydneyNew South WalesAustralia
| | - Malgorzata Lagisz
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental SciencesUniversity of New South WalesSydneyNew South WalesAustralia
- Diabetes and Metabolism DivisionGarvan Institute of Medical ResearchDarlinghurst, SydneyNew South WalesAustralia
| | - Shinichi Nakagawa
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental SciencesUniversity of New South WalesSydneyNew South WalesAustralia
- Diabetes and Metabolism DivisionGarvan Institute of Medical ResearchDarlinghurst, SydneyNew South WalesAustralia
| |
Collapse
|
17
|
Programming by maternal obesity: a pathway to poor cardiometabolic health in the offspring. Proc Nutr Soc 2022; 81:227-242. [DOI: 10.1017/s0029665122001914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There is an ever increasing prevalence of maternal obesity worldwide such that in many populations over half of women enter pregnancy either overweight or obese. This review aims to summarise the impact of maternal obesity on offspring cardiometabolic outcomes. Maternal obesity is associated with increased risk of adverse maternal and pregnancy outcomes. However, beyond this exposure to maternal obesity during development also increases the risk of her offspring developing long-term adverse cardiometabolic outcomes throughout their adult life. Both human studies and those in experimental animal models have shown that maternal obesity can programme increased risk of offspring developing obesity and adipose tissue dysfunction; type 2 diabetes with peripheral insulin resistance and β-cell dysfunction; CVD with impaired cardiac structure and function and hypertension via impaired vascular and kidney function. As female offspring themselves are therefore likely to enter pregnancy with poor cardiometabolic health this can lead to an inter-generational cycle perpetuating the transmission of poor cardiometabolic health across generations. Maternal exercise interventions have the potential to mitigate some of the adverse effects of maternal obesity on offspring health, although further studies into long-term outcomes and how these translate to a clinical context are still required.
Collapse
|
18
|
Sandovici I, Fernandez-Twinn DS, Hufnagel A, Constância M, Ozanne SE. Sex differences in the intergenerational inheritance of metabolic traits. Nat Metab 2022; 4:507-523. [PMID: 35637347 DOI: 10.1038/s42255-022-00570-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 04/05/2022] [Indexed: 02/02/2023]
Abstract
Strong evidence suggests that early-life exposures to suboptimal environmental factors, including those in utero, influence our long-term metabolic health. This has been termed developmental programming. Mounting evidence suggests that the growth and metabolism of male and female fetuses differ. Therefore, sexual dimorphism in response to pre-conception or early-life exposures could contribute to known sex differences in susceptibility to poor metabolic health in adulthood. However, until recently, many studies, especially those in animal models, focused on a single sex, or, often in the case of studies performed during intrauterine development, did not report the sex of the animal at all. In this review, we (a) summarize the evidence that male and females respond differently to a suboptimal pre-conceptional or in utero environment, (b) explore the potential biological mechanisms that underlie these differences and (c) review the consequences of these differences for long-term metabolic health, including that of subsequent generations.
Collapse
Affiliation(s)
- Ionel Sandovici
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Denise S Fernandez-Twinn
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Antonia Hufnagel
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Miguel Constância
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK.
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| | - Susan E Ozanne
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
19
|
Widen EM, Burns N, Daniels M, Backlund G, Rickman R, Foster S, Nichols AR, Hoepner LA, Kinsey EW, Ramirez-Carvey J, Hassoun A, Perera FP, Bukowski R, Rundle AG. Gestational weight change and childhood body composition trajectories from pregnancy to early adolescence. Obesity (Silver Spring) 2022; 30:707-717. [PMID: 35137558 PMCID: PMC8957403 DOI: 10.1002/oby.23367] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 12/03/2021] [Accepted: 12/10/2021] [Indexed: 11/09/2022]
Abstract
OBJECTIVE A mother-child dyad trajectory model of weight and body composition spanning from conception to adolescence was developed to understand how early life exposures shape childhood body composition. METHODS African American (49.3%) and Dominican (50.7%) pregnant mothers (n = 337) were enrolled during pregnancy, and their children (47.5% female) were followed from ages 5 to 14. Gestational weight gain (GWG) was abstracted from medical records. Child weight, height, percentage body fat, and waist circumference were measured. GWG and child body composition trajectories were jointly modeled with a flexible latent class model with a class membership component that included prepregnancy BMI. RESULTS Four prenatal and child body composition trajectory patterns were identified, and sex-specific patterns were observed for the joint GWG-postnatal body composition trajectories with more distinct patterns among girls but not boys. Girls of mothers with high GWG across gestation had the highest BMI z score, waist circumference, and percentage body fat trajectories from ages 5 to 14; however, boys in this high GWG group did not show similar growth patterns. CONCLUSIONS Jointly modeled prenatal weight and child body composition trajectories showed sex-specific patterns. Growth patterns from childhood though early adolescence appeared to be more profoundly affected by higher GWG patterns in females, suggesting sex differences in developmental programming.
Collapse
Affiliation(s)
- Elizabeth M Widen
- Department of Nutritional Sciences, School of Human Ecology, College of Natural Sciences, University of Texas at Austin, Austin, Texas, USA
- Department of Women's Health, Dell Medical School, University of Texas at Austin, Austin, Texas, USA
- Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin, Austin, Texas, USA
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University Medical Center, New York, New York, USA
| | - Natalie Burns
- Department of Statistics, University of Florida, Gainesville, Florida, USA
| | - Michael Daniels
- Department of Statistics, University of Florida, Gainesville, Florida, USA
| | - Grant Backlund
- Department of Statistics, University of Florida, Gainesville, Florida, USA
| | - Rachel Rickman
- Department of Nutritional Sciences, School of Human Ecology, College of Natural Sciences, University of Texas at Austin, Austin, Texas, USA
- Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin, Austin, Texas, USA
| | - Saralyn Foster
- Department of Nutritional Sciences, School of Human Ecology, College of Natural Sciences, University of Texas at Austin, Austin, Texas, USA
- Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin, Austin, Texas, USA
| | - Amy R Nichols
- Department of Nutritional Sciences, School of Human Ecology, College of Natural Sciences, University of Texas at Austin, Austin, Texas, USA
| | - Lori A Hoepner
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University Medical Center, New York, New York, USA
- Department of Environmental and Occupational Health Sciences, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Eliza W Kinsey
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University Medical Center, New York, New York, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Judyth Ramirez-Carvey
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University Medical Center, New York, New York, USA
| | - Abeer Hassoun
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University Medical Center, New York, New York, USA
| | - Frederica P Perera
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University Medical Center, New York, New York, USA
| | - Radek Bukowski
- Department of Women's Health, Dell Medical School, University of Texas at Austin, Austin, Texas, USA
| | - Andrew G Rundle
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University Medical Center, New York, New York, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| |
Collapse
|
20
|
Furigo IC, Dearden L. Mechanisms mediating the impact of maternal obesity on offspring hypothalamic development and later function. Front Endocrinol (Lausanne) 2022; 13:1078955. [PMID: 36619540 PMCID: PMC9813846 DOI: 10.3389/fendo.2022.1078955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
As obesity rates have risen around the world, so to have pregnancies complicated by maternal obesity. Obesity during pregnancy is not only associated with negative health outcomes for the mother and the baby during pregnancy and birth, there is also strong evidence that exposure to maternal obesity causes an increased risk to develop obesity, diabetes and cardiovascular disease later in life. Animal models have demonstrated that increased weight gain in offspring exposed to maternal obesity is usually preceded by increased food intake, implicating altered neuronal control of food intake as a likely area of change. The hypothalamus is the primary site in the brain for maintaining energy homeostasis, which it coordinates by sensing whole body nutrient status and appropriately adjusting parameters including food intake. The development of the hypothalamus is plastic and regulated by metabolic hormones such as leptin, ghrelin and insulin, making it vulnerable to disruption in an obese in utero environment. This review will summarise how the hypothalamus develops, how maternal obesity impacts on structure and function of the hypothalamus in the offspring, and the factors that are altered in an obese in utero environment that may mediate the permanent changes to hypothalamic function in exposed individuals.
