1
|
DeLacey S, Gurra M, Arzu J, Lowe LP, Lowe W, Scholtens D, Josefson JL. Leptin and adiposity measures from birth to later childhood: Findings from the Hyperglycemia and Adverse Pregnancy Outcomes Follow-Up Study. Pediatr Obes 2024; 19:e13087. [PMID: 38095062 PMCID: PMC10921990 DOI: 10.1111/ijpo.13087] [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: 06/19/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Cord blood (CB) leptin is positively associated with adiposity at birth, but the association with child adiposity is unclear. OBJECTIVES We hypothesized that CB leptin is positively associated with adiposity in peripubertal children and with childhood leptin. METHODS Leptin was measured in 986 CB and 931 childhood stored samples from a prospective birth cohort. Adiposity measures were collected at birth and mean age 11.5 years. Linear and logistic regression analyses were used to evaluate associations between log-transformed CB leptin and neonatal and childhood adiposity measures as continuous and categorical variables, respectively. RESULTS CB leptin was positively associated with neonatal and childhood adiposity. Childhood associations were attenuated when adjusted for maternal body mass index (BMI) and glucose, but remained statistically significant for childhood body fat percentage (β = 1.15%, confidence interval [CI] = 0.46-1.84), body fat mass (β = 0.69 kg, 95% CI = 0.16-1.23), sum of skin-folds (β = 1.77 mm, 95% CI = 0.31-3.24), log-transformed child serum leptin (β = 0.13, 95% CI = 0.06-0.20), overweight/obesity (OR = 1.21, 95% CI = 1.03-1.42), obesity (OR = 1.31, 95% CI = 1.04-1.66) and body fat percentage >85th percentile (OR = 1.38, 95% CI = 1.12-1.73). Positive associations between newborn adiposity measures and CB leptin confirmed previous reports. CONCLUSION CB leptin is positively associated with neonatal and childhood adiposity and child leptin levels, independent of maternal BMI and maternal hyperglycemia. CB leptin may be a biomarker of future adiposity risk.
Collapse
Affiliation(s)
- Sean DeLacey
- Department of Pediatrics, Division of Endocrinology, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
- Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Miranda Gurra
- Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jennifer Arzu
- Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Lynn P. Lowe
- Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - William Lowe
- Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Denise Scholtens
- Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jami L. Josefson
- Department of Pediatrics, Division of Endocrinology, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
- Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
2
|
Urbonaite G, Knyzeliene A, Bunn FS, Smalskys A, Neniskyte U. The impact of maternal high-fat diet on offspring neurodevelopment. Front Neurosci 2022; 16:909762. [PMID: 35937892 PMCID: PMC9354026 DOI: 10.3389/fnins.2022.909762] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/27/2022] [Indexed: 12/11/2022] Open
Abstract
A maternal high-fat diet affects offspring neurodevelopment with long-term consequences on their brain health and behavior. During the past three decades, obesity has rapidly increased in the whole human population worldwide, including women of reproductive age. It is known that maternal obesity caused by a high-fat diet may lead to neurodevelopmental disorders in their offspring, such as autism spectrum disorder, attention deficit hyperactivity disorder, anxiety, depression, and schizophrenia. A maternal high-fat diet can affect offspring neurodevelopment due to inflammatory activation of the maternal gut, adipose tissue, and placenta, mirrored by increased levels of pro-inflammatory cytokines in both maternal and fetal circulation. Furthermore, a maternal high fat diet causes gut microbial dysbiosis further contributing to increased inflammatory milieu during pregnancy and lactation, thus disturbing both prenatal and postnatal neurodevelopment of the offspring. In addition, global molecular and cellular changes in the offspring's brain may occur due to epigenetic modifications including the downregulation of brain-derived neurotrophic factor (BDNF) expression and the activation of the endocannabinoid system. These neurodevelopmental aberrations are reflected in behavioral deficits observed in animals, corresponding to behavioral phenotypes of certain neurodevelopmental disorders in humans. Here we reviewed recent findings from rodent models and from human studies to reveal potential mechanisms by which a maternal high-fat diet interferes with the neurodevelopment of the offspring.
Collapse
Affiliation(s)
- Gintare Urbonaite
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Agne Knyzeliene
- Centre for Cardiovascular Science, The Queen’s Medical Research Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - Fanny Sophia Bunn
- Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
| | - Adomas Smalskys
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Urte Neniskyte
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- VU LSC-EMBL Partnership for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
3
|
Meireles ALF, Segabinazi E, Spindler C, Gasperini NF, Souza Dos Santos A, Pochmann D, Elsner VR, Marcuzzo S. Maternal resistance exercise promotes changes in neuroplastic and epigenetic marks of offspring's hippocampus during adult life. Physiol Behav 2020; 230:113306. [PMID: 33359430 DOI: 10.1016/j.physbeh.2020.113306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/19/2020] [Accepted: 12/22/2020] [Indexed: 12/01/2022]
Abstract
Studies indicate that gestational exercise practice positively impacts the offspring's cognition. Nevertheless, the influence of maternal resistance exercise, different periods of exercise practice, and the inter- and transgenerational effects involved in these responses are not known. This study sought to report the influence of the maternal practice of resistance exercise on offspring's cognitive function, exploring behavior, and neuroplastic and epigenetic marks in the hippocampus. Female Wistar rats were divided into four groups: sedentary (SS), exercised during pregnancy (SE), exercised before pregnancy (ES), and exercised before and during pregnancy (EE). Exercised rats were submitted to a resistance exercise protocol (vertical ladder climbing). Between postnatal days (P)81 and P85, male offspring were submitted to the Morris water maze test. At P85, the following analyses were performed in offspring's hippocampus: expression of IGF-1 and BrdU+ cells, global DNA methylation, H3/H4 acetylation, and HDAC2 amount. Only the offspring of SE mothers presented subtly better performance on learning and memory tasks, associated with lower HDAC2 amount. Offspring from ES mothers presented an overexpression of hippocampal neuroplastic marks (BrdU+ and IGF-1), as well as a decrease of DNA methylation and an increase in H4 acetylation. Offspring from EE mothers (continuously exercised) did not present modifications in plasticity or epigenetic parameters. This is the first study to observe the influence of maternal resistance exercise on offspring's brains. The findings provide evidence that offspring's hippocampus plasticity is influenced by exercise performed in isolated periods (pre- or gestationally) more than that performed continually.
Collapse
Affiliation(s)
- André Luís Ferreira Meireles
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Histofisiologia Comparada, Departamento de Ciências Morfológicas, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Ethiane Segabinazi
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Histofisiologia Comparada, Departamento de Ciências Morfológicas, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Christiano Spindler
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Histofisiologia Comparada, Departamento de Ciências Morfológicas, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Natália Felix Gasperini
- Laboratório de Histofisiologia Comparada, Departamento de Ciências Morfológicas, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Adriana Souza Dos Santos
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Histofisiologia Comparada, Departamento de Ciências Morfológicas, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Daniela Pochmann
- Programa de Pós-Graduação em Biociências e Reabilitação, Centro Universitário Metodista-IPA, Porto Alegre, RS, Brazil
| | - Viviane Rostirola Elsner
- Programa de Pós-Graduação em Biociências e Reabilitação, Centro Universitário Metodista-IPA, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Simone Marcuzzo
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Histofisiologia Comparada, Departamento de Ciências Morfológicas, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
4
|
Khambadkone SG, Cordner ZA, Tamashiro KLK. Maternal stressors and the developmental origins of neuropsychiatric risk. Front Neuroendocrinol 2020; 57:100834. [PMID: 32084515 PMCID: PMC7243665 DOI: 10.1016/j.yfrne.2020.100834] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 01/23/2020] [Accepted: 02/12/2020] [Indexed: 12/14/2022]
Abstract
The maternal environment during pregnancy is critical for fetal development and perinatal perturbations can prime offspring disease risk. Here, we briefly review evidence linking two well-characterized maternal stressors - psychosocial stress and infection - to increased neuropsychiatric risk in offspring. In the current climate of increasing obesity and globalization of the Western-style diet, maternal overnutrition emerges as a pressing public health concern. We focus our attention on recent epidemiological and animal model evidence showing that, like psychosocial stress and infection, maternal overnutrition can also increase offspring neuropsychiatric risk. Using lessons learned from the psychosocial stress and infection literature, we discuss how altered maternal and placental physiology in the setting of overnutrition may contribute to abnormal fetal development and resulting neuropsychiatric outcomes. A better understanding of converging pathophysiological pathways shared between stressors may enable development of interventions against neuropsychiatric illnesses that may be beneficial across stressors.
Collapse
Affiliation(s)
- Seva G Khambadkone
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zachary A Cordner
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kellie L K Tamashiro
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
5
|
Reynolds CM, Vickers MH. The role of adipokines in developmental programming: evidence from animal models. J Endocrinol 2019. [DOI: 10.1530/joe-18-0686] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Alterations in the environment during critical periods of development, including altered maternal nutrition, can increase the risk for the development of a range of metabolic, cardiovascular and reproductive disorders in offspring in adult life. Following the original epidemiological observations of David Barker that linked perturbed fetal growth to adult disease, a wide range of experimental animal models have provided empirical support for the developmental programming hypothesis. Although the mechanisms remain poorly defined, adipose tissue has been highlighted as playing a key role in the development of many disorders that manifest in later life. In particular, adipokines, including leptin and adiponectin, primarily secreted by adipose tissue, have now been shown to be important mediators of processes underpinning several phenotypic features associated with developmental programming including obesity, insulin sensitivity and reproductive disorders. Moreover, manipulation of adipokines in early life has provided for potential strategies to ameliorate or reverse the adverse sequalae that are associated with aberrant programming and provided insight into some of the mechanisms involved in the development of chronic disease across the lifecourse.
Collapse
Affiliation(s)
- Clare M Reynolds
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Mark H Vickers
- Liggins Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
6
|
Christians JK, Lennie KI, Wild LK, Garcha R. Effects of high-fat diets on fetal growth in rodents: a systematic review. Reprod Biol Endocrinol 2019; 17:39. [PMID: 30992002 PMCID: PMC6469066 DOI: 10.1186/s12958-019-0482-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 04/09/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Maternal nutrition during pregnancy has life-long consequences for offspring. However, the effects of maternal overnutrition and/ or obesity on fetal growth remain poorly understood, e.g., it is not clear why birthweight is increased in some obese pregnancies but not in others. Maternal obesity is frequently studied using rodents on high-fat diets, but effects on fetal growth are inconsistent. The purpose of this review is to identify factors that contribute to reduced or increased fetal growth in rodent models of maternal overnutrition. METHODS We searched Web of Science and screened 2173 abstracts and 328 full texts for studies that fed mice or rats diets providing ~ 45% or ~ 60% calories from fat for 3 weeks or more prior to pregnancy. We identified 36 papers matching the search criteria that reported birthweight or fetal weight. RESULTS Studies that fed 45% fat diets to mice or 60% fat diets to rats generally did not show effects on fetal growth. Feeding a 45% fat diet to rats generally reduced birth and fetal weight. Feeding mice a 60% fat diet for 4-9 weeks prior to pregnancy tended to increase in fetal growth, whereas feeding this diet for a longer period tended to reduce fetal growth. CONCLUSIONS The high-fat diets used most often with rodents do not closely match Western diets and frequently reduce fetal growth, which is not a typical feature of obese human pregnancies. Adoption of standard protocols that more accurately mimic effects on fetal growth observed in obese human pregnancies will improve translational impact in this field.
