1
|
Calvino C, Almeida MMD, Reis-Gomes CF, Andrade BDS, Neves GA, Pazos-Moura CC, Trevenzoli IH. Maternal obesity induces sex-specific changes in the endocannabinoid system of the hypothalamus and dorsal hippocampus of offspring associated with anxiety-like behavior in adolescent female rats. Horm Behav 2024; 166:105648. [PMID: 39362071 DOI: 10.1016/j.yhbeh.2024.105648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024]
Abstract
Maternal obesity during perinatal period increases the risk of metabolic and behavioral deleterious outcomes in the offspring, since it is critical for brain development, maturation, and reorganization. These processes are highly modulated by the endocannabinoid system (ECS), which comprises the main lipid ligands anandamide and 2-arachidonoylglycerol, cannabinoid receptors 1 and 2 (CB1R and CB2R), and several metabolizing enzymes. The ECS is overactivated in obesity and it contributes to the physiological activity of the hypothalamus-pituitary-adrenal (HPA) axis, promoting stress relief. We have previously demonstrated that maternal high-fat diet during gestation and lactation programmed the food preference for fat in adolescent male offspring and adult male and female offspring. In the present study, we hypothesized that maternal diet-induced obesity would induce sex-specific changes of the ECS in the hypothalamus and dorsal hippocampus of rat offspring associated with dysregulation of the HPA axis and stress-related behavior in adolescence. Rat dams were fed a control (C) or an obesogenic high-fat high-sugar diet (OD) for nine weeks prior to mating and throughout gestation and lactation. Maternal obesity differentially altered the CB1R in the hypothalamus of neonate offspring, with significant increase in male but not in female pups, associated with decreased CB2R prior to obesity development. In adolescence, maternal obesity induced anxiety-like behavior only in adolescent females which was associated with increased content of CB1R in the dorsal hippocampus. Our findings suggest that the early origins of anxiety disorders induced by maternal exposome is associated with dysregulation of the brain ECS, with females being more susceptible.
Collapse
Affiliation(s)
- Camila Calvino
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana Macedo de Almeida
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Minas Gerais, Brazil
| | - Clara Figueredo Reis-Gomes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Brenda da Silva Andrade
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gilda Angela Neves
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Isis Hara Trevenzoli
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
2
|
Vasil TM, Fleury ES, Walker ED, Kuiper JR, Buckley JP, Cecil KM, Chen A, Kalkwarf HJ, Lanphear BP, Yolton K, Braun JM. Associations of pre- and postnatal per- and polyfluoroalkyl substance exposure with adolescents' eating behaviors. Environ Epidemiol 2024; 8:e343. [PMID: 39555184 PMCID: PMC11567689 DOI: 10.1097/ee9.0000000000000343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/02/2024] [Indexed: 11/19/2024] Open
Abstract
Background Per- and polyfluoroalkyl substances (PFAS), persistent environmental chemicals, may act as obesogens by interacting with neuroendocrine pathways regulating energy homeostasis and satiety signals influencing adolescent eating behaviors. Methods In 211 HOME Study adolescents (Cincinnati, OH; recruited 2003-2006), we measured PFAS concentrations in serum collected during pregnancy, at delivery, and at ages 3, 8, and 12 years. Caregivers completed the Child Eating Behavior Questionnaire (CEBQ) at age 12, and we calculated food approach and food avoidance scores. Using quantile-based g-computation, we estimated covariate-adjusted associations between a mixture of four gestational PFAS and CEBQ scores. We identified high (n = 76, 36%) and low (n = 135, 64%) longitudinal PFAS mixture exposure profiles between delivery and age 12 years using latent profile analysis and related these to CEBQ scores. We examined whether child sex or physical activity modified these associations. Results We observed no association of gestational PFAS mixture with food approach or food avoidance scores. Children in the higher longitudinal PFAS mixture profile had slightly higher food approach scores (β: 0.47, 95% CI: -0.27, 1.23) and similar food avoidance scores (β: -0.15, 95% CI: -0.75, 0.46) compared with children in the lower profile. We found some evidence that higher physical activity favorably modified the association between longitudinal PFAS mixture profiles and emotional overeating (interaction P value = 0.13). Child sex did not consistently modify any associations. Conclusions Serum PFAS concentrations were not consistently linked to adolescent eating behaviors in this study, suggesting alternative pathways, such as metabolic rate, may underlie previously observed associations between PFAS exposure and childhood obesity.