Collapse
Affiliation(s)
- Isadora C. Furigo
- Centre for Sport, Exercise and Life Sciences, School of Life Sciences, Coventry University, Coventry, United Kingdom
| | - Laura Dearden
- Metabolic Research Laboratories, Wellcome MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Laura Dearden,
| |
Collapse
|
21
|
Anwer H, Morris MJ, Noble DWA, Nakagawa S, Lagisz M. Transgenerational effects of obesogenic diets in rodents: A meta-analysis. Obes Rev 2022; 23:e13342. [PMID: 34595817 DOI: 10.1111/obr.13342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/22/2022]
Abstract
Obesity is a major health condition that affects millions worldwide. There is an increased interest in understanding the adverse outcomes associated with obesogenic diets. A multitude of studies have investigated the transgenerational impacts of maternal and parental obesogenic diets on subsequent generations of offspring, but results have largely been mixed. We conducted a systematic review and meta-analysis on rodent studies to elucidate how obesogenic diets impact the mean and variance of grand-offspring traits. Our study focused on transgenerational effects (i.e., F2 and F3 generations) in one-off and multigenerational exposure studies. From 33 included articles, we obtained 407 effect sizes representing pairwise comparisons of control and treatment grand-offspring groups pertaining to measures of body weight, adiposity, glucose, insulin, leptin, and triglycerides. We found evidence that male and female grand-offspring descended from grandparents exposed to an obesogenic diet displayed phenotypes consistent with metabolic syndrome, especially in cases where the obesogenic diet was continued across generations. Further, we found stronger evidence for the effects of grand-maternal than grand-paternal exposure on grand-offspring traits. A high-fat diet in one-off exposure studies did not seem to impact phenotypic variation, whereas in multigenerational exposure studies it reduced variation in several traits.
Collapse
Affiliation(s)
- Hamza Anwer
- Evolution and Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Margaret J Morris
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Daniel W A Noble
- Evolution and Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
- Division of Ecology and Evolution, Research School of Biology, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Shinichi Nakagawa
- Evolution and Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Malgorzata Lagisz
- Evolution and Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
22
|
Glavas MM, Lee AY, Miao I, Yang F, Mojibian M, O'Dwyer SM, Kieffer TJ. Developmental Timing of High-Fat Diet Exposure Impacts Glucose Homeostasis in Mice in a Sex-Specific Manner. Diabetes 2021; 70:2771-2784. [PMID: 34544729 PMCID: PMC8660987 DOI: 10.2337/db21-0310] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 09/03/2021] [Indexed: 11/24/2022]
Abstract
We previously demonstrated that male, but not female, Swiss Webster mice are susceptible to diabetes, with incidence increased by early overnutrition and high-fat diet (HFD). In this study, we investigated how HFD in Swiss Webster males and females during preweaning, peripubertal, and postpubertal periods alters glucose homeostasis and diabetes susceptibility. In males, HFD throughout life resulted in the highest diabetes incidence. Notably, switching to chow postpuberty was protective against diabetes relative to switching to chow at weaning, despite the longer period of HFD exposure. Similarly, HFD throughout life in males resulted in less liver steatosis relative to mice with shorter duration of postpubertal HFD. Thus, HFD timing relative to weaning and puberty, not simply exposure length, contributes to metabolic outcomes. Females were protected from hyperglycemia regardless of length or timing of HFD. However, postpubertal HFD resulted in a high degree of hepatic steatosis and adipose fibrosis, but glucose regulation and insulin sensitivity remained unchanged. Interestingly, peri-insulitis was observed in the majority of females but was not correlated with impaired glucose regulation. Our findings reveal critical periods of HFD-induced glucose dysregulation with striking sex differences in Swiss Webster mice, highlighting the importance of careful consideration of HFD timing relative to critical developmental periods.
Collapse
Affiliation(s)
- Maria M Glavas
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ann Y Lee
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ian Miao
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fan Yang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Majid Mojibian
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shannon M O'Dwyer
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Timothy J Kieffer
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
23
|
Song L, Yan J, Wang N, Wei X, Luo X, Meng K, Sun B. Prenatal exercise reverses high-fat-diet-induced placental alterations and alters male fetal hypothalamus during late gestation in rats†. Biol Reprod 2021; 102:705-716. [PMID: 31742332 DOI: 10.1093/biolre/ioz213] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/14/2019] [Accepted: 11/12/2019] [Indexed: 02/06/2023] Open
Abstract
Maternal high-fat (HF) diet negatively affects maternal metabolism and placental function. This study aimed to determine whether gestational exercise prevents the effect of HF diet on placental amino acid transporter expression and nutrient-sensing signaling and the fetal response. Pregnant Sprague-Dawley rats were either fed with a CHOW (13.5% fat) or HF (60% fat) diet during gestation and further divided into two subgroups: voluntary exercised and sedentary. Placentae were collected on gestational day (GD) 14 and GD20, and male placentae were used in this study. We found that gestational exercise ameliorated the detrimental effects of HF diet on dams' adiposity, plasma leptin, and insulin concentrations. Maternal exercise did not influence fetoplacental growth but affected male fetal hypothalamic Leprb, Stat3, Insr, Agrp, and Pomc expressions on GD20. Maternal HF diet decreased placental labyrinth thickness and increased system A amino acid transporter SNAT2 expression, while these changes were normalized by exercise. The activation of placental mechanistic target of rapamycin complex 1/4E-BP1 and LepRb/STAT3 signaling might contribute to the increased placental SNAT2 expression in HF-fed dams, which were reversed by exercise on GD20. These data highlight that gestational exercise reverses HF-diet-induced placental alterations during late gestation without influencing fetal growth. However, maternal exercise altered fetal hypothalamic gene expression, which may affect long-term offspring health.
Collapse
Affiliation(s)
- Lin Song
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jianqun Yan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Nan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Xiaojing Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiao Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Kai Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Bo Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
24
|
Kislal S, Jin W, Maesner C, Edlow AG. Mismatch between obesogenic intrauterine environment and low-fat postnatal diet may confer offspring metabolic advantage. Obes Sci Pract 2021; 7:450-461. [PMID: 34401203 PMCID: PMC8346367 DOI: 10.1002/osp4.501] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/25/2021] [Accepted: 02/19/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Mismatch between a depleted intrauterine environment and a substrate-rich postnatal environment confers an increased risk of offspring obesity and metabolic syndrome. Maternal diet-induced obesity (MATOB) is associated with the same outcomes. These experiments tested the hypothesis that a mismatch between a nutrient-rich intrauterine environment and a low-fat postnatal environment would ameliorate offspring metabolic morbidity. METHODS C57BL6/J female mice were fed either a 60% high-fat diet (HFD) or a 10% fat control diet (CD) for 14-week pre-breeding and during pregnancy/lactation. Offspring were weaned to CD. Weight was evaluated weekly; body composition was determined using EchoMRI. Serum fasting lipids and glucose and insulin tolerance tests were performed. Metabolic rate, locomotor, and sleep behavior were evaluated with indirect calorimetry. RESULTS MATOB-exposed/CD-weaned offspring of both sexes had improved glucose tolerance and insulin sensitivity compared to controls. Males had improved fasting lipids. Females had significantly increased weight and body fat percentage in adulthood compared to sex-matched controls. Females also had significantly increased sleep duration and reduced locomotor activity compared to males. CONCLUSIONS Reduced-fat dietary switch following intrauterine and lactational exposure to MATOB was associated with improved glucose handling and lipid profiles in adult offspring, more pronounced in males. A mismatch between a high-fat prenatal and low-fat postnatal environment may confer a metabolic advantage. The amelioration of deleterious metabolic programming by strict offspring adherence to a low-fat diet may have translational potential.
Collapse
Affiliation(s)
- Sezen Kislal
- Vincent Center for Reproductive BiologyMassachusetts General HospitalBostonMassachusettsUSA
| | - William Jin
- Vincent Center for Reproductive BiologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Claire Maesner
- Vincent Center for Reproductive BiologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Andrea G. Edlow
- Vincent Center for Reproductive BiologyMassachusetts General HospitalBostonMassachusettsUSA
- Department of Obstetrics and GynecologyMassachusetts General HospitalBostonMassachusettsUSA
| |
Collapse
|
25
|
Takaya J. Calcium-Deficiency during Pregnancy Affects Insulin Resistance in Offspring. Int J Mol Sci 2021; 22:ijms22137008. [PMID: 34209784 PMCID: PMC8268058 DOI: 10.3390/ijms22137008] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 02/08/2023] Open
Abstract
Prenatal malnutrition is known to affect the phenotype of the offspring through changes in epigenetic regulation. Growing evidence suggests that epigenetics is one of the mechanisms by which nutrients and minerals affect metabolic traits. Although the perinatal period is the time of highest phenotypic plasticity, which contributes largely to developmental programming, there is evidence of nutritional influence on epigenetic regulation during adulthood. Calcium (Ca) plays an important role in the pathogenesis of insulin resistance syndrome. Cortisol, the most important glucocorticoid, is considered to lead to insulin resistance and metabolic syndrome. 11β-hydroxysteroid dehydrogenase-1 is a key enzyme that catalyzes the intracellular conversion of cortisone to physiologically active cortisol. This brief review aims to identify the effects of Ca deficiency during pregnancy and/or lactation on insulin resistance in the offspring. Those findings demonstrate that maternal Ca deficiency during pregnancy may affect the epigenetic regulation of gene expression and thereby induce different metabolic phenotypes. We aim to address the need for Ca during pregnancy and propose the scaling-up of clinical and public health approaches that improved pregnancy outcomes.