Collapse
Affiliation(s)
- Julian K. Christians
- 0000 0004 1936 7494grid.61971.38Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6 Canada
| | - Kendra I. Lennie
- 0000 0004 1936 7494grid.61971.38Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6 Canada
| | - Lisa K. Wild
- 0000 0004 1936 7494grid.61971.38Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6 Canada
| | - Raajan Garcha
- 0000 0004 1936 7494grid.61971.38Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6 Canada
| |
Collapse
|
7
|
Niu X, Wu X, Ying A, Shao B, Li X, Zhang W, Lin C, Lin Y. Maternal high fat diet programs hypothalamic-pituitary-adrenal function in adult rat offspring. Psychoneuroendocrinology 2019; 102:128-138. [PMID: 30544004 DOI: 10.1016/j.psyneuen.2018.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/29/2018] [Accepted: 12/04/2018] [Indexed: 02/04/2023]
Abstract
Maternal environmental factors such as diet have profound effects on offspring development and later health. The hypothalamic-pituitary-adrenal (HPA) axis is an important stress neuroendocrine system that is subject to programming by early life challenges. The present study was further to investigate whether maternal high fat diet (HFD) exposure during rat pregnancy and lactation can alter the HPA axis activity in adult male offspring. We observed that maternal HFD consumption exerted long-term effects on the basal activity of the HPA axis in adult offspring, with increased mean plasma corticosterone levels that result from elevated steroid pulse frequence and pulse amplitude. More importantly, maternal HFD offspring displayed enhanced corticosterone responses to restraint (1 h) and lipopolysaccharide (25 μg/kg, iv) but not insulin-induced hypoglycemia (0.3U/kg, iv) stress, suggesting a stressor-specific effect of maternal diet on the hyperresponsiveness of the HPA axis to stress. Additionally, maternal HFD exposure markedly attenuated the habituation of HPA responses to repeated restraint stress. These findings demonstrate that perinatal HFD exposure has a potent and long-lasting influence on development of neuroendocrine regulatory mechanisms. Maternal HFD consumption significantly increased basal corticotropin-releasing factor (CRF) mRNA expression in the paraventricular nucleus; nevertheless, similar increments in CRF mRNA levels following restraint were observed between maternal HFD offspring and control rats. Furthermore, the medial and central nuclei of amygdala played a pivotal role in maternal HFD-induced sensitization of the HPA response to psychological and systemic stress, respectively, suggesting that different neural pathways may mediate maternal HFD-induced HPA hyperresponsivity to different types of stressors. Take together, the long-term effects of maternal HFD challenge on the central regulation of the HPA axis, therefore, expose the adult offspring to greater HPA function throughout lifespan, in stressor-specific and region-specific manner.
Collapse
Affiliation(s)
- XiaoTing Niu
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - XiaoYun Wu
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - AnNa Ying
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Bei Shao
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - XiaoFeng Li
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - WanLi Zhang
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - ChengCheng Lin
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - YuanShao Lin
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
8
|
Oxidative Stress, Maternal Diabetes, and Autism Spectrum Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3717215. [PMID: 30524654 PMCID: PMC6247386 DOI: 10.1155/2018/3717215] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/17/2018] [Indexed: 12/25/2022]
Abstract
Autism spectrum disorders (ASD) are a group of early-onset neurodevelopmental conditions characterized by alterations in brain connectivity with cascading effects on neuropsychological functions. To date, in the framework of an increasing interest about environmental conditions which could interact with genetic factors in ASD pathogenesis, many authors have stressed that changes in the intrauterine environment at different stages of pregnancy, such as those linked to maternal metabolic pathologies, may lead to long-term conditions in the newborn. In particular, a growing number of epidemiological studies have highlighted the role of obesity and maternal diabetes as a risk factor for developing both somatic and psychiatric disorders in humans, including ASD. While literature still fails in identifying specific etiopathological mechanisms, a growing body of evidence is available about the presence of a relationship between maternal immune dysregulation, inflammation, oxidative stress, and the development of ASD in the offspring. In this framework, results from high-fat diet animal models about the role played by oxidative stress in shaping offspring neurodevelopment may help in clarifying the pathways through which maternal metabolic conditions are linked with ASD. The aim of this review is to provide an overview of literature about the effects of early life insults linked to oxidative stress which may be involved in ASD etiopathogenesis and how this relationship can be explained in biological terms.
Collapse
|
9
|
Gautier Y, Luneau I, Coquery N, Meurice P, Malbert CH, Guerin S, Kemp B, Bolhuis JE, Clouard C, Le Huërou-Luron I, Blat S, Val-Laillet D. Maternal Western diet during gestation and lactation modifies adult offspring's cognitive and hedonic brain processes, behavior, and metabolism in Yucatan minipigs. FASEB J 2018; 32:fj201701541. [PMID: 29897815 DOI: 10.1096/fj.201701541] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This study explores the long-term effects of exposure to a maternal Western diet (WD) vs. standard diet (SD) in the Yucatan minipig, on the adult progeny at lean status ( n = 32), and then overweight status. We investigated eating behavior, cognitive abilities, brain basal glucose metabolism, dopamine transporter availability, microbiota activity, blood lipids, and glucose tolerance. Although both groups demonstrated similar cognitive abilities in a holeboard test, WD pigs expressed a higher stress level than did SD pigs (immobility, P < 0.05) and lower performance in an alley maze ( P = 0.06). WD pigs demonstrated lower dopamine transporter binding potential in the hippocampus and parahippocampal cortex ( P < 0.05 for both), as well as a trend in putamen ( P = 0.07), associated with lower basal brain activity in the prefrontal cortex and nucleus accumbens ( P < 0.05) compared with lean SD pigs. Lean WD pigs demonstrated a lower glucose tolerance than did SD animals (higher glucose peak, P < 0.05) and a tendency to a higher incremental area under the curve of insulin from 0 to 30 minutes after intravenous glucose injection ( P < 0.1). Both groups developed glucose intolerance with overweight, but WD animals were less impacted than SD animals. These results demonstrate that maternal diet shaped the offspring's brain functions and cognitive responses long term, even after being fed a balanced diet from weaning, but behavioral effects were only revealed in WD pigs under anxiogenic situation; however, WD animals seemed to cope better with the obesogenic diet from a metabolic standpoint.-Gautier, Y., Luneau, I., Coquery, N., Meurice, P., Malbert, C.-H., Guerin, S., Kemp, B., Bolhuis, J. E., Clouard, C., Le Huërou-Luron, I., Blat, S., Val-Laillet, D. Maternal Western diet during gestation and lactation modifies adult offspring's cognitive and hedonic brain processes, behavior, and metabolism in Yucatan minipigs.
Collapse
Affiliation(s)
- Yentl Gautier
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| | - Isabelle Luneau
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| | - Nicolas Coquery
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| | - Paul Meurice
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| | | | - Sylvie Guerin
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| | - Bas Kemp
- Department of Animal Sciences, Wageningen University and Research, Adaptation Physiology Group, Wageningen, The Netherlands
| | - J Elizabeth Bolhuis
- Department of Animal Sciences, Wageningen University and Research, Adaptation Physiology Group, Wageningen, The Netherlands
| | - Caroline Clouard
- Department of Animal Sciences, Wageningen University and Research, Adaptation Physiology Group, Wageningen, The Netherlands
| | - Isabelle Le Huërou-Luron
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| | - Sophie Blat
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| | - David Val-Laillet
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| |
Collapse
|
10
|
Wolfrum C, Peleg-Raibstein D. Maternal overnutrition leads to cognitive and neurochemical abnormalities in C57BL/6 mice. Nutr Neurosci 2018; 22:688-699. [DOI: 10.1080/1028415x.2018.1432096] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Christian Wolfrum
- Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| | - Daria Peleg-Raibstein
- Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| |
Collapse
|
11
|
Long-term consequences of obesity on female fertility and the health of the offspring. Curr Opin Obstet Gynecol 2018; 29:180-187. [PMID: 28448277 DOI: 10.1097/gco.0000000000000364] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Obesity has reached near epidemic levels among reproductive age women with a myriad of consequences. Obesity adversely affects the maternal milieu by creating conditions that decrease fertility and increase the risk of gestational diabetes, hypertensive disease in pregnancy, fetal growth abnormalities and congenital anomalies. The effects of obesity are not limited to pregnancy. Indeed, beyond the immediate postpartum period, obese women maintain a higher prevalence of insulin resistance and cardiovascular disease. In this article, we will review the pathophysiology underlying the effects of obesity on fertility, pregnancy outcome and health status of offspring. The purpose of this review is to outline proposed models responsible for the short-term and long-term consequences of obesity on fertility and offspring development, and identify knowledge gaps where additional research is needed. RECENT FINDINGS Maternal over or under nutrition adversely affect maternal reproductive capacity and pregnancy success. Separate from effects on maternal reproductive function, maternal over or under nutrition may also 'program' fetal pathophysiology through inheritance mechanisms that suggest epigenetic modification of DNA, differential RNA translation and protein expression, or modification of the fetal hypothalamic-pituitary axis function through programmed adverse effects on the developing hypothalamic circuitry. The concept of maternal health modifying the risk of developing noncommunicable diseases in the offspring is based on Developmental Origins of Health and Disease hypothesis. SUMMARY Of importance, the long-term effects of obesity are not limited to maternal health, but also programs pathophysiology in their offspring. Children of obese gravida are at increased risk for the development of cardiometabolic disease in childhood and throughout adulthood. Future studies directly interrogating mechanisms underlying the risks associated with obesity will allow us to develop interventions and therapies to decrease short-term and long-term morbidities associated with maternal obesity.
Collapse
|
12
|
Janthakhin Y, Rincel M, Costa AM, Darnaudéry M, Ferreira G. Maternal high-fat diet leads to hippocampal and amygdala dendritic remodeling in adult male offspring. Psychoneuroendocrinology 2017; 83:49-57. [PMID: 28595087 DOI: 10.1016/j.psyneuen.2017.05.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/07/2017] [Accepted: 05/01/2017] [Indexed: 12/16/2022]
Abstract
Early-life exposure to calorie-dense food, rich in fat and sugar, contributes to the increasing prevalence of obesity and its associated adverse cognitive and emotional outcomes at adulthood. It is thus critical to determine the impact of such nutritional environment on neurobehavioral development. In animals, maternal high-fat diet (HFD) consumption impairs hippocampal function in adult offspring, but its impact on hippocampal neuronal morphology is unknown. Moreover, the consequences of perinatal HFD exposure on the amygdala, another important structure for emotional and cognitive processes, remain to be established. In rats, we show that adult offspring from dams fed with HFD (45% from fat, throughout gestation and lactation) exhibit atrophy of pyramidal neuron dendrites in both the CA1 of the hippocampus and the basolateral amygdala (BLA). Perinatal HFD exposure also impairs conditioned odor aversion, a task highly dependent on BLA function, without affecting olfactory or malaise processing. Neuronal morphology and behavioral alterations elicited by perinatal HFD are not associated with body weight changes but with higher plasma leptin levels at postnatal day 15 and at adulthood. Taken together, our results suggest that perinatal HFD exposure alters hippocampal and amygdala neuronal morphology which could participate to memory alterations at adulthood.