Collapse
Affiliation(s)
| | - Elvira S. Fleury
- Department of Epidemiology, Brown University, Providence, Rhode Island
| | - Erica D. Walker
- Department of Epidemiology, Brown University, Providence, Rhode Island
| | - Jordan R. Kuiper
- Department of Environmental and Occupational Health, The George Washington University Milken Institute School of Public Health, Washington, District of Columbia
| | - Jessie P. Buckley
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina
| | - Kim M. Cecil
- Department of Radiology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Aimin Chen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Heidi J. Kalkwarf
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Bruce P. Lanphear
- Faculty of Health Sciences, Simon Fraser University, Vancouver, British Columbia, Canada
| | - Kimberly Yolton
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Joseph M. Braun
- Department of Epidemiology, Brown University, Providence, Rhode Island
| |
Collapse
|
3
|
Sullivan EL, Molloy KR, Dunn GA, Balanzar AL, Young AS, Loftis JM, Ablow JC, Nigg JT, Gustafsson HC. Adipokines measured during pregnancy and at birth are associated with infant negative affect. Brain Behav Immun 2024; 120:34-43. [PMID: 38772428 PMCID: PMC11401062 DOI: 10.1016/j.bbi.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 03/18/2024] [Accepted: 05/18/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Increased adiposity during pregnancy may be related to offspring risk for mental health disorders, although the biological mechanisms are poorly understood. One promising hypothesis is that factors secreted from adipocytes such as leptin and adiponectin may explain this association. The current study examined whether pregnancy or umbilical cord blood concentrations of leptin and/or adiponectin a) predict elevated infant negative affect at 6 months (an early life marker of risk for psychopathology); and b) help explain the association between pregnancy adiposity and increased infant negative affect. METHODS Data came from a prospective cohort (N = 305) of pregnant individuals and their offspring. Second trimester adiposity was assessed using air displacement plethysmography. Concentrations of leptin and adiponectin were measured in second trimester plasma and umbilical cord plasma. Infant negative affect was assessed by standardized observation at 6 months. Second trimester inflammation was assessed using a comprehensive panel of cytokines. RESULTS Lower second trimester adiponectin was associated with elevated infant negative affect, and mediated the effect of pregnancy adiposity on infant negative affect. This association was independent of the effect of second trimester inflammation. Umbilical cord leptin also predicted higher infant negative affect and mediated the association between pregnancy adiposity and infant negative affect. CONCLUSIONS This is the first study to link pregnancy adiponectin or cord blood leptin to infant markers of risk for psychopathology, and the first to demonstrate that these adipokines mediate the association between pregnancy adiposity and offspring behavioral outcomes, suggesting novel markers of risk and potential mechanisms of effect.
Collapse
Affiliation(s)
- Elinor L Sullivan
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States; Division of Neuroscience, Oregon National Primary Research Center, United States.
| | - Kelly R Molloy
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| | - Geoffrey A Dunn
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| | - Adriana L Balanzar
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| | - Anna S Young
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| | - Jennifer M Loftis
- Department of Psychiatry, Oregon Health & Science University, United States; VA Portland Health Care System, United States
| | | | - Joel T Nigg
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| | - Hanna C Gustafsson
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| |
Collapse
|
4
|
Sharma Y, Galvão AM. Maternal obesity and ovarian failure: is leptin the culprit? Anim Reprod 2023; 19:e20230007. [PMID: 36855701 PMCID: PMC9968511 DOI: 10.1590/1984-3143-ar2023-0007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 01/24/2023] [Indexed: 02/22/2023] Open
Abstract
At the time of its discovery and characterization in 1994, leptin was mostly considered a metabolic hormone able to regulate body weight and energy homeostasis. However, in recent years, a great deal of literature has revealed leptin's pleiotropic nature, through its involvement in numerous physiological contexts including the regulation of the female reproductive tract and ovarian function. Obesity has been largely associated with infertility, and leptin signalling is known to be dysregulated in the ovaries of obese females. Hence, the disruption of ovarian leptin signalling was shown to contribute to the pathophysiology of ovarian failure in obese females, affecting transcriptional programmes in the gamete and somatic cells. This review attempts to uncover the underlying mechanisms contributing to female infertility associated with obesity, as well as to shed light on the role of leptin in the metabolic dysregulation within the follicle, the effects on the oocyte epigenome, and the potential long-term consequence to embryo programming.