Collapse
Affiliation(s)
- Junji Takaya
- Department of Pediatrics, Kawachi General Hospital, 1-31 Yokomakura, Higashi-Osaka 578-0954, Osaka, Japan
| |
Collapse
|
26
|
Casasnovas J, Damron CL, Jarrell J, Orr KS, Bone RN, Archer-Hartmann S, Azadi P, Kua KL. Offspring of Obese Dams Exhibit Sex-Differences in Pancreatic Heparan Sulfate Glycosaminoglycans and Islet Insulin Secretion. Front Endocrinol (Lausanne) 2021; 12:658439. [PMID: 34108935 PMCID: PMC8181410 DOI: 10.3389/fendo.2021.658439] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/20/2021] [Indexed: 12/12/2022] Open
Abstract
Offspring of obese mothers suffer higher risks of type 2 diabetes due to increased adiposity and decreased β cell function. To date, the sex-differences in offspring islet insulin secretion during early life has not been evaluated extensively, particularly prior to weaning at postnatal day 21 (P21). To determine the role of maternal obesity on offspring islet insulin secretion, C57BL/6J female dams were fed chow or western diet from 4 weeks prior to mating to induce maternal obesity. First, offspring of chow-fed and obese dams were evaluated on postnatal day 21 (P21) prior to weaning for body composition, glucose and insulin tolerance, and islet phasic insulin-secretion. Compared to same-sex controls, both male and female P21 offspring born to obese dams (MatOb) had higher body adiposity and exhibited sex-specific differences in glucose tolerance and insulin secretion. The male MatOb offspring developed the highest extent of glucose intolerance and lowest glucose-induced insulin secretion. In contrast, P21 female offspring of obese dams had unimpaired insulin secretion. Using SAX-HPLC, we found that male MatOb had a decrease in pancreatic heparan sulfate glycosaminoglycan, which is a macromolecule critical for islet health. Notably, 8-weeks-old offspring of obese dams continued to exhibit a similar pattern of sex-differences in glucose intolerance and decreased islet insulin secretion. Overall, our study suggests that maternal obesity induces sex-specific changes to pancreatic HSG in offspring and a lasting effect on offspring insulin secretion, leading to the sex-differences in glucose intolerance.
Collapse
Affiliation(s)
- Jose Casasnovas
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Christopher Luke Damron
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - James Jarrell
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kara S. Orr
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Robert N. Bone
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | - Kok Lim Kua
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
27
|
Govindsamy A, Ghoor S, Cerf ME. Programming With Varying Dietary Fat Content Alters Cardiac Insulin Receptor, Glut4 and FoxO1 Immunoreactivity in Neonatal Rats, Whereas High Fat Programming Alters Cebpa Gene Expression in Neonatal Female Rats. Front Endocrinol (Lausanne) 2021; 12:772095. [PMID: 35069436 PMCID: PMC8766637 DOI: 10.3389/fendo.2021.772095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/01/2021] [Indexed: 12/20/2022] Open
Abstract
Fetal programming refers to an intrauterine stimulus or insult that shapes growth, development and health outcomes. Dependent on the quality and quantity, dietary fats can be beneficial or detrimental for the growth of the fetus and can alter insulin signaling by regulating the expression of key factors. The effects of varying dietary fat content on the expression profiles of factors in the neonatal female and male rat heart were investigated and analyzed in control (10% fat), 20F (20% fat), 30F (30% fat) and 40F (40% fat which was a high fat diet used to induce high fat programming) neonatal rats. The whole neonatal heart was immunostained for insulin receptor, glucose transporter 4 (Glut4) and forkhead box protein 1 (FoxO1), followed by image analysis. The expression of 84 genes, commonly associated with the insulin signaling pathway, were then examined in 40F female and 40F male offspring. Maintenance on diets, varying in fat content during fetal life, altered the expression of cardiac factors, with changes induced from 20% fat in female neonates, but from 30% fat in male neonates. Further, CCAAT/enhancer-binding protein alpha (Cebpa) was upregulated in 40F female neonates. There was, however, differential expression of several insulin signaling genes in 40F (high fat programmed) offspring, with some tending to significance but most differences were in fold changes (≥1.5 fold). The increased immunoreactivity for insulin receptor, Glut4 and FoxO1 in 20F female and 30F male neonatal rats may reflect a compensatory response to programming to maintain cardiac physiology. Cebpa was upregulated in female offspring maintained on a high fat diet, with fold increases in other insulin signaling genes viz. Aebp1, Cfd (adipsin), Adra1d, Prkcg, Igfbp, Retn (resistin) and Ucp1. In female offspring maintained on a high fat diet, increased Cebpa gene expression (concomitant with fold increases in other insulin signaling genes) may reflect cardiac stress and an adaptative response to cardiac inflammation, stress and/or injury, after high fat programming. Diet and the sex are determinants of cardiac physiology and pathophysiology, reflecting divergent mechanisms that are sex-specific.
Collapse
Affiliation(s)
- Annelene Govindsamy
- Discipline of Pharmaceutical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Samira Ghoor
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa
| | - Marlon E. Cerf
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa
- Grants, Innovation and Product Development, South African Medical Research Council, Cape Town, South Africa
- *Correspondence: Marlon E. Cerf,
| |
Collapse
|
28
|
Kulhanek D, Weigel R, Paulsen ME. Maternal High-Fat-High-Carbohydrate Diet-Induced Obesity Is Associated with Increased Appetite in Peripubertal Male but Not Female C57Bl/6J Mice. Nutrients 2020; 12:E2919. [PMID: 32987812 PMCID: PMC7598591 DOI: 10.3390/nu12102919] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/17/2022] Open
Abstract
Diet-induced maternal obesity might play a critical role in altering hypothalamic development, predisposing the offspring to obesity and metabolic disease later in life. The objective of this study was to describe both phenotypic and molecular sex differences in peripubertal offspring energy homeostasis, using a mouse model of maternal obesity induced by a high-fat-high-carbohydrate (HFHC) diet. We report that males, not females, exposed to a maternal HFHC diet had increased energy intake. Males exposed to a maternal HFHC diet had a 15% increased meal size and a 46% increased frequency, compared to the control (CON) males, without a change in energy expenditure. CON and HFHC offspring did not differ in body weight, composition, or plasma metabolic profile. HFHC diet caused decreased hypothalamic glucocorticoid expression, which was further decreased in males compared to females. Maternal weight, maternal caloric intake, and male offspring meal frequency were inversely correlated with offspring hypothalamic insulin receptor (IR) expression. There was a significant interaction between maternal-diet exposure and sex in hypothalamic IR. Based on our preclinical data, we suggest that interventions focusing on normalizing maternal nutrition might be considered to attenuate nutritional influences on obesity programming and curb the continuing rise in obesity rates.
Collapse
Affiliation(s)
| | | | - Megan E. Paulsen
- Department of Pediatrics, Division of Neonatology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (D.K.); (R.W.)
| |
Collapse
|
29
|
Tozour J, Hughes F, Carrier A, Vieau D, Delahaye F. Prenatal Hyperglycemia Exposure and Cellular Stress, a Sugar-Coated View of Early Programming of Metabolic Diseases. Biomolecules 2020; 10:E1359. [PMID: 32977673 PMCID: PMC7598660 DOI: 10.3390/biom10101359] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022] Open
Abstract
Worldwide, the number of people with diabetes has quadrupled since 1980 reaching 422 million in 2014 (World Health Organization). This distressing rise in diabetes also affects pregnant women and thus, in regard to early programming of adult diseases, creates a vicious cycle of metabolic dysfunction passed from one generation to another. Metabolic diseases are complex and caused by the interplay between genetic and environmental factors. High-glucose exposure during in utero development, as observed with gestational diabetes mellitus (GDM), is an established risk factor for metabolic diseases. Despite intense efforts to better understand this phenomenon of early memory little is known about the molecular mechanisms associating early exposure to long-term diseases risk. However, evidence promotes glucose associated oxidative stress as one of the molecular mechanisms able to influence susceptibility to metabolic diseases. Thus, we decided here to further explore the relationship between early glucose exposure and cellular stress in the context of early development, and focus on the concept of glycemic memory, its consequences, and sexual dimorphic and epigenetic aspects.