Collapse
Affiliation(s)
- Yoottana Janthakhin
- INRA, Nutrition and Integrative Neurobiology, UMR 1286, Bordeaux, France; Université de Bordeaux, Nutrition and Integrative Neurobiology, UMR 1286, Bordeaux, France
| | - Marion Rincel
- INRA, Nutrition and Integrative Neurobiology, UMR 1286, Bordeaux, France; Université de Bordeaux, Nutrition and Integrative Neurobiology, UMR 1286, Bordeaux, France
| | - Anna-Maria Costa
- INRA, Nutrition and Integrative Neurobiology, UMR 1286, Bordeaux, France; Université de Bordeaux, Nutrition and Integrative Neurobiology, UMR 1286, Bordeaux, France
| | - Muriel Darnaudéry
- INRA, Nutrition and Integrative Neurobiology, UMR 1286, Bordeaux, France; Université de Bordeaux, Nutrition and Integrative Neurobiology, UMR 1286, Bordeaux, France.
| | - Guillaume Ferreira
- INRA, Nutrition and Integrative Neurobiology, UMR 1286, Bordeaux, France; Université de Bordeaux, Nutrition and Integrative Neurobiology, UMR 1286, Bordeaux, France.
| |
Collapse
|
13
|
Tellechea ML, Mensegue MF, Pirola CJ. The Association between High Fat Diet around Gestation and Metabolic Syndrome-related Phenotypes in Rats: A Systematic Review and Meta-Analysis. Sci Rep 2017; 7:5086. [PMID: 28698653 PMCID: PMC5506021 DOI: 10.1038/s41598-017-05344-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 05/16/2017] [Indexed: 12/12/2022] Open
Abstract
Numerous rodent studies have evaluated the effects of a maternal high-fat diet (HFD) on later in life susceptibility to Metabolic Syndrome (MetS) with varying results. Our aim was to quantitatively synthesize the available data on effects of maternal HFD around gestation on offspring's body mass, body fat, plasma leptin, glucose, insulin, lipids and systolic blood pressure (SBP). Literature was screened and summary estimates of the effect of maternal HFD on outcomes were calculated by using fixed- or random-effects models. 362 effect sizes from 68 studies together with relevant moderators were collected. We found that maternal HFD is statistically associated with higher body fat, body weight, leptin, glucose, insulin and triglycerides levels, together with increased SBP in offspring later in life. Our analysis also revealed non-significant overall effect on offspring's HDL-cholesterol. A main source of variation among studies emerged from rat strain and lard-based diet type. Strain and sex -specific effects on particular data subsets were detected. Recommendations are suggested for future research in the field of developmental programming of the MetS. Despite significant heterogeneity, our meta-analysis confirms that maternal HFD had long-term metabolic effects in offspring.
Collapse
Affiliation(s)
- Mariana L Tellechea
- University of Buenos Aires, Institute of Medical Research A Lanari, Buenos Aires, Argentina. .,National Scientific and Technical Research Council (CONICET) - University of Buenos Aires, Institute of Medical Research (IDIM), Department of Molecular Genetics and Biology of Complex Diseases, Buenos Aires, Argentina.
| | - Melisa F Mensegue
- University of Buenos Aires, Institute of Medical Research A Lanari, Buenos Aires, Argentina.,National Scientific and Technical Research Council (CONICET) - University of Buenos Aires, Institute of Medical Research (IDIM), Department of Molecular Genetics and Biology of Complex Diseases, Buenos Aires, Argentina
| | - Carlos J Pirola
- University of Buenos Aires, Institute of Medical Research A Lanari, Buenos Aires, Argentina. .,National Scientific and Technical Research Council (CONICET) - University of Buenos Aires, Institute of Medical Research (IDIM), Department of Molecular Genetics and Biology of Complex Diseases, Buenos Aires, Argentina.
| |
Collapse
|
14
|
Moody L, Chen H, Pan YX. Early-Life Nutritional Programming of Cognition-The Fundamental Role of Epigenetic Mechanisms in Mediating the Relation between Early-Life Environment and Learning and Memory Process. Adv Nutr 2017; 8:337-350. [PMID: 28298276 PMCID: PMC5347110 DOI: 10.3945/an.116.014209] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The perinatal period is a window of heightened plasticity that lays the groundwork for future anatomic, physiologic, and behavioral outcomes. During this time, maternal diet plays a pivotal role in the maturation of vital organs and the establishment of neuronal connections. However, when perinatal nutrition is either lacking in specific micro- and macronutrients or overloaded with excess calories, the consequences can be devastating and long lasting. The brain is particularly sensitive to perinatal insults, with several neurologic and psychiatric disorders having been linked to a poor in utero environment. Diseases characterized by learning and memory impairments, such as autism, schizophrenia, and Alzheimer disease, are hypothesized to be attributed in part to environmental factors, and evidence suggests that the etiology of these conditions may date back to very early life. In this review, we discuss the role of the early-life diet in shaping cognitive outcomes in offspring. We explore the endocrine and immune mechanisms responsible for these phenotypes and discuss how these systemic factors converge to change the brain's epigenetic landscape and regulate learning and memory across the lifespan. Through understanding the maternal programming of cognition, critical steps may be taken toward preventing and treating diseases that compromise learning and memory.
Collapse
Affiliation(s)
| | - Hong Chen
- Division of Nutritional Sciences,,Department of Food Science and Human Nutrition, and
| | - Yuan-Xiang Pan
- Division of Nutritional Sciences, .,Department of Food Science and Human Nutrition, and.,Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, IL
| |
Collapse
|
15
|
De Luca SN, Ziko I, Sominsky L, Nguyen JCD, Dinan T, Miller AA, Jenkins TA, Spencer SJ. Early life overfeeding impairs spatial memory performance by reducing microglial sensitivity to learning. J Neuroinflammation 2016; 13:112. [PMID: 27193330 PMCID: PMC4872342 DOI: 10.1186/s12974-016-0578-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 05/11/2016] [Indexed: 11/22/2022] Open
Abstract
Background Obesity can lead to cognitive dysfunction including poor performance in memory tasks. However, poor memory is not seen in all obese humans and takes several months to develop in animal models, indicating the adult brain is relatively resistant to obesity’s cognitive effects. We have seen that, in the rat, overfeeding for as little as 3 weeks in early life leads to lasting obesity and microglial priming in the hypothalamus. Here we hypothesized that microglial hyper-sensitivity in the neonatally overfed rats extends beyond the hypothalamus into memory-associated brain regions, resulting in cognitive deficits. Methods We tested this idea by manipulating Wistar rat litter sizes to suckle pups in litters of 4 (overfed) or 12 (control). Results Neonatally overfed rats had microgliosis in the hippocampus after only 14 days overfeeding, and this persisted into adulthood. These changes were coupled with poor performance in radial arm maze and novel object recognition tests relative to controls. In controls, the experience of the radial arm maze reduced cell proliferation in the dentate gyrus and neuron numbers in the CA3. The learning task also suppressed microglial number and density in hippocampus and retrosplenial cortex. Neonatally overfed brains had impaired sensitivity to learning, with no neuronal or cell proliferative effects and less effective microglial suppression. Conclusions Thus, early life overfeeding contributes to a long-term impairment in learning and memory with a likely role for microglia. These data may partially explain why some obese individuals display cognitive dysfunction and some do not, i.e. the early life dietary environment is likely to have a vital long-term contribution. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0578-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Simone N De Luca
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., 3083, Australia
| | - Ilvana Ziko
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., 3083, Australia
| | - Luba Sominsky
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., 3083, Australia
| | - Jason C D Nguyen
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., 3083, Australia
| | - Tara Dinan
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., 3083, Australia
| | - Alyson A Miller
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., 3083, Australia
| | - Trisha A Jenkins
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., 3083, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., 3083, Australia.
| |
Collapse
|
16
|
Cordner ZA, Tamashiro KLK. Effects of high-fat diet exposure on learning & memory. Physiol Behav 2015; 152:363-71. [PMID: 26066731 PMCID: PMC5729745 DOI: 10.1016/j.physbeh.2015.06.008] [Citation(s) in RCA: 201] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 05/14/2015] [Accepted: 06/05/2015] [Indexed: 01/13/2023]
Abstract
The associations between consumption of a high-fat or 'Western' diet and metabolic disorders such as obesity, diabetes, and cardiovascular disease have long been recognized and a great deal of evidence now suggests that diets high in fat can also have a profound impact on the brain, behavior, and cognition. Here, we will review the techniques most often used to assess learning and memory in rodent models and discuss findings from studies assessing the cognitive effects of high-fat diet consumption. The review will then consider potential underlying mechanisms in the brain and conclude by reviewing emerging literature suggesting that maternal consumption of a high-fat diet may have effects on the learning and memory of offspring.
Collapse
Affiliation(s)
- Zachary A Cordner
- Cellular & Molecular Medicine Graduate Program, Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Kellie L K Tamashiro
- Cellular & Molecular Medicine Graduate Program, Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States.
| |
Collapse
|
17
|
Penfold NC, Ozanne SE. Developmental programming by maternal obesity in 2015: Outcomes, mechanisms, and potential interventions. Horm Behav 2015; 76:143-52. [PMID: 26145566 DOI: 10.1016/j.yhbeh.2015.06.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 06/23/2015] [Accepted: 06/24/2015] [Indexed: 02/06/2023]
Abstract
This article is part of a Special Issue "SBN 2014". Obesity in women of child-bearing age is a growing problem in developed and developing countries. Evidence from human studies indicates that maternal BMI correlates with offspring adiposity from an early age and predisposes to metabolic disease in later life. Thus the early life environment is an attractive target for intervention to improve public health. Animal models have been used to investigate the specific physiological outcomes and mechanisms of developmental programming that result from exposure to maternal obesity in utero. From this research, targeted intervention strategies can be designed. In this review we summarise recent progress in this field, with a focus on cardiometabolic disease and central control of appetite and behaviour. We highlight key factors that may mediate programming by maternal obesity, including leptin, insulin, and ghrelin. Finally, we explore potential lifestyle and pharmacological interventions in humans and the current state of evidence from animal models.
Collapse
Affiliation(s)
- Naomi C Penfold
- University of Cambridge, Metabolic Research Laboratories MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom.
| | - Susan E Ozanne
- University of Cambridge, Metabolic Research Laboratories MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
18
|
Pollock KE, Stevens D, Pennington KA, Thaisrivongs R, Kaiser J, Ellersieck MR, Miller DK, Schulz LC. Hyperleptinemia During Pregnancy Decreases Adult Weight of Offspring and Is Associated With Increased Offspring Locomotor Activity in Mice. Endocrinology 2015. [PMID: 26196541 DOI: 10.1210/en.2015-1247] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Pregnant women who are obese or have gestational diabetes mellitus have elevated leptin levels and their children have an increased risk for child and adult obesity. The goals of this study were to determine whether offspring weights are altered by maternal hyperleptinemia, and whether this occurs via behavioral changes that influence energy balance. We used 2 hyperleptinemic mouse models. The first was females heterozygous for a leptin receptor mutation (DB/+), which were severely hyperleptinemic, and that were compared with wild-type females. The second model was wild-type females infused with leptin (LEP), which were moderately hyperleptinemic, and were compared with wild-type females infused with saline (SAL). Total food consumption, food preference, locomotor activity, coordinated motor skills, and anxiety-like behaviors were assessed in wild-type offspring from each maternal group at 3 postnatal ages: 4-6, 11-13, and 19-21 weeks. Half the offspring from each group were then placed on a high-fat diet, and behaviors were reassessed. Adult offspring from both groups of hyperleptinemic dams weighed less than their respective controls beginning at 23 weeks of age, independent of diet or sex. Weight differences were not explained by food consumption or preference, because female offspring from hyperleptinemic dams tended to consume more food and had reduced preference for palatable, high-fat and sugar, food compared with controls. Offspring from DB/+ dams were more active than offspring of controls, as were female offspring of LEP dams. Maternal hyperleptinemia during pregnancy did not predispose offspring to obesity, and in fact, reduced weight gain.