Collapse
Affiliation(s)
- Yashaswi Sharma
- Institute of Animal Reproduction and Food Research of PAS, Department of Reproductive Immunology and Pathology, Olsztyn, Poland
| | - António Miguel Galvão
- Institute of Animal Reproduction and Food Research of PAS, Department of Reproductive Immunology and Pathology, Olsztyn, Poland,Babraham Institute, Epigenetics Programme, Cambridge, United Kingdom UK,Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom UK,Corresponding author: ;
| |
Collapse
|
5
|
Regensburger M, Rasul Chaudhry S, Yasin H, Zhao Y, Stadlbauer A, Buchfelder M, Kinfe T. Emerging roles of leptin in Parkinson's disease: Chronic inflammation, neuroprotection and more? Brain Behav Immun 2023; 107:53-61. [PMID: 36150585 DOI: 10.1016/j.bbi.2022.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/22/2022] [Accepted: 09/16/2022] [Indexed: 12/13/2022] Open
Abstract
An increasing body of experimental evidence implicates a relationship between immunometabolic deterioration and the progression of Parkinson's disease (PD) with a dysregulation of central and peripheral neuroinflammatory networks mediated by circulating adipokines, in particular leptin. We screened the current literature on the role of adipokines in PD. Hence, we searched known databases (PubMed, MEDLINE/OVID) and reviewed original and review articles using the following terms: "leptin/ObR", "Parkinson's disease", "immune-metabolism", "biomarkers" and "neuroinflammation". Focusing on leptin, we summarize and discuss the existing in vivo and in vitro evidence on how adipokines may be protective against neurodegeneration, but at the same time contribute to the progression of PD. These components of the adipose brain axis represent a hitherto underestimated pathway to study systemic influences on dopaminergic degeneration. In addition, we give a comprehensive update on the potential of adjunctive therapeutics in PD targeting leptin, leptin-receptors, and associated pathways. Further experimental and clinical trials are needed to elucidate the mechanisms of action and the value of central and peripheral adipose-immune-metabolism molecular phenotyping in order to develop and validate the differential roles of different adipokines as potential therapeutic target for PD patients.
Collapse
Affiliation(s)
- Martin Regensburger
- Department of Molecular Neurology, Friedrich-Alexander University (FAU), Erlangen-Nürnberg, 91054 Erlangen, Germany; Center for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, 91054 Erlangen, Germany
| | - Shafqat Rasul Chaudhry
- Obaid Noor Institute of Medical Sciences (ONIMS), Mianwali, Pakistan; Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, 44000 Islamabad, Pakistan
| | - Hammad Yasin
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, 44000 Islamabad, Pakistan
| | - Yining Zhao
- Department of Neurosurgery, Friedrich-Alexander University (FAU), Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Andreas Stadlbauer
- Department of Neurosurgery, Friedrich-Alexander University (FAU), Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Michael Buchfelder
- Department of Neurosurgery, Friedrich-Alexander University (FAU), Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Thomas Kinfe
- Division of Functional Neurosurgery and Stereotaxy, Friedrich-Alexander University (FAU), Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|
6
|
Mitchell AJ, Khambadkone SG, Dunn G, Bagley J, Tamashiro KLK, Fair D, Gustafsson H, Sullivan EL. Maternal Western-style diet reduces social engagement and increases idiosyncratic behavior in Japanese macaque offspring. Brain Behav Immun 2022; 105:109-121. [PMID: 35809877 PMCID: PMC9987715 DOI: 10.1016/j.bbi.2022.07.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/29/2022] [Accepted: 07/03/2022] [Indexed: 01/27/2023] Open
Abstract
Recent evidence in humans and animals indicates an association between maternal obesity and offspring behavioral outcomes. In humans, increased maternal body mass index has been linked to an increased risk of children receiving a diagnosis of early-emerging neurodevelopmental disorders such as Attention Deficit/Hyperactivity Disorder (ADHD) and/or Autism Spectrum Disorder (ASD). However, a limited number of preclinical studies have examined associations between maternal Western-Style Diet (mWSD) exposure and offspring social behavior. To our knowledge, this is the first study to investigate relationships between mWSD exposure and social behavior in non-human primates. Since aberrant social behavior is a diagnostic criterion for several neurodevelopmental disorders, the current study focuses on examining the influence of maternal nutrition and metabolic state on offspring social behavior in Japanese macaques (Macaca fuscata). We found that mWSD offspring initiated less affiliative social behaviors as well as proximity to a peer. Using path analysis, we found that the association between mWSD consumption and reduced offspring social engagement was statistically mediated by increased maternal interleukin (IL)-12 during the third trimester of pregnancy. Additionally, mWSD offspring displayed increased idiosyncratic behavior, which was related to alterations in maternal adiposity and leptin in the third trimester. Together, these results suggest that NHP offspring exposed to mWSD exhibit behavioral phenotypes similar to what is described in some early-emerging neurodevelopmental disorders. These results provide evidence that mWSD exposure during gestation may be linked to increased risk of neurodevelopmental disorders and provides targets for prevention and intervention efforts.