Collapse
Affiliation(s)
- Jessica Tozour
- Department of Obstetrics and Gynecology, NYU Winthrop Hospital, Mineola, NY 11501, USA;
| | - Francine Hughes
- Obstetrics & Gynecology and Women’s Health, Division of Maternal-Fetal Medicine, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Arnaud Carrier
- Institut Pasteur de Lille, U1283-UMR 8199 EGID, Université de Lille, Inserm, CNRS, CHU Lille, F-59000 Lille, France;
| | - Didier Vieau
- BiologyDepartment, LilNCog Lille Neurosciences and Cognition U 1172, Université de Lille, Inserm, CHU Lille, F-59000 Lille, France;
| | - Fabien Delahaye
- Institut Pasteur de Lille, U1283-UMR 8199 EGID, Université de Lille, Inserm, CNRS, CHU Lille, F-59000 Lille, France;
| |
Collapse
|
30
|
Kislal S, Shook LL, Edlow AG. Perinatal exposure to maternal obesity: Lasting cardiometabolic impact on offspring. Prenat Diagn 2020; 40:1109-1125. [PMID: 32643194 DOI: 10.1002/pd.5784] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 06/25/2020] [Accepted: 07/05/2020] [Indexed: 12/11/2022]
Abstract
Evidence from epidemiological, clinical, and animal model studies clearly demonstrates that prenatal and lactational maternal obesity and high-fat diet consumption are associated with cardiometabolic morbidity in offspring. Fetal and offspring sex may be an important effect modifier. Adverse offspring cardiometabolic outcomes observed in the setting of maternal obesity include an increased risk for obesity, features of metabolic syndrome (hypertension, hyperglycemia and insulin resistance, hyperlipidemia, increased adiposity), and non-alcoholic fatty liver disease. This review article synthesizes human and animal data linking maternal obesity and high-fat diet consumption in pregnancy and lactation to adverse cardiometabolic outcomes in offspring. We review key mechanisms underlying skeletal muscle, adipose tissue, pancreatic, liver, and central brain reward programming in obesity-exposed offspring, and how such malprogramming contributes to offspring cardiometabolic morbidity.
Collapse
Affiliation(s)
- Sezen Kislal
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lydia L Shook
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrea G Edlow
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA.,Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
31
|
Glendining KA, Fisher LC, Jasoni CL. Maternal Obesity Modulates Expression of Satb2 in Hypothalamic VMN of Female Offspring. Life (Basel) 2020; 10:life10040048. [PMID: 32344561 PMCID: PMC7235991 DOI: 10.3390/life10040048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 01/24/2023] Open
Abstract
Maternal obesity during pregnancy is associated with a greater risk of poor health outcomes in offspring, including obesity, metabolic disorders, and anxiety, however the incidence of these diseases differs for males and females. Similarly, animal models of maternal obesity have reported sex differences in offspring, for both metabolic outcomes and anxiety-like behaviors. The ventromedial nucleus of the hypothalamus (VMN) is a brain region known to be involved in the regulation of both metabolism and anxiety, and is well documented to be sexually dimorphic. As the VMN is largely composed of glutamatergic neurons, which are important for its functions in modulating metabolism and anxiety, we hypothesized that maternal obesity may alter the number of glutamatergic neurons in the offspring VMN. We used a mouse model of a maternal high-fat diet (mHFD), to examine mRNA expression of the glutamatergic neuronal marker Satb2 in the mediobasal hypothalamus of control and mHFD offspring at GD17.5. We found sex differences in Satb2 expression, with mHFD-induced upregulation of Satb2 mRNA in the mediobasal hypothalamus of female offspring, compared to controls, but not males. Using immunohistochemistry, we found an increase in the number of SATB2-positive cells in female mHFD offspring VMN, compared to controls, which was localized to the rostral region of the nucleus. These data provide evidence that maternal nutrition during gestation alters the developing VMN, possibly increasing its glutamatergic drive of offspring in a sex-specific manner, which may contribute to sexual dimorphism in offspring health outcomes later in life.
Collapse
|
32
|
Kruse M, Keyhani-Nejad F, Osterhoff MA, Pfeiffer AFH. Sexually dimorphic metabolic responses to exposure of a high fat diet during pregnancy, lactation and early adulthood in Gipr -/- mice. Peptides 2020; 125:170250. [PMID: 31917165 DOI: 10.1016/j.peptides.2019.170250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/22/2019] [Accepted: 12/26/2019] [Indexed: 12/25/2022]
Abstract
Obesity has a multifactorial origin. It is known that alterations of the intra uterine milieu induce developmental programming effects leading to metabolic diseases in offspring. Obesity is diminished in mice lacking the glucose-dependent insulinotropic polypeptide receptor (Gipr-/-) when exposed to a high fat diet (HFD). We investigated whether Gipr-/- mice are still protected from obesity when additionally exposure to a HFD during pregnancy and lactation occurs. Male and female wild type (WT) and Gipr-/- offspring received either a control/ low fat diet or HFD during pregnancy and lactation and were then either left on this diet or placed on the opposite diet after weaning until 24 weeks of life. Female WT mice showed increased body weight and adiposity when exposed to a HFD during pregnancy and lactation and post-weaning compared to female WT that received the HFD after weaning only. This exacerbated effect of a HFD during pregnancy and lactation was abolished in female Gipr-/- mice. Male Gipr-/- mice were protected from obesity to a much lesser extent. Male Gipr-/- mice exposed to a HFD during pregnancy and lactation and after weaning exhibited significantly increased fed serum glucose compared to Gipr-/- mice exposed to a HFD after weaning only. In female Gipr-/- mice no differences in fed blood glucose were observed between these groups. Our data indicate that female Gipr-/- mice are more protected from obesity. This protection is preserved in female Gipr-/- mice when additional deleterious effects of a HFD occur during fetal development.
Collapse
Affiliation(s)
- Michael Kruse
- Department of Clinical Nutrition, German Institute of Human Nutrition, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; Department for Endocrinology, Diabetes and Nutrition, Charité - University of Medicine, Hindenburgdamm 30, 12200 Berlin, Germany
| | - Farnaz Keyhani-Nejad
- Department of Clinical Nutrition, German Institute of Human Nutrition, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; Department for Endocrinology, Diabetes and Nutrition, Charité - University of Medicine, Hindenburgdamm 30, 12200 Berlin, Germany
| | - Martin A Osterhoff
- Department of Clinical Nutrition, German Institute of Human Nutrition, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; Department for Endocrinology, Diabetes and Nutrition, Charité - University of Medicine, Hindenburgdamm 30, 12200 Berlin, Germany
| | - Andreas F H Pfeiffer
- Department of Clinical Nutrition, German Institute of Human Nutrition, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; Department for Endocrinology, Diabetes and Nutrition, Charité - University of Medicine, Hindenburgdamm 30, 12200 Berlin, Germany; German Center for Diabetes Research, Germany.
| |
Collapse
|
33
|
Alves JM, Luo S, Chow T, Herting M, Xiang AH, Page KA. Sex differences in the association between prenatal exposure to maternal obesity and hippocampal volume in children. Brain Behav 2020; 10:e01522. [PMID: 31903710 PMCID: PMC7010582 DOI: 10.1002/brb3.1522] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 12/05/2019] [Accepted: 12/09/2019] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION Animal studies have shown that male but not female offspring exposed to maternal obesity have abnormal hippocampal development. Similar sex differences were observed in animal models of developmental programming by prenatal stress or maternal diabetes. We aimed to translate this work into humans by examining sex-specific effects of exposure to maternal obesity on hippocampal volume in children. METHODS Eighty-eight children (37 boys and 51 girls) aged 7-11 years completed the study. Maternal prepregnancy body mass index (BMI) was obtained from electronic medical records. A high-resolution anatomical scan was performed using a 3-Tesla magnetic resonance imaging (MRI) scanner. Total hippocampal volume and hippocampal subfield volumes were analyzed using FreeSurfer 6.0. Linear regression was used to investigate sex differences in relationships between maternal prepregnancy BMI and child hippocampal volume. RESULTS Maternal prepregnancy BMI ranged from 19.0 to 50.4 kg/m2 . We observed a significant interaction between maternal prepregnancy BMI and sex on total hippocampal volume (p < .001) such that boys (r = -.39, p = .018) but not girls (r = .11, p = .45) had a significant negative relationship between maternal prepregnancy BMI and total hippocampal volume. This relationship in boys remained significant after adjusting for child and maternal covariates (β = -126.98, p = .012). The sex interactions with prepregnancy BMI were consistently observed in hippocampal subfields CA1 (p = .008), CA2/3 (p = .016), CA4 (p = .002), dentate gyrus (p < .001), and subiculum (p < .001). CONCLUSIONS Our results support findings in animal models and suggest that boys may be more vulnerable to the adverse effects of exposure to maternal obesity on hippocampal development than girls.