Collapse
Affiliation(s)
- Kelly E Pollock
- Department of Obstetrics, Gynecology, and Women's Health (K.E.P., K.A.P., L.C.S.), University of Missouri, Columbia, Missouri 65212; Department of Animal Sciences (K.E.P., M.R.E., L.C.S.), University of Missouri, Columbia, Missouri 65211; Department of Health Sciences (D.S.), University of Missouri, Columbia, Missouri 65212; School of Medicine (R.T.), University of Missouri, Columbia, Missouri 65212; Division of Biological Sciences (J.K., L.C.S.), University of Missouri, Columbia, Missouri 65211; and Department of Psychological Sciences (D.K.M.), University of Missouri, Columbia, Missouri 65211
| | - Damaiyah Stevens
- Department of Obstetrics, Gynecology, and Women's Health (K.E.P., K.A.P., L.C.S.), University of Missouri, Columbia, Missouri 65212; Department of Animal Sciences (K.E.P., M.R.E., L.C.S.), University of Missouri, Columbia, Missouri 65211; Department of Health Sciences (D.S.), University of Missouri, Columbia, Missouri 65212; School of Medicine (R.T.), University of Missouri, Columbia, Missouri 65212; Division of Biological Sciences (J.K., L.C.S.), University of Missouri, Columbia, Missouri 65211; and Department of Psychological Sciences (D.K.M.), University of Missouri, Columbia, Missouri 65211
| | - Kathleen A Pennington
- Department of Obstetrics, Gynecology, and Women's Health (K.E.P., K.A.P., L.C.S.), University of Missouri, Columbia, Missouri 65212; Department of Animal Sciences (K.E.P., M.R.E., L.C.S.), University of Missouri, Columbia, Missouri 65211; Department of Health Sciences (D.S.), University of Missouri, Columbia, Missouri 65212; School of Medicine (R.T.), University of Missouri, Columbia, Missouri 65212; Division of Biological Sciences (J.K., L.C.S.), University of Missouri, Columbia, Missouri 65211; and Department of Psychological Sciences (D.K.M.), University of Missouri, Columbia, Missouri 65211
| | - Rose Thaisrivongs
- Department of Obstetrics, Gynecology, and Women's Health (K.E.P., K.A.P., L.C.S.), University of Missouri, Columbia, Missouri 65212; Department of Animal Sciences (K.E.P., M.R.E., L.C.S.), University of Missouri, Columbia, Missouri 65211; Department of Health Sciences (D.S.), University of Missouri, Columbia, Missouri 65212; School of Medicine (R.T.), University of Missouri, Columbia, Missouri 65212; Division of Biological Sciences (J.K., L.C.S.), University of Missouri, Columbia, Missouri 65211; and Department of Psychological Sciences (D.K.M.), University of Missouri, Columbia, Missouri 65211
| | - Jennifer Kaiser
- Department of Obstetrics, Gynecology, and Women's Health (K.E.P., K.A.P., L.C.S.), University of Missouri, Columbia, Missouri 65212; Department of Animal Sciences (K.E.P., M.R.E., L.C.S.), University of Missouri, Columbia, Missouri 65211; Department of Health Sciences (D.S.), University of Missouri, Columbia, Missouri 65212; School of Medicine (R.T.), University of Missouri, Columbia, Missouri 65212; Division of Biological Sciences (J.K., L.C.S.), University of Missouri, Columbia, Missouri 65211; and Department of Psychological Sciences (D.K.M.), University of Missouri, Columbia, Missouri 65211
| | - Mark R Ellersieck
- Department of Obstetrics, Gynecology, and Women's Health (K.E.P., K.A.P., L.C.S.), University of Missouri, Columbia, Missouri 65212; Department of Animal Sciences (K.E.P., M.R.E., L.C.S.), University of Missouri, Columbia, Missouri 65211; Department of Health Sciences (D.S.), University of Missouri, Columbia, Missouri 65212; School of Medicine (R.T.), University of Missouri, Columbia, Missouri 65212; Division of Biological Sciences (J.K., L.C.S.), University of Missouri, Columbia, Missouri 65211; and Department of Psychological Sciences (D.K.M.), University of Missouri, Columbia, Missouri 65211
| | - Dennis K Miller
- Department of Obstetrics, Gynecology, and Women's Health (K.E.P., K.A.P., L.C.S.), University of Missouri, Columbia, Missouri 65212; Department of Animal Sciences (K.E.P., M.R.E., L.C.S.), University of Missouri, Columbia, Missouri 65211; Department of Health Sciences (D.S.), University of Missouri, Columbia, Missouri 65212; School of Medicine (R.T.), University of Missouri, Columbia, Missouri 65212; Division of Biological Sciences (J.K., L.C.S.), University of Missouri, Columbia, Missouri 65211; and Department of Psychological Sciences (D.K.M.), University of Missouri, Columbia, Missouri 65211
| | - Laura Clamon Schulz
- Department of Obstetrics, Gynecology, and Women's Health (K.E.P., K.A.P., L.C.S.), University of Missouri, Columbia, Missouri 65212; Department of Animal Sciences (K.E.P., M.R.E., L.C.S.), University of Missouri, Columbia, Missouri 65211; Department of Health Sciences (D.S.), University of Missouri, Columbia, Missouri 65212; School of Medicine (R.T.), University of Missouri, Columbia, Missouri 65212; Division of Biological Sciences (J.K., L.C.S.), University of Missouri, Columbia, Missouri 65211; and Department of Psychological Sciences (D.K.M.), University of Missouri, Columbia, Missouri 65211
| |
Collapse
|
19
|
Reynolds CM, Gray C, Li M, Segovia SA, Vickers MH. Early Life Nutrition and Energy Balance Disorders in Offspring in Later Life. Nutrients 2015; 7:8090-111. [PMID: 26402696 PMCID: PMC4586579 DOI: 10.3390/nu7095384] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 08/31/2015] [Accepted: 09/11/2015] [Indexed: 02/07/2023] Open
Abstract
The global pandemic of obesity and type 2 diabetes is often causally linked to changes in diet and lifestyle; namely increased intake of calorically dense foods and concomitant reductions in physical activity. Epidemiological studies in humans and controlled animal intervention studies have now shown that nutritional programming in early periods of life is a phenomenon that affects metabolic and physiological functions throughout life. This link is conceptualised as the developmental programming hypothesis whereby environmental influences during critical periods of developmental plasticity can elicit lifelong effects on the health and well-being of the offspring. The mechanisms by which early environmental insults can have long-term effects on offspring remain poorly defined. However there is evidence from intervention studies which indicate altered wiring of the hypothalamic circuits that regulate energy balance and epigenetic effects including altered DNA methylation of key adipokines including leptin. Studies that elucidate the mechanisms behind these associations will have a positive impact on the health of future populations and adopting a life course perspective will allow identification of phenotype and markers of risk earlier, with the possibility of nutritional and other lifestyle interventions that have obvious implications for prevention of non-communicable diseases.
Collapse
Affiliation(s)
- Clare M Reynolds
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, Auckland 1142, New Zealand.
| | - Clint Gray
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, Auckland 1142, New Zealand.
| | - Minglan Li
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, Auckland 1142, New Zealand.
| | - Stephanie A Segovia
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, Auckland 1142, New Zealand.
| | - Mark H Vickers
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, Auckland 1142, New Zealand.
| |
Collapse
|
20
|
Little appetite for obesity: meta-analysis of the effects of maternal obesogenic diets on offspring food intake and body mass in rodents. Int J Obes (Lond) 2015; 39:1669-78. [PMID: 26293233 DOI: 10.1038/ijo.2015.160] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 07/15/2015] [Accepted: 08/02/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND There is increasing recognition that maternal effects contribute to variation in individual food intake and metabolism. For example, many experimental studies on model animals have reported the effect of a maternal obesogenic diet during pregnancy on the appetite of offspring. However, the consistency of effects and the causes of variation among studies remain poorly understood. METHODS After a systematic search for relevant publications, we selected 53 studies on rats and mice for a meta-analysis. We extracted and analysed data on the differences in food intake and body weight between offspring of dams fed obesogenic diets and dams fed standard diets during gestation. We used meta-regression to study predictors of the strength and direction of the effect sizes. RESULTS We found that experimental offspring tended to eat more than control offspring but this difference was small and not statistically significant (0.198, 95% highest posterior density (HPD)=-0.118-0.627). However, offspring from dams on obesogenic diets were significantly heavier than offspring of control dams (0.591, 95% HPD=0.052-1.056). Meta-regression analysis revealed no significant influences of tested predictor variables (for example, use of choice vs no-choice maternal diet, offspring sex) on differences in offspring appetite. Dietary manipulations that extended into lactation had the largest effect on body weight. Subgroup analysis revealed that high protein to non-protein ratio of the maternal diet may promote increased body weight in experimental offspring in comparison with control offspring; low protein content in the maternal chow can have opposite effect. CONCLUSIONS Exposure to maternal obesogenic diets in early life is not likely to result in a substantial change in offspring appetite. Nevertheless, we found an effect on offspring body weight, consistent with permanent alterations of offspring metabolism in response to maternal diet. Additionally, it appears that protein content of the obesogenic diet and timing of manipulation modulate the effects on offspring body weight in later life.
Collapse
|
21
|
López-Gallardo M, Antón-Fernández A, Llorente R, Mela V, Llorente-Berzal A, Prada C, Viveros MP. Neonatal Treatment with a Pegylated Leptin Antagonist Induces Sexually Dimorphic Effects on Neurones and Glial Cells, and on Markers of Synaptic Plasticity in the Developing Rat Hippocampal Formation. J Neuroendocrinol 2015; 27:658-69. [PMID: 25981175 DOI: 10.1111/jne.12294] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 04/04/2015] [Accepted: 04/11/2015] [Indexed: 01/03/2023]
Abstract
The present study aimed to better understand the role of the neonatal leptin surge, which peaks on postnatal day (PND)9-10, on the development of the hippocampal formation. Accordingly, male and female rats were administered with a pegylated leptin antagonist on PND9 and the expression of neurones, glial cells and diverse markers of synaptic plasticity was then analysed by immunohistochemistry in the hippocampal formation. Antagonism of the actions of leptin at this specific postnatal stage altered the number of glial fibrillary acidic protein positive cells, and also affected type 1 cannabinoid receptors, synaptophysin and brain-derived neurotrophic factor (BDNF), with the latter effect being sexually dimorphic. The results indicate that the physiological leptin surge occurring around PND 9-10 is critical for hippocampal formation development and that the dynamics of leptin activity might be different in males and females. The data obtained also suggest that some but not all the previously reported effects of maternal deprivation on hippocampal formation development (which markedly reduces leptin levels at PND 9-10) might be mediated by leptin deficiency in these animals.
Collapse
Affiliation(s)
- M López-Gallardo
- Department of Physiology, Faculty of Medicine, Universidad Complutense, Madrid, Spain
| | - A Antón-Fernández
- Department of Physiology, Faculty of Medicine, Universidad Complutense, Madrid, Spain
| | - R Llorente
- Department of Physiology (Animal Physiology II), Faculty of Biology, Universidad Complutense, Madrid, Spain
| | - V Mela
- Department of Physiology (Animal Physiology II), Faculty of Biology, Universidad Complutense, Madrid, Spain
| | - A Llorente-Berzal
- Department of Physiology (Animal Physiology II), Faculty of Biology, Universidad Complutense, Madrid, Spain
| | - C Prada
- Department of Physiology, Faculty of Medicine, Universidad Complutense, Madrid, Spain
| | - M P Viveros
- Department of Physiology (Animal Physiology II), Faculty of Biology, Universidad Complutense, Madrid, Spain
| |
Collapse
|
22
|
Kuhn FT, Trevizol F, Dias VT, Barcelos RCS, Pase CS, Roversi K, Antoniazzi CTDD, Roversi K, Boufleur N, Benvegnú DM, Emanuelli T, Bürger ME. Toxicological aspects of trans fat consumption over two sequential generations of rats: Oxidative damage and preference for amphetamine. Toxicol Lett 2014; 232:58-67. [PMID: 25290576 DOI: 10.1016/j.toxlet.2014.10.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 09/24/2014] [Accepted: 10/03/2014] [Indexed: 11/25/2022]
Abstract
Chronic consumption of processed food causes structural changes in membrane phospholipids, affecting brain neurotransmission. Here we evaluated noxious influences of dietary fats over two generations of rats on amphetamine (AMPH)-conditioned place preference (CPP). Female rats received soybean oil (SO, rich in n-6 fatty acids (FA)), fish oil (FO, rich in n-3 FA) and hydrogenated vegetable fat (HVF, rich in trans fatty acids (TFA)) for two successive generations. Male pups from the 2nd generation were maintained on the same supplementation until 41 days of age, when they were conditioned with AMPH in CPP. While the FO group showed higher incorporation of n-3 polyunsaturated-FA (PUFA) in cortex/hippocampus, the HVF group showed TFA incorporation in these same brain areas. The SO and HVF groups showed AMPH-preference and anxiety-like symptoms during abstinence. Higher levels of protein carbonyl (PC) and lower levels of non-protein thiols (NPSH) were observed in cortex/hippocampus of the HVF group, indicating antioxidant defense system impairment. In contrast, the FO group showed no drug-preference and lower PC levels in cortex. Cortical PC was positively correlated with n-6/n-3 PUFA ratio, locomotion and anxiety-like behavior, and hippocampal PC was positively correlated with AMPH-preference, reinforcing connections between oxidative damage and AMPH-induced preference/abstinence behaviors. As brain incorporation of trans and n-6 PUFA modifies its physiological functions, it may facilitate drug addiction.