Collapse
Affiliation(s)
- A J Mitchell
- Oregon National Primate Research Center, Division of Neuroscience, Beaverton, OR, USA; Oregon Health & Science University, Department of Behavioral Neuroscience, Portland, OR, USA
| | - Seva G Khambadkone
- Johns Hopkins University, School of Medicine, Department of Psychiatry & Behavioral Sciences, Baltimore, MD, USA
| | - Geoffrey Dunn
- University of Oregon, Department of Human Physiology, Eugene, OR, USA
| | - Jennifer Bagley
- Oregon National Primate Research Center, Division of Neuroscience, Beaverton, OR, USA
| | - Kellie L K Tamashiro
- Johns Hopkins University, School of Medicine, Department of Psychiatry & Behavioral Sciences, Baltimore, MD, USA
| | - Damien Fair
- University of Minnesota School of Medicine, Masonic Institute of Child Development, Minneapolis, MN, USA
| | - Hanna Gustafsson
- Oregon Health & Science University, Department of Psychiatry, Portland, OR, USA
| | - Elinor L Sullivan
- Oregon National Primate Research Center, Division of Neuroscience, Beaverton, OR, USA; Oregon Health & Science University, Department of Behavioral Neuroscience, Portland, OR, USA; University of Oregon, Department of Human Physiology, Eugene, OR, USA; Oregon Health & Science University, Department of Psychiatry, Portland, OR, USA.
| |
Collapse
|
7
|
Abstract
Leptin is a hormone primarily produced by the adipose tissue in proportion to the size of fat stores, with a primary function in the control of lipid reserves. Besides adipose tissue, leptin is also produced by other tissues, such as the stomach, placenta, and mammary gland. Altogether, leptin exerts a broad spectrum of short, medium, and long-term regulatory actions at the central and peripheral levels, including metabolic programming effects that condition the proper development and function of the adipose organ, which are relevant for its main role in energy homeostasis. Comprehending how leptin regulates adipose tissue may provide important clues to understand the pathophysiology of obesity and related diseases, such as type 2 diabetes, as well as its prevention and treatment. This review focuses on the physiological and long-lasting regulatory effects of leptin on adipose tissue, the mechanisms and pathways involved, its main outcomes on whole-body physiological homeostasis, and its consequences on chronic diseases.
Collapse
Affiliation(s)
- Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands. CIBER de Fisiopatología de La Obesidad Y Nutrición (CIBEROBN). Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Mariona Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands. CIBER de Fisiopatología de La Obesidad Y Nutrición (CIBEROBN). Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Catalina Amadora Pomar
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands. CIBER de Fisiopatología de La Obesidad Y Nutrición (CIBEROBN). Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Ana María Rodríguez
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands. CIBER de Fisiopatología de La Obesidad Y Nutrición (CIBEROBN). Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands. CIBER de Fisiopatología de La Obesidad Y Nutrición (CIBEROBN). Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| |
Collapse
|
8
|
Vickers MH. Early life nutrition and neuroendocrine programming. Neuropharmacology 2021; 205:108921. [PMID: 34902348 DOI: 10.1016/j.neuropharm.2021.108921] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/26/2022]
Abstract
Alterations in the nutritional environment in early life can significantly increase the risk for obesity and a range of development of metabolic disorders in offspring in later life, effects that can be passed onto future generations. This process, termed development programming, provides the framework of the developmental origins of health and disease (DOHaD) paradigm. Early life nutritional compromise including undernutrition, overnutrition or specific macro/micronutrient deficiencies, results in a range of adverse health outcomes in offspring that can be further exacerbated by a poor postnatal nutritional environment. Although the mechanisms underlying programming remain poorly defined, a common feature across the phenotypes displayed in preclinical models is that of altered wiring of neuroendocrine circuits that regulate satiety and energy balance. As such, altered maternal nutritional exposures during critical early periods of developmental plasticity can result in aberrant hardwiring of these circuits with lasting adverse consequences for the offspring. There is also increasing evidence around the role of an altered epigenome and the gut-brain axis in mediating some of the central programming effects observed. Further, although such programming was once considered to result in a permanent change in developmental trajectory, there is evidence, at least from preclinical models, that programming can be reversed via targeted nutritional manipulations during early development. Further work is required at a mechanistic level to allow for identification for early markers of later disease risk, delineation of sex-specific effects and pathways to implementation of strategies aimed at breaking the transgenerational transmission of disease.