Collapse
Affiliation(s)
- Jasmin M Alves
- Division of Endocrinology, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Diabetes and Obesity Research Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shan Luo
- Division of Endocrinology, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Diabetes and Obesity Research Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ting Chow
- Department of Research and Evaluation, Pasadena, CA, USA
| | - Megan Herting
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anny H Xiang
- Department of Research and Evaluation, Pasadena, CA, USA
| | - Kathleen A Page
- Division of Endocrinology, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Diabetes and Obesity Research Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
34
|
Badran M, Yassin BA, Lin DTS, Kobor MS, Ayas N, Laher I. Gestational intermittent hypoxia induces endothelial dysfunction, reduces perivascular adiponectin and causes epigenetic changes in adult male offspring. J Physiol 2019; 597:5349-5364. [PMID: 31441069 DOI: 10.1113/jp277936] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS Obstructive sleep apnoea (OSA) is characterized by intermittent hypoxia, which causes oxidative stress and inflammation and increases the risk of cardiovascular disease. OSA during pregnancy causes adverse maternal and fetal outcomes. The effects of pre-existing OSA in pregnant women on cardiometabolic outcomes in the offspring are unknown. We evaluated basic metabolic parameters, as well as aortic vascular and perivascular adipose tissue (PVAT) function in response to adiponectin, and examined DNA methylation of adiponectin gene promoter in PVAT in 16-week-old adult offspring exposed to gestational intermittent hypoxia (GIH). GIH decreased body weights at week 1 in both male and female offspring, and caused subsequent increases in body weight and food consumption in male offspring only. Adult female offspring had normal levels of lipids, glucose and insulin, with no endothelial dysfunction. Adult male offspring exhibited dyslipidaemia, insulin resistance and hyperleptinaemia. Decreased endothelial-dependent vasodilatation, loss of anti-contractile activity of PVAT and low circulating PVAT adiponectin levels, as well as increased pro-inflammatory gene expression and DNA methylation of adiponectin gene promoter, occurred in adult male offspring. Our results suggest that male offspring of women with OSA could be at risk of developing cardiometabolic disease during adulthood. ABSTRACT Perturbations during pregnancy can program the offspring to develop cardiometabolic diseases later in life. Obstructive sleep apnoea (OSA) is a chronic condition that frequently affects pregnancies and leads to adverse fetal outcomes. We assessed the offspring of female mice experiencing gestational intermittent hypoxia (GIH), a hallmark of OSA, for changes in metabolic profiles, aortic nitric oxide (NO)-dependent relaxations, perivascular adipose tissue (PVAT) anti-contractile activities and the responses to adiponectin, and DNA methylation of the adiponectin gene promoter in PVAT tissue. Pregnant mouse dams were exposed to intermittent hypoxic cycles ( F I O 2 21-12%) for 18 days. GIH resulted in lower body weights of pups at week 1, followed by significant weight gain by week 16 of age in male but not female offspring. Plasma lipids, leptin and insulin resistance were higher in GIH male adult offspring. Endothelium-dependent relaxation in response to ACh and the anti-contractile activity of PVAT in the abdominal aorta was reduced in GIH adult male offspring. Incubation of arteries from GIH adult male offspring with adiponectin restored the anti-contractile activity of PVAT. Both circulating and PVAT tissue homogenate levels of adiponectin, as well as gene expression of adiponectin in PVAT, were lower in GIH male offspring, along with an increased gene expression of inflammatory cytokines. Pyrosequencing of adiponectin gene promoter in PVAT showed increased DNA methylation in GIH male offspring. Our results indicate that GIH leads to vascular disease in adult male offspring through PVAT dysfunction, which was associated with low adiponectin levels and epigenetic modifications on the adiponectin gene promoter.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Bisher Abu Yassin
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - David Tse Shen Lin
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Najib Ayas
- Divisions of Critical Care and Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Sleep Disorders Program, UBC Hospital, Vancouver, BC, Canada.,Division of Critical Care Medicine, Providence Healthcare, Vancouver, BC, Canada
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
35
|
Bouwman LMS, Fernández-Calleja JMS, van der Stelt I, Oosting A, Keijer J, van Schothorst EM. Replacing Part of Glucose with Galactose in the Postweaning Diet Protects Female But Not Male Mice from High-Fat Diet-Induced Adiposity in Later Life. J Nutr 2019; 149:1140-1148. [PMID: 31076770 PMCID: PMC6602901 DOI: 10.1093/jn/nxz028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/11/2018] [Accepted: 02/04/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Duration of breastfeeding is positively associated with decreased adiposity and increased metabolic health in later life, which might be related to galactose. OBJECTIVE The aim of this study was to investigate if partial replacement of glucose with galactose in the postweaning diet had a metabolic programming effect. METHODS Male and female mice (C57BL/6JRccHsd) received an isocaloric diet (16 energy% fat; 64 energy% carbohydrates; 20 energy% protein) with either glucose (32 energy%) (GLU) or glucose + galactose (GLU + GAL, 16 energy% each) for 3 wk postweaning. Afterwards, all mice were switched to the same 40 energy% high-fat diet (HFD) for 9 wk to evaluate potential programming effects in an obesogenic environment. Data were analyzed within sex. RESULTS Female body weight (-14%) and fat mass (-47%) were significantly lower at the end of the HFD period (both P < 0.001) among those fed GLU + GAL than among those fed GLU; effects in males were in line with these findings but nonsignificant. Food intake was affected in GLU + GAL-fed females (+8% on postweaning diet, -9% on HFD) compared with GLU-fed females, but not for hypothalamic transcript levels at endpoint. Also, in GLU + GAL-fed females, serum insulin concentrations (-48%, P < 0.05) and the associated homeostasis model assessment of insulin resistance (HOMA-IR) were significantly lower ( P < 0.05) at endpoint, but there were no changes in pancreas morphology. In GLU + GAL-fed females, expression of insulin receptor substrate 2 (Irs2) (-27%, P < 0.01 ; -44%, P < 0.001) and the adipocyte size markers leptin (Lep) (-40%, P < 0.05; -63% , P < 0.05) and mesoderm-specific transcript homolog protein (Mest) (-80%, P < 0.05; -72%, P < 0.05) was lower in gonadal and subcutaneous white adipose tissue (WAT), respectively. Expression of insulin receptor substrate1 (Irs1) (-24%, P < 0.05) was only lower in subcutaneous WAT in GLU + GAL-fed females. CONCLUSIONS Partial replacement of glucose with galactose, resulting in a 1:1 ratio mimicking lactose, in a 3-wk postweaning diet lowered body weight, adiposity, HOMA-IR, and expression of WAT insulin signaling in HFD-challenged female mice in later life. This suggests that prolonged galactose intake may improve metabolic and overall health in later life.
Collapse
Affiliation(s)
- Lianne M S Bouwman
- Human and Animal Physiology, Wageningen University, Wageningen, Netherlands
| | | | - Inge van der Stelt
- Human and Animal Physiology, Wageningen University, Wageningen, Netherlands
| | | | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, Wageningen, Netherlands
| | - Evert M van Schothorst
- Human and Animal Physiology, Wageningen University, Wageningen, Netherlands,Address correspondence to EMvS (e-mail: )
| |
Collapse
|
36
|
Hsu CN, Tain YL. The Good, the Bad, and the Ugly of Pregnancy Nutrients and Developmental Programming of Adult Disease. Nutrients 2019; 11:nu11040894. [PMID: 31010060 PMCID: PMC6520975 DOI: 10.3390/nu11040894] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 12/14/2022] Open
Abstract
Maternal nutrition plays a decisive role in developmental programming of many non-communicable diseases (NCDs). A variety of nutritional insults during gestation can cause programming and contribute to the development of adult-onset diseases. Nutritional interventions during pregnancy may serve as reprogramming strategies to reverse programming processes and prevent NCDs. In this review, firstly we summarize epidemiological evidence for nutritional programming of human disease. It will also discuss evidence from animal models, for the common mechanisms underlying nutritional programming, and potential nutritional interventions used as reprogramming strategies.
Collapse
Affiliation(s)
- Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan.
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| |
Collapse
|
37
|
Guo J, Li B, Zuo Z, Chen M, Wang C. Maternal Supplementation with β‐Carotene During Pregnancy Disturbs Lipid Metabolism and Glucose Homoeostasis in F1 Female Mice. Mol Nutr Food Res 2019; 63:e1900072. [DOI: 10.1002/mnfr.201900072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Jiaojiao Guo
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen University Xiamen 36110 P. R. China
| | - Bingshui Li
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen University Xiamen 36110 P. R. China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen University Xiamen 36110 P. R. China
| | - Meng Chen
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen University Xiamen 36110 P. R. China
- Key Laboratory of Ministry of Education for Subtropical Wetland Ecosystem ResearchXiamen University Xiamen 36110 P. R. China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen University Xiamen 36110 P. R. China
- Key Laboratory of Ministry of Education for Subtropical Wetland Ecosystem ResearchXiamen University Xiamen 36110 P. R. China
| |
Collapse
|
38
|
Cerf ME. High Fat Programming and Cardiovascular Disease. MEDICINA (KAUNAS, LITHUANIA) 2018; 54:E86. [PMID: 30428585 PMCID: PMC6262472 DOI: 10.3390/medicina54050086] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/31/2018] [Accepted: 11/08/2018] [Indexed: 02/06/2023]
Abstract
Programming is triggered through events during critical developmental phases that alter offspring health outcomes. High fat programming is defined as the maintenance on a high fat diet during fetal and/or early postnatal life that induces metabolic and physiological alterations that compromise health. The maternal nutritional status, including the dietary fatty acid composition, during gestation and/or lactation, are key determinants of fetal and postnatal development. A maternal high fat diet and obesity during gestation compromises the maternal metabolic state and, through high fat programming, presents an unfavorable intrauterine milieu for fetal growth and development thereby conferring adverse cardiac outcomes to offspring. Stressors on the heart, such as a maternal high fat diet and obesity, alter the expression of cardiac-specific factors that alter cardiac structure and function. The proper nutritional balance, including the fatty acid balance, particularly during developmental windows, are critical for maintaining cardiac structure, preserving cardiac function and enhancing the cardiac response to metabolic challenges.