Collapse
Affiliation(s)
- Fábio Teixeira Kuhn
- Programa de pós Graduação em Farmacologia, Universidade Federal de Santa Maria-UFSM, RS, Brazil
| | - Fabíola Trevizol
- Programa de pós Graduação em Farmacologia, Universidade Federal de Santa Maria-UFSM, RS, Brazil
| | - Verônica Tironi Dias
- Programa de pós Graduação em Farmacologia, Universidade Federal de Santa Maria-UFSM, RS, Brazil
| | | | - Camila Simonetti Pase
- Programa de pós Graduação em Farmacologia, Universidade Federal de Santa Maria-UFSM, RS, Brazil
| | - Karine Roversi
- Departamento de Fisiologia e Farmacologia, UFSM, RS, Brazil
| | | | | | - Nardeli Boufleur
- Programa de pós Graduação em Farmacologia, Universidade Federal de Santa Maria-UFSM, RS, Brazil
| | - Dalila Moter Benvegnú
- Programa de pós Graduação em Farmacologia, Universidade Federal de Santa Maria-UFSM, RS, Brazil
| | - Tatiana Emanuelli
- Programa de pós Graduação em Farmacologia, Universidade Federal de Santa Maria-UFSM, RS, Brazil; Departamento de Ciência e Tecnologia dos Alimentos, UFSM, RS, Brazil
| | - Marilise Escobar Bürger
- Programa de pós Graduação em Farmacologia, Universidade Federal de Santa Maria-UFSM, RS, Brazil; Departamento de Fisiologia e Farmacologia, UFSM, RS, Brazil.
| |
Collapse
|
23
|
Kang SS, Kurti A, Fair DA, Fryer JD. Dietary intervention rescues maternal obesity induced behavior deficits and neuroinflammation in offspring. J Neuroinflammation 2014; 11:156. [PMID: 25212412 PMCID: PMC4172780 DOI: 10.1186/s12974-014-0156-9] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 08/21/2014] [Indexed: 12/27/2022] Open
Abstract
Obesity induces a low-grade inflammatory state and has been associated with behavioral and cognitive alterations. Importantly, maternal environmental insults can adversely impact subsequent offspring behavior and have been linked with neurodevelopmental disorders such as autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (AHDH). It is unknown if maternal obesity significantly alters offspring sociability, a key ASD feature, and if altering maternal diet will provide an efficacious intervention paradigm for behavioral deficits. Here we investigated the impact of maternal high fat diet (HFD) and maternal dietary intervention during lactation on offspring behavior and brain inflammation in mice. We found that maternal HFD increased anxiety and decreased sociability in female offspring. Additionally, female offspring from HFD-fed dams also exhibited increased brain IL-1β and TNFα and microglial activation. Importantly, maternal dietary intervention during lactation was sufficient to alleviate social deficits and brain inflammation. Maternal obesity during gestation alone was sufficient to increase hyperactivity in male offspring, a phenotype that was not ameliorated by dietary intervention. These data suggest that maternal HFD acts as a prenatal/perinatal insult that significantly impacts offspring behavior and inflammation and that dietary intervention during lactation may be an easily translatable, efficacious intervention to offset some of these manifestations.
Collapse
|
24
|
Endocannabinoid signals in the developmental programming of delayed-onset neuropsychiatric and metabolic illnesses. Biochem Soc Trans 2014; 41:1569-76. [PMID: 24256256 DOI: 10.1042/bst20130117] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
It is increasingly recognized that maternal exposure to metabolic (nutritional) stimuli, infections, illicit or prescription drugs and environmental stressors during pregnancy can predispose affected offspring to developing devastating postnatal illnesses. If detrimental maternal stimuli coincide with critical periods of tissue production and organogenesis then they can permanently derail key cellular differentiation programs. Maternal programming can thus either provoke developmental failure directly ('direct hit') or introduce latent developmental errors that enable otherwise sub-threshold secondary stressors to manifest as disease ('double hit') postnatally. Accumulating evidence suggests that nervous system development is tightly controlled by maternal metabolic stimuli, and whose synaptic wiring and integrative capacity are adversely affected by dietary and hormonal challenges, infections or episodes of illicit drug use. Endocannabinoids, a family of signal lipids derived from polyunsaturated fatty acids, have been implicated in neuronal fate determination, the control of axonal growth, synaptogenesis and synaptic neurotransmission. Therefore the continuum and interdependence of endocannabinoid actions during the formation and function of synapses together with dynamic changes in focal and circulating endocannabinoid levels upon maternal nutritional imbalance suggest that endocannabinoids can execute the 'reprogramming' of specific neuronal networks. In the present paper, we review molecular evidence suggesting that maternal nutrition and metabolism during pregnancy can affect the formation and function of the hippocampus and hypothalamus by altering endocannabinoid signalling such that neuropsychiatric diseases and obesity respectively ensue in affected offspring. Moreover, we propose that the placenta, fetal adipose and nervous tissues interact via endocannabinoid signals. Thus endocannabinoids are hypothesized to act as a molecular substrate of maternal programming.
Collapse
|
25
|
Vilches N, Spichiger C, Mendez N, Abarzua-Catalan L, Galdames HA, Hazlerigg DG, Richter HG, Torres-Farfan C. Gestational chronodisruption impairs hippocampal expression of NMDA receptor subunits Grin1b/Grin3a and spatial memory in the adult offspring. PLoS One 2014; 9:e91313. [PMID: 24663672 PMCID: PMC3963867 DOI: 10.1371/journal.pone.0091313] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 02/10/2014] [Indexed: 11/18/2022] Open
Abstract
Epidemiological and experimental evidence correlates adverse intrauterine conditions with the onset of disease later in life. For a fetus to achieve a successful transition to extrauterine life, a myriad of temporally integrated humoral/biophysical signals must be accurately provided by the mother. We and others have shown the existence of daily rhythms in the fetus, with peripheral clocks being entrained by maternal cues, such as transplacental melatonin signaling. Among developing tissues, the fetal hippocampus is a key structure for learning and memory processing that may be anticipated as a sensitive target of gestational chronodisruption. Here, we used pregnant rats exposed to constant light treated with or without melatonin as a model of gestational chronodisruption, to investigate effects on the putative fetal hippocampus clock, as well as on adult offspring’s rhythms, endocrine and spatial memory outcomes. The hippocampus of fetuses gestated under light:dark photoperiod (12:12 LD) displayed daily oscillatory expression of the clock genes Bmal1 and Per2, clock-controlled genes Mtnr1b, Slc2a4, Nr3c1 and NMDA receptor subunits 1B-3A-3B. In contrast, in the hippocampus of fetuses gestated under constant light (LL), these oscillations were suppressed. In the adult LL offspring (reared in LD during postpartum), we observed complete lack of day/night differences in plasma melatonin and decreased day/night differences in plasma corticosterone. In the adult LL offspring, overall hippocampal day/night difference of gene expression was decreased, which was accompanied by a significant deficit of spatial memory. Notably, maternal melatonin replacement to dams subjected to gestational chronodisruption prevented the effects observed in both, LL fetuses and adult LL offspring. Collectively, the present data point to adverse effects of gestational chronodisruption on long-term cognitive function; raising challenging questions about the consequences of shift work during pregnancy. The present study also supports that developmental plasticity in response to photoperiodic cues may be modulated by maternal melatonin.
Collapse
Affiliation(s)
- Nelson Vilches
- Laboratorio de Cronobiologia del Desarrollo, Instituto de Anatomia, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Carlos Spichiger
- Laboratorio de Cronobiologia del Desarrollo, Instituto de Anatomia, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Natalia Mendez
- Laboratorio de Cronobiologia del Desarrollo, Instituto de Anatomia, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Lorena Abarzua-Catalan
- Laboratorio de Cronobiologia del Desarrollo, Instituto de Anatomia, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Hugo A. Galdames
- Laboratorio de Cronobiologia del Desarrollo, Instituto de Anatomia, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - David G. Hazlerigg
- Institute of Biological and Environmental Sciences, Zoology Building, Tillydrone Avenue, University of Aberdeen, Aberdeen, United Kingdom
| | - Hans G. Richter
- Laboratorio de Cronobiologia del Desarrollo, Instituto de Anatomia, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Claudia Torres-Farfan
- Laboratorio de Cronobiologia del Desarrollo, Instituto de Anatomia, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
- * E-mail:
| |
Collapse
|
26
|
Coplan JD, Fathy HM, Abdallah CG, Ragab SA, Kral JG, Mao X, Shungu DC, Mathew SJ. Reduced hippocampal N-acetyl-aspartate (NAA) as a biomarker for overweight. NEUROIMAGE-CLINICAL 2014; 4:326-35. [PMID: 24501701 PMCID: PMC3913836 DOI: 10.1016/j.nicl.2013.12.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 12/22/2013] [Accepted: 12/23/2013] [Indexed: 12/11/2022]
Abstract
OBJECTIVE We previously demonstrated an inverse relationship between both dentate gyrus neurogenesis - a form of neuroplasticity - and expression of the antiapoptotic gene marker, BCL-2 and adult macaque body weight. We therefore explored whether a similar inverse correlation existed in humans between body mass index (BMI) and hippocampal N-acetyl-aspartate (NAA), a marker of neuronal integrity and putatively, neuroplasticity. We also studied the relationship of a potentially neurotoxic process, worry, to hippocampal NAA in patients with generalized anxiety disorder (GAD) and control subjects (CS). METHODS We combined two previously studied cohorts of GAD and control subjects. Using proton magnetic resonance spectroscopy imaging ((1)H MRSI) in medication-free patients with GAD (n = 29) and a matched healthy control group (n = 22), we determined hippocampal concentrations of (1) NAA (2) choline containing compounds (CHO), and (3) Creatine + phosphocreatine (CR). Data were combined from 1.5 T and 3 T scans by converting values from each cohort to z-scores. Overweight and GAD diagnosis were used as categorical variables while the Penn State Worry Questionnaire (PSWQ) and Anxiety Sensitivity Index (ASI) were used as dependent variables. RESULTS Overweight subjects (BMI ≥ 25) exhibited lower NAA levels in the hippocampus than normal-weight subjects (BMI < 25) (partial Eta-squared = 0.14) controlling for age, sex and psychiatric diagnosis, and the effect was significant for the right hippocampus in both GAD patients and control subjects. An inverse linear correlation was noted in all subjects between right hippocampal NAA and BMI. High scores on the PSWQ predicted low hippocampal NAA and CR. Both BMI and worry were independent inverse predictors of hippocampal NAA. CONCLUSION Overweight was associated with reduced NAA concentrations in the hippocampus with a strong effect size. Future mechanistic studies are warranted.