Collapse
Affiliation(s)
- M H Vickers
- Liggins Institute, University of Auckland, 85 Park Road, Grafton, Auckland, 1142, New Zealand.
| |
Collapse
|
9
|
Vasistha NA, Khodosevich K. The impact of (ab)normal maternal environment on cortical development. Prog Neurobiol 2021; 202:102054. [PMID: 33905709 DOI: 10.1016/j.pneurobio.2021.102054] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/01/2021] [Accepted: 04/20/2021] [Indexed: 12/24/2022]
Abstract
The cortex in the mammalian brain is the most complex brain region that integrates sensory information and coordinates motor and cognitive processes. To perform such functions, the cortex contains multiple subtypes of neurons that are generated during embryogenesis. Newly born neurons migrate to their proper location in the cortex, grow axons and dendrites, and form neuronal circuits. These developmental processes in the fetal brain are regulated to a large extent by a great variety of factors derived from the mother - starting from simple nutrients as building blocks and ending with hormones. Thus, when the normal maternal environment is disturbed due to maternal infection, stress, malnutrition, or toxic substances, it might have a profound impact on cortical development and the offspring can develop a variety of neurodevelopmental disorders. Here we first describe the major developmental processes which generate neuronal diversity in the cortex. We then review our knowledge of how most common maternal insults affect cortical development, perturb neuronal circuits, and lead to neurodevelopmental disorders. We further present a concept of selective vulnerability of cortical neuronal subtypes to maternal-derived insults, where the vulnerability of cortical neurons and their progenitors to an insult depends on the time (developmental period), place (location in the developing brain), and type (unique features of a cell type and an insult). Finally, we provide evidence for the existence of selective vulnerability during cortical development and identify the most vulnerable neuronal types, stages of differentiation, and developmental time for major maternal-derived insults.
Collapse
Affiliation(s)
- Navneet A Vasistha
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| | - Konstantin Khodosevich
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| |
Collapse
|
10
|
Kim JG, Lee BJ, Jeong JK. Temporal Leptin to Determine Cardiovascular and Metabolic Fate throughout the Life. Nutrients 2020; 12:nu12113256. [PMID: 33114326 PMCID: PMC7690895 DOI: 10.3390/nu12113256] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 01/01/2023] Open
Abstract
Leptin links peripheral adiposity and the central nervous system (CNS) to regulate cardiometabolic physiology. Within the CNS, leptin receptor-expressing cells are a counterpart to circulating leptin, and leptin receptor-mediated neural networks modulate the output of neuroendocrine and sympathetic nervous activity to balance cardiometabolic homeostasis. Therefore, disrupted CNS leptin signaling is directly implicated in the development of metabolic diseases, such as hypertension, obesity, and type 2 diabetes. Independently, maternal leptin also plays a central role in the development and growth of the infant during gestation. Accumulating evidence points to the dynamic maternal leptin environment as a predictor of cardiometabolic fate in their offspring as it is directly associated with infant metabolic parameters at birth. In postnatal life, the degree of serum leptin is representative of the level of body adiposity/weight, a driving factor for cardiometabolic alterations, and therefore, the levels of blood leptin through the CNS mechanism, in a large part, are a strong determinant for future cardiometabolic fate. The current review focuses on highlighting and discussing recent updates for temporal dissection of leptin-associated programing of future cardiometabolic fate throughout the entire life.