Collapse
Affiliation(s)
- Marlon E Cerf
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg 7505, South Africa.
| |
Collapse
|
39
|
McCoski SR, Vailes MT, Owens CE, Cockrum RR, Ealy AD. Exposure to maternal obesity alters gene expression in the preimplantation ovine conceptus. BMC Genomics 2018; 19:737. [PMID: 30305020 PMCID: PMC6180665 DOI: 10.1186/s12864-018-5120-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023] Open
Abstract
Background Embryonic and fetal exposure to maternal obesity causes several maladaptive morphological and epigenetic changes in exposed offspring. The timing of these events is unclear, but changes can be observed even after a short exposure to maternal obesity around the time of conception. The hypothesis of this work is that maternal obesity influences the ovine preimplantation conceptus early in pregnancy, and this exposure will affect gene expression in embryonic and extraembryonic tissues. Results Obese and lean ewe groups were established by overfeeding or normal feeding, respectively. Ewes were then bred to genetically similar rams. Conceptuses were collected at day 14 of gestation. Morphological assessments were made, conceptuses were sexed by genomic PCR analysis, and samples underwent RNA-sequencing analysis. While no obvious morphological differences existed between conceptuses, differentially expressed genes (≥ 2-fold; ≥ 0.2 RPKM; ≤ 0.05 FDR) were detected based on maternal obesity exposure (n = 21). Also, differential effects of maternal obesity were noted on each conceptus sex (n = 347). A large portion of differentially expressed genes were associated with embryogenesis and placental development. Conclusions Findings reveal that the preimplantation ovine conceptus genome responds to maternal obesity in a sex-dependent manner. The sexual dimorphism in response to the maternal environment coupled with changes in placental gene expression may explain aberrations in phenotype observed in offspring derived from obese females. Electronic supplementary material The online version of this article (10.1186/s12864-018-5120-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sarah R McCoski
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, 3430 Litton-Reaves Hall (0306), Virginia, Blacksburg, VA, 24061, USA
| | - McCauley T Vailes
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, 3430 Litton-Reaves Hall (0306), Virginia, Blacksburg, VA, 24061, USA
| | - Connor E Owens
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Rebecca R Cockrum
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Alan D Ealy
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, 3430 Litton-Reaves Hall (0306), Virginia, Blacksburg, VA, 24061, USA.
| |
Collapse
|
40
|
Csongová M, Gurecká R, Koborová I, Celec P, Domonkos E, Uličná O, Somoza V, Šebeková K. The effects of a maternal advanced glycation end product-rich diet on somatic features, reflex ontogeny and metabolic parameters of offspring mice. Food Funct 2018; 9:3432-3446. [PMID: 29877548 DOI: 10.1039/c8fo00183a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Maternal exposure to a Western type diet during pregnancy might predispose the offspring to manifestation of metabolic and behavioral disturbances in later life. The Western type diet contains large amounts of advanced glycation end products (AGEs). In humans and experimental rodents, the intake of an AGE-rich diet (AGE-RD) negatively affected glucose homeostasis, and initiated the production of reactive oxygen species. Rats consuming the AGE-RD presented changes in behavior. It remains unclear whether maternal intake of the AGE-RD might affect developmental plasticity in offspring. We examined early somatic (weight, incisor eruption, ear unfolding, and eye opening) and neuromotor development, oxidative status, insulin sensitivity (HOMA index) and locomotor activity assessed in PhenoTyper cages in the offspring of mice fed during pregnancy with either the AGE-RD (25% bread crusts/75% control chow) or control chow. Until weaning, the somatic development of offspring did not differ between the two dietary groups. The AGE-RD offspring manifested physiological reflexes (auditory startle, eye lid, ear twitch and righting reflexes) earlier. As young adults, the male offspring of the AGE-RD dams were heavier and less insulin sensitive compared with their control counterparts. The AGE-RD offspring showed higher locomotor activity during the active phase. Our data indicate that the maternal AGE-RD during pregnancy might accelerate the maturation of reflexes in offspring, predispose the male progeny to weight gain and affect their glucose homeostasis. These effects manifest without the direct consumption of the AGE-RD by offspring. Further work is needed to determine the mechanisms by which the maternal AGE-RD affects neurobehavioral pathways in offspring, as well as sex differences in adverse metabolic responses.
Collapse
Affiliation(s)
- Melinda Csongová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Berends LM, Dearden L, Tung YCL, Voshol P, Fernandez-Twinn DS, Ozanne SE. Programming of central and peripheral insulin resistance by low birthweight and postnatal catch-up growth in male mice. Diabetologia 2018; 61:2225-2234. [PMID: 30043179 PMCID: PMC6133152 DOI: 10.1007/s00125-018-4694-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/13/2018] [Indexed: 02/06/2023]
Abstract
AIMS Intra-uterine growth restriction (IUGR) followed by accelerated postnatal growth is associated with an increased risk of obesity and type 2 diabetes. We aimed to determine central and peripheral insulin sensitivity in mice that underwent IUGR followed by postnatal catch-up growth and investigate potential molecular mechanisms underpinning their physiology. METHODS We used a C57BL/6J mouse model of maternal diet-induced IUGR (maternal diet, 8% protein) followed by cross-fostering to a normal nutrition dam (maternal diet, 20% protein) and litter size manipulation to cause accelerated postnatal catch-up growth. We performed intracerebroventricular insulin injection and hyperinsulinaemic-euglycaemic clamp studies to examine the effect of this early nutritional manipulation on central and peripheral insulin resistance. Furthermore, we performed quantitative real-time PCR and western blotting to examine the expression of key insulin-signalling components in discrete regions of the hypothalamus. RESULTS IUGR followed by accelerated postnatal growth caused impaired glucose tolerance and peripheral insulin resistance. In addition, these 'recuperated' animals were resistant to the anorectic effects of central insulin administration. This central insulin resistance was associated with reduced protein levels of the p110β subunit of phosphoinositide 3-kinase (PI3K) and increased serine phosphorylation of IRS-1 in the arcuate nucleus (ARC) of the hypothalamus. Expression of the gene encoding protein tyrosine phosphatase 1B (PTP1B; Ptpn1) was also increased specifically in this region of the hypothalamus. CONCLUSIONS/INTERPRETATION Mice that undergo IUGR followed by catch-up growth display peripheral and central insulin resistance in adulthood. Recuperated offspring show changes in expression/phosphorylation of components of the insulin signalling pathway in the ARC. These defects may contribute to the resistance to the anorectic effects of central insulin, as well as the impaired glucose homeostasis seen in these animals.
Collapse
Affiliation(s)
- Lindsey M Berends
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Laura Dearden
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Yi Chun L Tung
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Peter Voshol
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Denise S Fernandez-Twinn
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
42
|
Dearden L, Bouret SG, Ozanne SE. Sex and gender differences in developmental programming of metabolism. Mol Metab 2018; 15:8-19. [PMID: 29773464 PMCID: PMC6066743 DOI: 10.1016/j.molmet.2018.04.007] [Citation(s) in RCA: 223] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The early life environment experienced by an individual in utero and during the neonatal period is a major factor in shaping later life disease risk-including susceptibility to develop obesity, diabetes, and cardiovascular disease. The incidence of metabolic disease is different between males and females. How the early life environment may underlie these sex differences is an area of active investigation. SCOPE OF REVIEW The purpose of this review is to summarize our current understanding of how the early life environment influences metabolic disease risk in a sex specific manner. We also discuss the possible mechanisms responsible for mediating these sexually dimorphic effects and highlight the results of recent intervention studies in animal models. MAJOR CONCLUSIONS Exposure to states of both under- and over-nutrition during early life predisposes both sexes to develop metabolic disease. Females seem particularly susceptible to develop increased adiposity and disrupted glucose homeostasis as a result of exposure to in utero undernutrition or high sugar environments, respectively. The male placenta is particularly vulnerable to damage by adverse nutritional states and this may underlie some of the metabolic phenotypes observed in adulthood. More studies investigating both sexes are needed to understand how changes to the early life environment impact differently on the long-term health of male and female individuals.