Collapse
Affiliation(s)
- Jeremy D Coplan
- Department of Psychiatry, Division of Neuropsychopharmacology, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Hassan M Fathy
- Department of Psychiatry, Division of Neuropsychopharmacology, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Chadi G Abdallah
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Sherif A Ragab
- Department of Psychiatry, Division of Neuropsychopharmacology, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - John G Kral
- Department of Surgery and Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Xiangling Mao
- Department of Radiology, Weill Medical College of Cornell University, New York, NY, USA ; Department of Psychiatry, Weill Medical College of Cornell University, New York, NY, USA ; Department of Biophysics, Weill Medical College of Cornell University, New York, NY, USA
| | - Dikoma C Shungu
- Department of Radiology, Weill Medical College of Cornell University, New York, NY, USA ; Department of Psychiatry, Weill Medical College of Cornell University, New York, NY, USA ; Department of Biophysics, Weill Medical College of Cornell University, New York, NY, USA
| | - Sanjay J Mathew
- Menninger Department of Psychiatry, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
27
|
Iozzo P, Holmes M, Schmidt MV, Cirulli F, Guzzardi MA, Berry A, Balsevich G, Andreassi MG, Wesselink JJ, Liistro T, Gómez-Puertas P, Eriksson JG, Seckl J. Developmental ORIgins of Healthy and Unhealthy AgeiNg: the role of maternal obesity--introduction to DORIAN. Obes Facts 2014; 7:130-51. [PMID: 24801105 PMCID: PMC5644840 DOI: 10.1159/000362656] [Citation(s) in RCA: 22] [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: 01/09/2014] [Accepted: 02/12/2014] [Indexed: 12/31/2022] Open
Abstract
Europe has the highest proportion of elderly people in the world. Cardiovascular disease, type 2 diabetes, sarcopenia and cognitive decline frequently coexist in the same aged individual, sharing common early risk factors and being mutually reinforcing. Among conditions which may contribute to establish early risk factors, this review focuses on maternal obesity, since the epidemic of obesity involves an ever growing number of women of reproductive age and children, calling for appropriate studies to understand the consequences of maternal obesity on the offspring's health and for developing effective measures and policies to improve people's health before their conception and birth. Though the current knowledge suggests that the long-term impact of maternal obesity on the offspring's health may be substantial, the outcomes of maternal obesity over the lifespan have not been quantified, and the molecular changes induced by maternal obesity remain poorly characterized. We hypothesize that maternal insulin resistance and reduced placental glucocorticoid catabolism, leading to oxidative stress, may damage the DNA, either in its structure (telomere shortening) or in its function (via epigenetic changes), resulting in altered gene expression/repair, disease during life, and pathological ageing. This review illustrates the background to the EU-FP7-HEALTH-DORIAN project.
Collapse
Affiliation(s)
- Patricia Iozzo
- Institute of Clinical Physiology, National Research Council (CNR), Pis
- *Patricia Iozzo, MD, PhD, Institute of Clinical Physiology, National Research Council (CNR), Via Moruzzi 1, 56124 Pisa (Italy),
| | - Megan Holmes
- Endocrinology Unit, Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | | | | | | | | | | | | | | | - Tiziana Liistro
- Institute of Clinical Physiology, National Research Council (CNR), Pis
| | | | - Johan G. Eriksson
- Samfundet Folkhälsan i Svenska Finland rf (Folkhälsan), Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
| | - Jonathan Seckl
- Endocrinology Unit, Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
28
|
Prenatal exposure to nicotine stimulates neurogenesis of orexigenic peptide-expressing neurons in hypothalamus and amygdala. J Neurosci 2013; 33:13600-11. [PMID: 23966683 DOI: 10.1523/jneurosci.5835-12.2013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Animal and clinical studies show that gestational exposure to nicotine increases the propensity of offspring to consume nicotine, but the precise mechanism mediating this behavioral phenomenon is unclear. The present study in Sprague Dawley rats examined the possibility that the orexigenic peptide systems, enkephalin (ENK) and orexin (OX), which are stimulated by nicotine in adult animals and promote consummatory behavior, may be similarly responsive to nicotine's stimulatory effect in utero while having long-term behavioral consequences. The results demonstrated that nicotine exposure during gestation at low doses (0.75 or 1.5 mg/kg/d) significantly increased mRNA levels and density of neurons that express ENK in the hypothalamic paraventricular nucleus and central nucleus of the amygdala, OX, and another orexigenic peptide, melanin-concentrating hormone, in the perifornical lateral hypothalamus in preweanling offspring. These effects persisted in the absence of nicotine, at least until puberty. Colabeling of the cell proliferation marker BrdU with the neuronal marker NeuN and peptides revealed a marked stimulatory effect of prenatal nicotine on neurogenesis, but not gliogenesis, and also on the number of newly generated neurons expressing ENK, OX, or melanin-concentrating hormone. During adolescence, offspring also exhibited significant behavioral changes, increased consumption of nicotine and other substances of abuse, ethanol and a fat-rich diet, with no changes in chow and water intake or body weight. These findings reveal a marked sensitivity during gestation of the orexigenic peptide neurons to low nicotine doses that may increase the offspring's propensity to overconsume substances of abuse during adolescence.
Collapse
|
29
|
Ferro Cavalcante TC, Lima da Silva JM, da Marcelino da Silva AA, Muniz GS, da Luz Neto LM, Lopes de Souza S, Manhães de Castro R, Ferraz KM, do Nascimento E. Effects of a Westernized Diet on the Reflexes and Physical Maturation of Male Rat Offspring During the Perinatal Period. Lipids 2013; 48:1157-68. [DOI: 10.1007/s11745-013-3833-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 08/13/2013] [Indexed: 11/30/2022]
|
30
|
Naef L, Gratton A, Walker CD. Exposure to high fat during early development impairs adaptations in dopamine and neuroendocrine responses to repeated stress. Stress 2013; 16:540-8. [PMID: 23786443 DOI: 10.3109/10253890.2013.805321] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Perturbations in the perinatal environment have been shown to significantly alter mesolimbic dopamine (DA) and hypothalamic-pituitary-adrenal (HPA) responses to stressors in adulthood. We have previously demonstrated that adult offspring exposed to high fat during the last week of gestation and throughout lactation display permanent alterations in mesolimbic DA function and behavior. The goal of the present study was to investigate nucleus accumbens (NAc) DA and HPA responses to acute and repeated stress in high fat exposed (HFD, 30% fat) and control (CD, 5% fat) offspring. Using microdialysis to monitor extracellular DA, we report that adult HFD offspring show an enhanced NAc DA response to acute tail-pinch compared to CD offspring. With repeated tail-pinch, the response of the HFD animals remains unchanged while CD offspring exhibit a sensitized DA response. The pattern of the DA response to both acute and repeated stress is also significantly altered by early diet exposure with an earlier peak and faster return to baseline levels in CD compared with HFD offspring. Similarly, neuroendocrine adaptations to repeated tail-pinch are observed in CD animals, but not in HFD animals. While controls display a habituated adrenocorticotropic hormone (ACTH) response to repeated tail-pinch, and an exacerbated ACTH response to a novel stressor, this effect was not observed in the HFD offspring. Together, our data demonstrate that exposure to high fat during early development impairs adaptations in NAc DA and HPA responses usually observed with repeated stress.
Collapse
Affiliation(s)
- Lindsay Naef
- Department of Psychiatry and Integrated Program in Neuroscience, McGill University, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | | | | |
Collapse
|
31
|
Perinatal programming of adult hippocampal structure and function; emerging roles of stress, nutrition and epigenetics. Trends Neurosci 2013; 36:621-31. [PMID: 23998452 DOI: 10.1016/j.tins.2013.08.002] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 08/05/2013] [Accepted: 08/06/2013] [Indexed: 11/22/2022]
Abstract
Early-life stress lastingly affects adult cognition and increases vulnerability to psychopathology, but the underlying mechanisms remain elusive. In this Opinion article, we propose that early nutritional input together with stress hormones and sensory stimuli from the mother during the perinatal period act synergistically to program the adult brain, possibly via epigenetic mechanisms. We hypothesize that stress during gestation or lactation affects the intake of macro- and micronutrients, including dietary methyl donors, and/or impairs the dam's metabolism, thereby altering nutrient composition and intake by the offspring. In turn, this may persistently modulate gene expression via epigenetic programming, thus altering hippocampal structure and cognition. Understanding how the combination of stress, nutrition, and epigenetics shapes the adult brain is essential for effective therapies.
Collapse
|
32
|
Moon HS, Dalamaga M, Kim SY, Polyzos SA, Hamnvik OP, Magkos F, Paruthi J, Mantzoros CS. Leptin's role in lipodystrophic and nonlipodystrophic insulin-resistant and diabetic individuals. Endocr Rev 2013; 34:377-412. [PMID: 23475416 PMCID: PMC3660716 DOI: 10.1210/er.2012-1053] [Citation(s) in RCA: 187] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Leptin is an adipocyte-secreted hormone that has been proposed to regulate energy homeostasis as well as metabolic, reproductive, neuroendocrine, and immune functions. In the context of open-label uncontrolled studies, leptin administration has demonstrated insulin-sensitizing effects in patients with congenital lipodystrophy associated with relative leptin deficiency. Leptin administration has also been shown to decrease central fat mass and improve insulin sensitivity and fasting insulin and glucose levels in HIV-infected patients with highly active antiretroviral therapy (HAART)-induced lipodystrophy, insulin resistance, and leptin deficiency. On the contrary, the effects of leptin treatment in leptin-replete or hyperleptinemic obese individuals with glucose intolerance and diabetes mellitus have been minimal or null, presumably due to leptin tolerance or resistance that impairs leptin action. Similarly, experimental evidence suggests a null or a possibly adverse role of leptin treatment in nonlipodystrophic patients with nonalcoholic fatty liver disease. In this review, we present a description of leptin biology and signaling; we summarize leptin's contribution to glucose metabolism in animals and humans in vitro, ex vivo, and in vivo; and we provide insights into the emerging clinical applications and therapeutic uses of leptin in humans with lipodystrophy and/or diabetes.
Collapse
Affiliation(s)
- Hyun-Seuk Moon
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Rivera P, Pérez-Martín M, Pavón FJ, Serrano A, Crespillo A, Cifuentes M, López-Ávalos MD, Grondona JM, Vida M, Fernández-Llebrez P, de Fonseca FR, Suárez J. Pharmacological administration of the isoflavone daidzein enhances cell proliferation and reduces high fat diet-induced apoptosis and gliosis in the rat hippocampus. PLoS One 2013; 8:e64750. [PMID: 23741384 PMCID: PMC3669353 DOI: 10.1371/journal.pone.0064750] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 04/17/2013] [Indexed: 12/16/2022] Open
Abstract
Soy extracts have been claimed to be neuroprotective against brain insults, an effect related to the estrogenic properties of isoflavones. However, the effects of individual isoflavones on obesity-induced disruption of adult neurogenesis have not yet been analyzed. In the present study we explore the effects of pharmacological administration of daidzein, a main soy isoflavone, in cell proliferation, cell apoptosis and gliosis in the adult hippocampus of animals exposed to a very high-fat diet. Rats made obese after 12-week exposure to a standard or high-fat (HFD, 60%) diets were treated with daidzein (50 mg kg(-1)) for 13 days. Then, plasma levels of metabolites and metabolic hormones, cell proliferation in the subgranular zone of the dentate gyrus (SGZ), and immunohistochemical markers of hippocampal cell apoptosis (caspase-3), gliosis (GFAP and Iba-1), food reward factor FosB and estrogen receptor alpha (ERα) were analyzed. Treatment with daidzein reduced food/caloric intake and body weight gain in obese rats. This was associated with glucose tolerance, low levels of HDL-cholesterol, insulin, adiponectin and testosterone, and high levels of leptin and 17β-estradiol. Daidzein increased the number of phospho-histone H3 and 5-bromo-2-deoxyuridine (BrdU)-ir cells detected in the SGZ of standard diet and HFD-fed rats. Daidzein reversed the HFD-associated enhanced immunohistochemical expression of caspase-3, FosB, GFAP, Iba-1 and ERα in the hippocampus, being more prominent in the dentate gyrus. These results suggest that pharmacological treatment with isoflavones regulates metabolic alterations associated with enhancement of cell proliferation and reduction of apoptosis and gliosis in response to high-fat diet.
Collapse
Affiliation(s)
- Patricia Rivera
- Laboratorio de Medicina Regenerativa (UGC Salud Mental), Instituto de Investigación Biomédica (IBIMA), Complejo Hospitalario de Málaga (Hospital Carlos Haya), Pabellón de Gobierno, Málaga, Spain
- CIBER OBN, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Madrid, Spain
| | - Margarita Pérez-Martín
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Francisco J. Pavón
- Laboratorio de Medicina Regenerativa (UGC Salud Mental), Instituto de Investigación Biomédica (IBIMA), Complejo Hospitalario de Málaga (Hospital Carlos Haya), Pabellón de Gobierno, Málaga, Spain
- CIBER OBN, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Madrid, Spain
| | - Antonia Serrano
- Laboratorio de Medicina Regenerativa (UGC Salud Mental), Instituto de Investigación Biomédica (IBIMA), Complejo Hospitalario de Málaga (Hospital Carlos Haya), Pabellón de Gobierno, Málaga, Spain
- CIBER OBN, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Madrid, Spain
| | - Ana Crespillo
- Laboratorio de Medicina Regenerativa (UGC Salud Mental), Instituto de Investigación Biomédica (IBIMA), Complejo Hospitalario de Málaga (Hospital Carlos Haya), Pabellón de Gobierno, Málaga, Spain
- CIBER OBN, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Madrid, Spain
| | - Manuel Cifuentes
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- CIBER BBN, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Madrid, Spain
| | - María-Dolores López-Ávalos
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Jesús M. Grondona
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Margarita Vida
- Laboratorio de Medicina Regenerativa (UGC Salud Mental), Instituto de Investigación Biomédica (IBIMA), Complejo Hospitalario de Málaga (Hospital Carlos Haya), Pabellón de Gobierno, Málaga, Spain
- CIBER OBN, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Madrid, Spain
| | - Pedro Fernández-Llebrez
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- Laboratorio de Medicina Regenerativa (UGC Salud Mental), Instituto de Investigación Biomédica (IBIMA), Complejo Hospitalario de Málaga (Hospital Carlos Haya), Pabellón de Gobierno, Málaga, Spain
- CIBER OBN, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Madrid, Spain
| | - Juan Suárez
- Laboratorio de Medicina Regenerativa (UGC Salud Mental), Instituto de Investigación Biomédica (IBIMA), Complejo Hospitalario de Málaga (Hospital Carlos Haya), Pabellón de Gobierno, Málaga, Spain
- CIBER OBN, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Madrid, Spain
| |
Collapse
|
34
|
Perinatal high fat diet alters glucocorticoid signaling and anxiety behavior in adulthood. Neuroscience 2013; 240:1-12. [PMID: 23454542 DOI: 10.1016/j.neuroscience.2013.02.044] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 02/19/2013] [Accepted: 02/20/2013] [Indexed: 12/23/2022]
Abstract
Maternal obesity carries significant health risks for offspring that manifest later in life, including metabolic syndrome, cardiovascular disease and affective disorders. Programming of the hypothalamic-pituitary-adrenal (HPA) axis during development mediates both metabolic homeostasis and the response to psychosocial stress in offspring. A diet high in fat alters maternal systemic corticosterone levels, but effects in offspring on limbic brain areas regulating the HPA axis and anxiety behavior are poorly understood. In addition to their role in the response to psychosocial stress, corticosteroid receptors form part of the glucocorticoid signaling pathway comprising downstream inflammatory processes. Increased systemic inflammation is a hallmark of high-fat diet exposure, though altered expression of these genes in limbic brain areas has not been examined. We studied the influence of high-fat diet exposure during pre-weaning development in rats on gene expression in the amygdala and hippocampus by quantitative real-time polymerase chain reaction (PCR), anxiety behavior in the Open field, elevated plus maze and light-dark transition tasks, and corticosterone levels in response to stress by radioimmunoassay. As adults, offspring exposed to perinatal high-fat diet show increased expression of corticosterone receptors in the amygdala and altered pro-inflammatory and anti-inflammatory expression in the hippocampus and amygdala in genes known to be regulated by the glucocorticoid receptor. These changes were associated with increased anxiety behavior, decreased basal corticosterone levels and a slower return to baseline levels following a stress challenge. The data indicate that the dietary environment during development programs glucocorticoid signaling pathways in limbic areas relevant for the regulation of HPA function and anxiety behavior.
Collapse
|
35
|
Sullivan EL, Nousen EK, Chamlou KA. Maternal high fat diet consumption during the perinatal period programs offspring behavior. Physiol Behav 2012; 123:236-42. [PMID: 23085399 DOI: 10.1016/j.physbeh.2012.07.014] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 07/20/2012] [Accepted: 07/30/2012] [Indexed: 12/16/2022]
Abstract
The environment that a developing offspring experiences during the perinatal period is markedly influenced by maternal health and diet composition. Evidence from both epidemiological studies and animal models indicates that maternal diet and metabolic status play a critical role in programming the neural circuitry that regulates behavior, resulting in long-term consequences for offspring behavior. Maternal diet and metabolic state influence the behavior of offspring directly by impacting the intrauterine environment and indirectly by modulating maternal behavior. The mechanisms by which maternal diet and metabolic profile shape the perinatal environment remain largely unknown, but recent research has found that increases in inflammatory cytokines, nutrients (glucose and fatty acids), and hormones (insulin and leptin) affect the environment of the developing offspring. Offspring exposed to maternal obesity and high fat diet consumption during development are more susceptible to developing mental health and behavioral disorders such as anxiety, depression, attention deficit hyperactivity disorder, and autism spectrum disorders. Recent evidence suggests that this increased risk for behavioral disorders is driven by modifications in the development of neural pathways involved in behavioral regulation. In particular, research indicates that the development of the serotonergic system is impacted by exposure to maternal obesity and high fat diet consumption, and this disruption may underlie many of the behavioral disturbances observed in these offspring. Given the high rates of obesity and high fat diet consumption in pregnant women, it is vital to examine the influence that maternal nutrition and metabolic profile have on the developing offspring.
Collapse
Affiliation(s)
- Elinor L Sullivan
- Department of Biology, University of Portland, Portland, OR, USA; Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA.
| | | | | |
Collapse
|
36
|
Fetal stress and programming of hypoxic/ischemic-sensitive phenotype in the neonatal brain: mechanisms and possible interventions. Prog Neurobiol 2012; 98:145-65. [PMID: 22627492 DOI: 10.1016/j.pneurobio.2012.05.010] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 05/10/2012] [Accepted: 05/11/2012] [Indexed: 12/12/2022]
Abstract
Growing evidence of epidemiological, clinical and experimental studies has clearly shown a close link between adverse in utero environment and the increased risk of neurological, psychological and psychiatric disorders in later life. Fetal stresses, such as hypoxia, malnutrition, and fetal exposure to nicotine, alcohol, cocaine and glucocorticoids may directly or indirectly act at cellular and molecular levels to alter the brain development and result in programming of heightened brain vulnerability to hypoxic-ischemic encephalopathy and the development of neurological diseases in the postnatal life. The underlying mechanisms are not well understood. However, glucocorticoids may play a crucial role in epigenetic programming of neurological disorders of fetal origins. This review summarizes the recent studies about the effects of fetal stress on the abnormal brain development, focusing on the cellular, molecular and epigenetic mechanisms and highlighting the central effects of glucocorticoids on programming of hypoxic-ischemic-sensitive phenotype in the neonatal brain, which may enhance the understanding of brain pathophysiology resulting from fetal stress and help explore potential targets of timely diagnosis, prevention and intervention in neonatal hypoxic-ischemic encephalopathy and other brain disorders.
Collapse
|
37
|
Maniam J, Morris MJ. The link between stress and feeding behaviour. Neuropharmacology 2012; 63:97-110. [PMID: 22710442 DOI: 10.1016/j.neuropharm.2012.04.017] [Citation(s) in RCA: 173] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 04/11/2012] [Accepted: 04/20/2012] [Indexed: 02/06/2023]
Abstract
Exposure to stress is inevitable, and it may occur, to varying degrees, at different phases throughout the lifespan. The impact of stress experienced in later life has been well documented as many populations in modern society experience increasing socio-economic demands. The effects of stress early in life are less well known, partly as the impact of an early exposure may be difficult to quantify, however emerging evidence shows it can impact later in life. One of the major impacts of stress besides changes in psychosocial behaviour is altered feeding responses. The system that regulates stress responses, the hypothalamo-pituitary-adrenal axis, also regulates feeding responses because the neural circuits that regulate food intake converge on the paraventricular nucleus, which contains corticotrophin releasing hormone (CRH), and urocortin containing neurons. In other words the systems that control food intake and stress responses share the same anatomy and thus each system can influence each other in eliciting a response. Stress is known to alter feeding responses in a bidirectional pattern, with both increases and decreases in intake observed. Stress-induced bidirectional feeding responses underline the complex mechanisms and multiple contributing factors, including the levels of glucocorticoids (dependent on the severity of a stressor), the interaction between glucocorticoids and feeding related neuropeptides such as neuropeptide Y (NPY), alpha-melanocyte stimulating hormone (α-MSH), agouti-related protein (AgRP), melanocortins and their receptors, CRH, urocortin and peripheral signals (leptin, insulin and ghrelin). This review discusses the neuropeptides that regulate feeding behaviour and how their function can be altered through cross-talk with hormones and neuropeptides that also regulate the hypothalamo-pituitary-adrenal axis. In addition, long-term stress induced alterations in feeding behaviour, and changes in gene expression of neuropeptides regulating stress and food intake through epigenetic modifications will be discussed. This article is part of a Special Issue entitled 'SI: Central Control of Food Intake'.
Collapse
Affiliation(s)
- Jayanthi Maniam
- Pharmacology, School of Medical Sciences, University of New South Wales, Sydney NSW 2052, Australia
| | | |
Collapse
|
38
|
Rodriguez J, Rodríguez‐González G, Reyes‐Castro L, Ibáñez C, Ramírez A, Chavira R, Larrea F, Nathanielsz P, Zambrano E. Maternal obesity in the rat programs male offspring exploratory, learning and motivation behavior: prevention by dietary intervention pre‐gestation or in gestation. Int J Dev Neurosci 2012; 30:75-81. [DOI: 10.1016/j.ijdevneu.2011.12.012] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 12/16/2011] [Accepted: 12/28/2011] [Indexed: 01/19/2023] Open
Affiliation(s)
- J.S. Rodriguez
- Center for Pregnancy and Newborn ResearchDepartment of Obstetrics and GynecologyUniversity of Texas Health Sciences CenterSan AntonioTX78229USA
| | - G.L. Rodríguez‐González
- Department of Reproductive BiologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMéxico City14000Mexico
| | - L.A. Reyes‐Castro
- Department of Reproductive BiologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMéxico City14000Mexico
| | - C. Ibáñez
- Department of Reproductive BiologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMéxico City14000Mexico
| | - A. Ramírez
- Department of Reproductive BiologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMéxico City14000Mexico
| | - R. Chavira
- Department of Reproductive BiologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMéxico City14000Mexico
| | - F. Larrea
- Department of Reproductive BiologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMéxico City14000Mexico
| | - P.W. Nathanielsz
- Center for Pregnancy and Newborn ResearchDepartment of Obstetrics and GynecologyUniversity of Texas Health Sciences CenterSan AntonioTX78229USA
| | - E. Zambrano
- Department of Reproductive BiologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMéxico City14000Mexico
| |
Collapse
|
39
|
Sullivan EL, Nousen EK, Chamlou KA, Grove KL. The Impact of Maternal High-Fat Diet Consumption on Neural Development and Behavior of Offspring. INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2012; 2:S7-S13. [PMID: 26069734 PMCID: PMC4460829 DOI: 10.1038/ijosup.2012.15] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Maternal diet and metabolic state are important factors in determining the environment experienced during perinatal development. Epidemiological studies and evidence from animal models provide evidence that a mother's diet and metabolic condition are important in programming the neural circuitry that regulates behavior, resulting in a persistent impact on the offspring's behavior. Potential mechanisms by which maternal diet and metabolic profile influence the perinatal environment include placental dysfunction and increases in circulating factors such as inflammatory cytokines, nutrients (glucose and fatty acids) and hormones (insulin and leptin). Maternal obesity and high-fat diet (HFD) consumption exposure during development have been observed to increase the risk of developing serious mental health and behavioral disorders including anxiety, depression, attention deficit hyperactivity disorder and autism spectrum disorder. The increased risk of developing these behavioral disorders is postulated to be due to perturbations in the development of neural pathways that regulate behavior, including the serotonergic, dopaminergic and melanocortinergic systems. It is critical to examine the influence that a mother's nutrition and metabolic profile have on the developing offspring considering the current and alarmingly high prevalence of obesity and HFD consumption in pregnant women.