Collapse
Affiliation(s)
- Laura Dearden
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Level 4, Box 289, Cambridge, CB2 0QQ, United Kingdom
| | - Sebastien G Bouret
- The Saban Research Institute, Developmental Neuroscience Program & Diabetes and Obesity Program, Center for Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, 90027, USA; Inserm, Jean-Pierre Aubert Research Center, U1172, University Lille 2, Lille, 59045, France
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Level 4, Box 289, Cambridge, CB2 0QQ, United Kingdom.
| |
Collapse
|
43
|
Maternal overnutrition programs epigenetic changes in the regulatory regions of hypothalamic Pomc in the offspring of rats. Int J Obes (Lond) 2018; 42:1431-1444. [PMID: 29777232 PMCID: PMC6113193 DOI: 10.1038/s41366-018-0094-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/27/2018] [Accepted: 03/16/2018] [Indexed: 02/02/2023]
Abstract
Background and objective Maternal overnutrition has been implicated in affecting the offspring by programming metabolic disorders such as obesity and diabetes, by mechanisms that are not clearly understood. This study aimed to determine the long-term impact of maternal high-fat (HF) diet feeding on epigenetic changes in the offspring’s hypothalamic Pomc gene, coding a key factor in the control of energy balance. Further, it aimed to study the additional effects of postnatal overnutrition on epigenetic programming by maternal nutrition. Methods Eight-week-old female Sprague–Dawley rats were fed HF diet or low-fat (LF) diet for 6 weeks before mating, and throughout gestation and lactation. At postnatal day 21, samples were collected from a third offspring and the remainder were weaned onto LF diet for 5 weeks, after which they were either fed LF or HF diet for 12 weeks, resulting in four groups of offspring differing by their maternal and postweaning diet. Results With maternal HF diet, offspring at weaning had rapid early weight gain, increased adiposity, and hyperleptinemia. The programmed adult offspring, subsequently fed LF diet, retained the increased body weight. Maternal HF diet combined with offspring HF diet caused more pronounced hyperphagia, fat mass, and insulin resistance. The ARC Pomc gene from programmed offspring at weaning showed hypermethylation in the enhancer (nPE1 and nPE2) regions and in the promoter sequence mediating leptin effects. Interestingly, hypermethylation at the Pomc promoter but not at the enhancer region persisted long term into adulthood in the programmed offspring. However, there were no additive effects on methylation levels in the regulatory regions of Pomc in programmed offspring fed a HF diet. Conclusion Maternal overnutrition programs long-term epigenetic alterations in the offspring’s hypothalamic Pomc promoter. This predisposes the offspring to metabolic disorders later in life.
Collapse
|
44
|
Shift-work: is time of eating determining metabolic health? Evidence from animal models. Proc Nutr Soc 2018; 77:199-215. [DOI: 10.1017/s0029665117004128] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The circadian disruption in shift-workers is suggested to be a risk factor to develop overweight and metabolic dysfunction. The conflicting time signals given by shifted activity, shifted food intake and exposure to light at night occurring in the shift-worker are proposed to be the cause for the loss of internal synchrony and the consequent adverse effects on body weight and metabolism. Because food elicited signals have proven to be potent entraining signals for peripheral oscillations, here we review the findings from experimental models of shift-work and verify whether they provide evidence about the causal association between shifted feeding schedules, circadian disruption and altered metabolism. We found mainly four experimental models that mimic the conditions of shift-work: protocols of forced sleep deprivation, of forced activity during the normal rest phase, exposure to light at night and shifted food timing. A big variability in the intensity and duration of the protocols was observed, which led to a diversity of effects. A common result was the disruption of temporal patterns of activity; however, not all studies explored the temporal patterns of food intake. According to studies that evaluate time of food intake as an experimental model of shift-work and studies that evaluate shifted food consumption, time of food intake may be a determining factor for the loss of balance at the circadian and metabolic level.
Collapse
|
45
|
Barrand S, Crowley TM, Wood-Bradley RJ, De Jong KA, Armitage JA. Impact of maternal high fat diet on hypothalamic transcriptome in neonatal Sprague Dawley rats. PLoS One 2017; 12:e0189492. [PMID: 29240779 PMCID: PMC5730210 DOI: 10.1371/journal.pone.0189492] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/27/2017] [Indexed: 12/14/2022] Open
Abstract
Maternal consumption of a high fat diet during early development has been shown to impact the formation of hypothalamic neurocircuitry, thereby contributing to imbalances in appetite and energy homeostasis and increasing the risk of obesity in subsequent generations. Early in postnatal life, the neuronal projections responsible for energy homeostasis develop in response to appetite-related peptides such as leptin. To date, no study characterises the genome-wide transcriptional changes that occur in response to exposure to high fat diet during this critical window. We explored the effects of maternal high fat diet consumption on hypothalamic gene expression in Sprague Dawley rat offspring at postnatal day 10. RNA-sequencing enabled discovery of differentially expressed genes between offspring of dams fed a high fat diet and offspring of control diet fed dams. Female high fat diet offspring displayed altered expression of 86 genes (adjusted P-value<0.05), including genes coding for proteins of the extra cellular matrix, particularly Collagen 1a1 (Col1a1), Col1a2, Col3a1, and the imprinted Insulin-like growth factor 2 (Igf2) gene. Male high fat diet offspring showed significant changes in collagen genes (Col1a1 and Col3a1) and significant upregulation of two genes involved in regulation of dopamine availability in the brain, tyrosine hydroxylase (Th) and dopamine reuptake transporter Slc6a3 (also known as Dat1). Transcriptional changes were accompanied by increased body weight, body fat and body length in the high fat diet offspring, as well as altered blood glucose and plasma leptin. Transcriptional changes identified in the hypothalamus of offspring of high fat diet mothers could alter neuronal projection formation during early development leading to abnormalities in the neuronal circuitry controlling appetite in later life, hence priming offspring to the development of obesity.
Collapse
Affiliation(s)
- Sanna Barrand
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Tamsyn M. Crowley
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
- MMR, BCRG, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Ryan J. Wood-Bradley
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Kirstie A. De Jong
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - James A. Armitage
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| |
Collapse
|
46
|
Ruegsegger GN, Grigsby KB, Kelty TJ, Zidon TM, Childs TE, Vieira-Potter VJ, Klinkebiel DL, Matheny M, Scarpace PJ, Booth FW. Maternal Western diet age-specifically alters female offspring voluntary physical activity and dopamine- and leptin-related gene expression. FASEB J 2017; 31:5371-5383. [PMID: 28794174 DOI: 10.1096/fj.201700389r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/25/2017] [Indexed: 01/30/2023]
Abstract
Prenatal overnutrition affects development into adulthood and influences risk of obesity. We assessed the transgenerational effect of maternal Western diet (WD) consumption on offspring physical activity. Voluntary wheel running was increased in juvenile (4-7 wk of age), but decreased in adult (16-19 wk of age), F1 female WD offspring In contrast, no wheel-running differences in F1 male offspring were observed. Increased wheel running in juvenile female WD offspring was associated with up-regulated dopamine receptor (DRD)-1 and -2 in the nucleus accumbens (NAc) and with down-regulated Lepr in the ventral tegmental area (VTA). Conversely, decreased wheel running by adult female WD offspring was associated with down-regulated DRD1 in the NAc and with up-regulated Lepr in the VTA. Body fat, leptin, and insulin were increased in male, but not in female, F1 WD offspring. Recombinant virus (rAAV) leptin antagonism in the VTA decreased wheel running in standard diet but not in WD F1 female offspring. Analysis of F2 offspring found no differences in wheel running or adiposity in male or female offspring, suggesting that changes in the F1 generation were related to in utero somatic reprogramming. Our findings indicate prenatal WD exposure leads to age-specific changes in voluntary physical activity in female offspring that are differentially influenced by VTA leptin antagonism.-Ruegsegger, G. N., Grigsby, K. B., Kelty, T. J., Zidon, T. M., Childs, T. E., Vieira-Potter, V. J., Klinkebiel, D. L., Matheny, M., Scarpace, P. J., Booth, F. W. Maternal Western diet age-specifically alters female offspring voluntary physical activity and dopamine- and leptin-related gene expression.