Collapse
Affiliation(s)
- E L Sullivan
- Department of Biology, University of Portland, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - E K Nousen
- Department of Biology, University of Portland, Portland, OR, USA
| | - K A Chamlou
- Department of Biology, University of Portland, Portland, OR, USA
| | - K L Grove
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| |
Collapse
|
40
|
Abstract
Suboptimal maternal nutritional status has been implicated in the development of cardiovascular risk in the child. Initially inferred from studies of low-birthweight children, investigations in cohorts of women subjected to famine provide direct evidence for an independent influence of the mother's diet on the cardiovascular health of her child. Animal studies from rodents and sheep have shown associations between maternal undernutrition and raised blood pressure, as well as abnormalities in resistance artery function, particularly in endothelium-dependent responses. Early life exposure to the influences of maternal over nutritional states, e.g. obesity and excessive gestational weight gain, has also been associated with markers of cardiovascular risk in man, and animal models have shown raised blood pressure and endothelial dysfunction in offspring of diet-induced obese dams. Increased sympathetic tone is commonly associated with hypertension in animal models of both under nutritional and over nutritional states. This and several other similarities may indicate commonality of mechanism and could reflect supranormal nutritional status in postnatal life in both conditions.
Collapse
Affiliation(s)
- Lucilla Poston
- Division of Women's Health, King's College London and King's Health Partners, London, UK.
| |
Collapse
|
41
|
Ong ZY, Muhlhausler BS. Maternal "junk-food" feeding of rat dams alters food choices and development of the mesolimbic reward pathway in the offspring. FASEB J 2011; 25:2167-79. [PMID: 21427213 DOI: 10.1096/fj.10-178392] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Individuals exposed to high-fat, high-sugar diets before birth have an increased risk of obesity in later life. Recent studies have shown that these offspring exhibit increased preference for fat, leading to suggestions that perinatal exposure to high-fat, high-sugar foods results in permanent changes within the central reward system that increase the subsequent drive to overconsume palatable foods. The present study has determined the effect of a maternal "junk-food" diet on the expression of key components of the mesolimbic reward pathway in the offspring of rat dams at 6 wk and 3 mo of age. We show that offspring of junk-food-fed (JF) dams exhibit higher fat intake from weaning until at least 3 mo of age (males: 16 ± 0.6 vs. 11 ± 0.8 g/kg/d; females: 19 ± 1.3 vs. 13 ± 0.4 g/kg/d; P<0.01). mRNA expression of μ-opioid receptor (Mu) was 1.6-fold higher (P<0.01) and dopamine active transporter (DAT) was 2-fold lower (P<0.05) in JF offspring at 6 wk of age. By 3 mo, these differences were reversed, and Mu mRNA expression was 2.8-fold lower (P<0.01) and DAT mRNA expression was 1.9-fold higher (P<0.01) in the JF offspring. These findings suggest that perinatal exposure to high-fat, high-sugar diets results in altered development of the central reward system, resulting in increased fat intake and altered response of the reward system to excessive junk-food intake in postnatal life.
Collapse
Affiliation(s)
- Z Y Ong
- Sansom Institute for Health Research, School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia
| | | |
Collapse
|
42
|
Naef L, Moquin L, Dal Bo G, Giros B, Gratton A, Walker CD. Maternal high-fat intake alters presynaptic regulation of dopamine in the nucleus accumbens and increases motivation for fat rewards in the offspring. Neuroscience 2011; 176:225-36. [DOI: 10.1016/j.neuroscience.2010.12.037] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 12/17/2010] [Accepted: 12/21/2010] [Indexed: 11/28/2022]
|
43
|
Sullivan EL, Smith MS, Grove KL. Perinatal exposure to high-fat diet programs energy balance, metabolism and behavior in adulthood. Neuroendocrinology 2011; 93:1-8. [PMID: 21079387 PMCID: PMC3700139 DOI: 10.1159/000322038] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Accepted: 10/15/2010] [Indexed: 12/16/2022]
Abstract
The perinatal environment plays an important role in programming many aspects of physiology and behavior including metabolism, body weight set point, energy balance regulation and predisposition to mental health-related disorders such as anxiety, depression and attention deficit hyperactivity disorder. Maternal health and nutritional status heavily influence the early environment and have a long-term impact on critical central pathways, including the melanocortinergic, serotonergic system and dopaminergic systems. Evidence from a variety of animal models including rodents and nonhuman primates indicates that exposure to maternal high-fat diet (HFD) consumption programs offspring for increased risk of adult obesity. Hyperphagia and increased preference for fatty and sugary foods are implicated as mechanisms for the increased obesity risk. The effects of maternal HFD consumption on energy expenditure are unclear, and future studies need to address the impact of perinatal HFD exposure on this important component of energy balance regulation. Recent evidence from animal models also indicates that maternal HFD consumption increases the risk of offspring developing mental health-related disorders such as anxiety. Potential mechanisms for perinatal HFD programming of neural pathways include circulating factors, such as hormones (leptin, insulin), nutrients (fatty acids, triglycerides and glucose) and inflammatory cytokines. As maternal HFD consumption and obesity are common and rapidly increasing, we speculate that future generations will be at increased risk for both metabolic and mental health disorders. Thus, it is critical that future studies identify therapeutic strategies that are effective at preventing maternal HFD-induced malprogramming.
Collapse
Affiliation(s)
- Elinor L Sullivan
- Department of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, USA.
| | | | | |
Collapse
|
44
|
Walker CD. Maternal touch and feed as critical regulators of behavioral and stress responses in the offspring. Dev Psychobiol 2010; 52:638-50. [DOI: 10.1002/dev.20492] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
45
|
Mattson MP. The impact of dietary energy intake on cognitive aging. Front Aging Neurosci 2010; 2:5. [PMID: 20552045 PMCID: PMC2874403 DOI: 10.3389/neuro.24.005.2010] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Accepted: 01/27/2010] [Indexed: 01/21/2023] Open
Abstract
Rodents that are insulin resistant and obese as the result of genetic factors, overeating and/or a sedentary lifestyle, exhibit cognitive deficits that worsen with advancing age compared to their more svelte counterparts. Data from epidemiological and clinical studies suggest similar adverse effects of excessive dietary energy intake and insulin resistance on cognition in humans. Our findings from studies of animal models suggest that dietary energy restriction can enhance neural plasticity and reduce the vulnerability of the brain to age-related dysfunction and disease. Dietary energy restriction may exert beneficial effects on the brain by engaging adaptive cellular stress response pathways resulting in the up-regulation of genes that encode proteins that promote neural plasticity and cell survival (e.g., neurotrophic factors, protein chaperones and redox enzymes). Two energy state-sensitive factors that are proving particularly important in regulating energy balance and improving/preserving cognitive function are brain-derived neurotrophic factor and glucagon-like peptide 1. Alternate day calorie restriction, novel insulin-sensitizing and neuroprotective agents, and drugs that activate adaptive stress response pathways, are examples of approaches for preserving cognitive function that show promise in preclinical studies.
Collapse
Affiliation(s)
- Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research ProgramBaltimore, MD, USA
| |
Collapse
|
46
|
Grayson BE, Kievit P, Smith MS, Grove KL. Critical determinants of hypothalamic appetitive neuropeptide development and expression: species considerations. Front Neuroendocrinol 2010; 31:16-31. [PMID: 19822169 PMCID: PMC2813940 DOI: 10.1016/j.yfrne.2009.10.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Accepted: 10/05/2009] [Indexed: 01/21/2023]
Abstract
Over the last decade there has been a striking increase in the early onset of metabolic disease, including obesity and diabetes. The regulation of energy homeostasis is complex and involves the intricate integration of peripheral and central systems, including the hypothalamus. This review provides an overview of the development of brain circuitry involved in the regulation of energy homeostasis as well as recent findings related to the impact of both prenatal and postnatal maternal environment on the development of these circuits. There is surprising evidence that both overnutrition and undernutrition impact the development of these circuits in a similar manner as well as having similar consequences of increased obesity and diabetes later in life. There is also a special focus on relevant species differences in the development of hypothalamic circuits. A deeper understanding of the mechanisms involved in the development of brain circuitry is needed to fully understand how the nutritional and/or maternal environments impact the functional circuitry as well as the behavior and physiological outcomes.
Collapse
Affiliation(s)
- B E Grayson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | | | |
Collapse
|
47
|
Morris MJ. Early life influences on obesity risk: maternal overnutrition and programming of obesity. Expert Rev Endocrinol Metab 2009; 4:625-637. [PMID: 30780787 DOI: 10.1586/eem.09.45] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
While adult lifestyle factors undoubtedly contribute to the incidence of obesity and its attendant disorders, mounting evidence suggests that programming of obesity may occur following over-nutrition during development. As hypothalamic control of appetite and energy expenditure is set early in life and can be perturbed by certain exposures, such as undernutrition and altered metabolic and hormonal signals, in utero exposure to maternal obesity-related changes may contribute to programming of obesity in offspring. Data from animal studies indicate both intrauterine and postnatal environments are critical determinants of the development of pathways regulating energy homeostasis. This review summarizes recent evidence of the impact of maternal obesity on subsequent obesity risk, paying particular attention to the hypothalamic regulation of appetite and markers of metabolic control. The extraordinary rise in the rates of maternal obesity underlines an urgent need to investigate the mechanisms contributing to its transgenerational effects.
Collapse
Affiliation(s)
- Margaret J Morris
- a Department of Pharmacology, School of Medical Sciences, University of New South Wales, NSW 2052, Australia.
| |
Collapse
|
48
|
Niculescu MD, Lupu DS. High fat diet-induced maternal obesity alters fetal hippocampal development. Int J Dev Neurosci 2009; 27:627-33. [PMID: 19695321 DOI: 10.1016/j.ijdevneu.2009.08.005] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Revised: 07/24/2009] [Accepted: 08/09/2009] [Indexed: 01/01/2023] Open
Abstract
The importance of maternal nutrition for fetal brain development is increasingly recognized. Previous studies have suggested that maternal obesity or maternal exposure to obesogenic diets may permanently alter brain structure and function in the offspring. To test whether maternal exposure to a high-fat diet, prior and during gestation, alters fetal hippocampal development, we fed 8-week old C57BL/6 females with a high-fat diet (60% calories from fat) for 10 weeks prior to matting and 17 days after. Fetal brains at embryonic day E17 were used to determine developmental changes in the hippocampus. We report that maternal exposure to the high-fat diet induced small for gestational age (SGA) status and fetal resorption. The proliferation of neural progenitors was increased in the neuroepithelium from hippocampus and cortex in fetuses from mothers fed the high-fat diet when compared to controls, but decreased within the dentate gyrus (DG). Apoptosis in the hippocampus was decreased (Ammon's Horn and fimbria). The differentiation of calretinin-positive neurons within the DG was also decreased. These data indicate that, under the influence of a maternal high-fat diet administered prior and during gestation, fetal hippocampal development is altered at embryonic day 17, as indicated by region-specific changes in proliferation of neural precursors, decreased apoptosis, and by decreased neuronal differentiation within the dentate gyrus.
Collapse
Affiliation(s)
- Mihai D Niculescu
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC, United States.
| | | |
Collapse
|