Collapse
Affiliation(s)
- Gregory N Ruegsegger
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Kolter B Grigsby
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Taylor J Kelty
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Terese M Zidon
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, USA
| | - Thomas E Childs
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Victoria J Vieira-Potter
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, USA
| | - David L Klinkebiel
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Michael Matheny
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, USA
| | - Phillip J Scarpace
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, USA
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA; .,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, USA.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
47
|
Rozo AV, Babu DA, Suen PA, Groff DN, Seeley RJ, Simmons RA, Seale P, Ahima RS, Stoffers DA. Neonatal GLP1R activation limits adult adiposity by durably altering hypothalamic architecture. Mol Metab 2017; 6:748-759. [PMID: 28702330 PMCID: PMC5485307 DOI: 10.1016/j.molmet.2017.05.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/03/2017] [Accepted: 05/10/2017] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Adult obesity risk is influenced by alterations to fetal and neonatal environments. Modifying neonatal gut or neurohormone signaling pathways can have negative metabolic consequences in adulthood. Here we characterize the effect of neonatal activation of glucagon like peptide-1 (GLP-1) receptor (GLP1R) signaling on adult adiposity and metabolism. METHODS Wild type C57BL/6 mice were injected with 1 nmol/kg Exendin-4 (Ex-4), a GLP1R agonist, for 6 consecutive days after birth. Growth, body composition, serum analysis, energy expenditure, food intake, and brain and fat pad histology and gene expression were assessed at multiple time points through 42 weeks. Similar analyses were conducted in a Glp1r conditional allele crossed with a Sim1Cre deleter strain to produce Sim1Cre;Glp1rloxP/loxP mice and control littermates. RESULTS Neonatal administration of Ex-4 reduced adult body weight and fat mass, increased energy expenditure, and conferred protection from diet-induced obesity in female mice. This was associated with induction of brown adipose genes and increased noradrenergic fiber density in parametrial white adipose tissue (WAT). We further observed durable alterations in orexigenic and anorexigenic projections to the paraventricular hypothalamic nucleus (PVH). Genetic deletion of Glp1r in the PVH by Sim1-Cre abrogated the impact of neonatal Ex-4 on adult body weight, WAT browning, and hypothalamic architecture. CONCLUSION These observations suggest that the acute activation of GLP1R in neonates durably alters hypothalamic architecture to limit adult weight gain and adiposity, identifying GLP1R as a therapeutic target for obesity prevention.
Collapse
Affiliation(s)
- Andrea V. Rozo
- Institute for Diabetes, Obesity and Metabolism and the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Daniella A. Babu
- Institute for Diabetes, Obesity and Metabolism and the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - PoMan A. Suen
- Institute for Diabetes, Obesity and Metabolism and the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - David N. Groff
- Institute for Diabetes, Obesity and Metabolism and the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Randy J. Seeley
- Department of Surgery, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Rebecca A. Simmons
- Department of Pediatrics, Division of Neonatology, The Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity and Metabolism and the Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Rexford S. Ahima
- Institute for Diabetes, Obesity and Metabolism and the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Doris A. Stoffers
- Institute for Diabetes, Obesity and Metabolism and the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| |
Collapse
|
48
|
Klein MO, MacKay H, Edwards A, Park S, Kiss ACI, Felicio LF, Abizaid A. POMC and NPY mRNA expression during development is increased in rat offspring brain from mothers fed with a high fat diet. Int J Dev Neurosci 2017; 64:14-20. [DOI: 10.1016/j.ijdevneu.2017.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/21/2017] [Accepted: 03/10/2017] [Indexed: 01/06/2023] Open
Affiliation(s)
- Marianne Orlandini Klein
- Department of NeuroscienceCarleton UniversityOttawaONCanada
- Department of PharmacologyInstitute of Biomedical Science, University of São PauloSão PauloSPBrazil
| | - Harry MacKay
- Department of NeuroscienceCarleton UniversityOttawaONCanada
| | | | - Su‐Bin Park
- Department of NeuroscienceCarleton UniversityOttawaONCanada
| | | | - Luciano Freitas Felicio
- Department of PharmacologyInstitute of Biomedical Science, University of São PauloSão PauloSPBrazil
- Department of PathologySchool of Veterinary Medicine, University of São PauloSão PauloSPBrazil
| | | |
Collapse
|
49
|
Robb JL, Messa I, Lui E, Yeung D, Thacker J, Satvat E, Mielke JG. A maternal diet high in saturated fat impairs offspring hippocampal function in a sex-specific manner. Behav Brain Res 2017; 326:187-199. [PMID: 28259676 DOI: 10.1016/j.bbr.2017.02.049] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/23/2017] [Accepted: 02/28/2017] [Indexed: 01/07/2023]
Abstract
While a maternal diet high in saturated fat is likely to affect foetal brain development, whether the effects are the same for male and female offspring is unclear. As a result, we randomly assigned female, Sprague-Dawley rats to either a control, or high-fat diet (HFD; 45% of calories from saturated fat) for 10 weeks. A range of biometrics were collected, and hippocampal function was assessed at both the tissue level (by measuring synaptic plasticity) and at the behavioural level (using the Morris water maze; MWM). Subsequently, a subset of animals was bred and remained on their respective diets throughout gestation and lactation. On post-natal day 21, offspring were weaned and placed onto the control diet; biometrics and spatial learning and memory were then assessed at both adolescence and young adulthood. Although the HFD led to changes in the maternal generation consistent with an obese phenotype, no impairments were noted at the level of hippocampal synaptic plasticity, or MWM performance. Unexpectedly, among the offspring, a sexually dimorphic effect upon MWM performance became apparent. In particular, adolescent male offspring displayed a greater latency to reach the platform during training trials and spent less time in the target quadrant during the probe test; notably, when re-examined during young adulthood, the performance deficit was no longer present. Overall, our work suggests the existence of sexual dimorphism with regard to how a maternal HFD affects hippocampal-dependent function in the offspring brain.
Collapse
Affiliation(s)
- Jamie-Lee Robb
- Neuroplasticity Research Group, School of Public Health and Health Systems, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
| | - Isabelle Messa
- Neuroplasticity Research Group, School of Public Health and Health Systems, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
| | - Erika Lui
- Neuroplasticity Research Group, School of Public Health and Health Systems, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
| | - Derrick Yeung
- Neuroplasticity Research Group, School of Public Health and Health Systems, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
| | - Jonathan Thacker
- Neuroplasticity Research Group, School of Public Health and Health Systems, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
| | - Elham Satvat
- Neuroplasticity Research Group, School of Public Health and Health Systems, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
| | - John G Mielke
- Neuroplasticity Research Group, School of Public Health and Health Systems, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada.
| |
Collapse
|
50
|
Ramírez-López MT, Arco R, Decara J, Vázquez M, Noemí Blanco R, Alén F, Suárez J, Gómez de Heras R, Rodríguez de Fonseca F. Exposure to a Highly Caloric Palatable Diet during the Perinatal Period Affects the Expression of the Endogenous Cannabinoid System in the Brain, Liver and Adipose Tissue of Adult Rat Offspring. PLoS One 2016; 11:e0165432. [PMID: 27806128 PMCID: PMC5091916 DOI: 10.1371/journal.pone.0165432] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 10/11/2016] [Indexed: 12/27/2022] Open
Abstract
Recent studies have linked gestational exposure to highly caloric diets with a disrupted endogenous cannabinoid system (ECS). In the present study, we have extended these studies by analyzing the impact of the exposure to a palatable diet during gestation and lactation on a) the adult expression of endocannabinoid-related behaviors, b) the metabolic profile of adult offspring and c) the mRNA expression of the signaling machinery of the ECS in the hypothalamus, the liver and the adipose tissue of adult offspring of both sexes. Exposure to a palatable diet resulted in a) sex-dimorphic and perinatal diet specific feeding behaviors, including the differential response to the inhibitory effects of the cannabinoid receptor inverse agonist AM251, b) features of metabolic syndrome including increased adiposity, hyperleptinemia, hypertriglyceridemia and hypercholesterolemia and c) tissue and sex-specific changes in the expression of both CB1 and CB2 receptors and in that of the endocannabinoid-degrading enzymes FAAH and MAGL, being the adipose tissue the most affected organ analyzed. Since the effects were observed in adult animals that were weaned while consuming a normal diet, the present results indicate that the ECS is one of the targets of maternal programming of the offspring energy expenditure. These results clearly indicate that the maternal diet has long-term effects on the development of pups through multiple alterations of signaling homeostatic pathways that include the ECS. The potential relevance of these alterations for the current obesity epidemic is discussed.
Collapse
Affiliation(s)
- María Teresa Ramírez-López
- Departamento de Psicobiología, Facultad de Psicología, Universidad Complutense de Madrid, Campus de Somosaguas s/n, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Raquel Arco
- Instituto de Investigación Biomédica de Málaga (IBIMA), Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010, Málaga, Spain
| | - Juan Decara
- Instituto de Investigación Biomédica de Málaga (IBIMA), Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010, Málaga, Spain
| | - Mariam Vázquez
- Instituto de Investigación Biomédica de Málaga (IBIMA), Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010, Málaga, Spain
| | - Rosario Noemí Blanco
- Departamento de Psicobiología, Facultad de Psicología, Universidad Complutense de Madrid, Campus de Somosaguas s/n, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Francisco Alén
- Instituto de Investigación Biomédica de Málaga (IBIMA), Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010, Málaga, Spain
| | - Juan Suárez
- Instituto de Investigación Biomédica de Málaga (IBIMA), Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010, Málaga, Spain
| | - Raquel Gómez de Heras
- Departamento de Psicobiología, Facultad de Psicología, Universidad Complutense de Madrid, Campus de Somosaguas s/n, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga (IBIMA), Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010, Málaga, Spain
| |
Collapse
|