Collapse
Affiliation(s)
- Jae Geun Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea;
- Institute for New Drug Development, Division of Life Sciences, Incheon National University, Incheon 22012, Korea
| | - Byung Ju Lee
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 44610, Korea
- Correspondence: (B.J.L.); (J.K.J.); Tel.: +82-52-259-2351 (B.J.L.); +1-202-994-9815 (J.K.J.)
| | - Jin Kwon Jeong
- Department of Pharmacology and Physiology, School of Medicine & Health Sciences, The George Washington University, Washington, DC 20037, USA
- Correspondence: (B.J.L.); (J.K.J.); Tel.: +82-52-259-2351 (B.J.L.); +1-202-994-9815 (J.K.J.)
| |
Collapse
|
11
|
Yau-Qiu ZX, Picó C, Rodríguez AM, Palou A. Leptin Distribution in Rat Foetal and Extraembryonic Tissues in Late Gestation: A Physiological View of Amniotic Fluid Leptin. Nutrients 2020; 12:E2542. [PMID: 32825787 PMCID: PMC7551401 DOI: 10.3390/nu12092542] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 12/14/2022] Open
Abstract
Prenatal leptin is key to regulating foetal growth and early metabolic programming. The presence of intact leptin in rat foetal (at late gestation) and neonatal (immediately after birth) stomach content and mucosa has been previously described, suggesting that it may act as a regulatory nutrient for the neonate rats, be internalised by the stomach, and play a physiological role early in life, which requires to be further investigated, including its origin. We aimed to study the ontogeny of the presence of leptin in the foetal stomach and key extraembryonic tissues in rats at late gestation (days 18-21). Leptin concentration was determined by enzyme-linked immunosorbent assay, and placental leptin immunolocalisation was analysed by immunohistochemistry. Leptin showed a sudden appearance in the amniotic fluid (AF) at day 20 of gestation, gastric content (swallowed AF), stomach, and umbilical cord, significantly increasing at day 21. Leptin levels in these fluids and tissues were positively correlated. In the placenta, leptin was detectable at all the studied days, but its localisation changed from widespread throughout the placenta at day 18 to well-defined in the labyrinth zone from day 19 onwards. The results support a possible internalisation of AF leptin by the immature stomach of near-term foetuses and suggest that changes in placental leptin localisation might help to explain the sudden appearance of leptin in AF at gestational day 20, with potential physiological significance regarding short-term feeding control and metabolic programming in the developing offspring.
Collapse
Affiliation(s)
- Zhi Xin Yau-Qiu
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics and Obesity), University of the Balearic Islands (UIB), Palma de Mallorca, 07122 Balearic Islands, Spain; (Z.X.Y.-Q.); (C.P.); (A.P.)
- Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, 07010 Balearic Islands, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Palma de Mallorca, 07122 Balearic Islands, Spain
| | - Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics and Obesity), University of the Balearic Islands (UIB), Palma de Mallorca, 07122 Balearic Islands, Spain; (Z.X.Y.-Q.); (C.P.); (A.P.)
- Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, 07010 Balearic Islands, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Palma de Mallorca, 07122 Balearic Islands, Spain
| | - Ana María Rodríguez
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics and Obesity), University of the Balearic Islands (UIB), Palma de Mallorca, 07122 Balearic Islands, Spain; (Z.X.Y.-Q.); (C.P.); (A.P.)
- Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, 07010 Balearic Islands, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Palma de Mallorca, 07122 Balearic Islands, Spain
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics and Obesity), University of the Balearic Islands (UIB), Palma de Mallorca, 07122 Balearic Islands, Spain; (Z.X.Y.-Q.); (C.P.); (A.P.)
- Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, 07010 Balearic Islands, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Palma de Mallorca, 07122 Balearic Islands, Spain
| |
Collapse
|
12
|
Easterling MR, Engbrecht KM, Crespi EJ. Endocrine Regulation of Epimorphic Regeneration. Endocrinology 2019; 160:2969-2980. [PMID: 31593236 DOI: 10.1210/en.2019-00321] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 10/01/2019] [Indexed: 12/16/2022]
Abstract
Studies aiming to uncover primary mechanisms of regeneration have predominantly focused on genetic pathways regulating specific stages in the regeneration process: wound healing, blastema formation, and pattern formation. However, studies across organisms show that environmental conditions and the physiological state of the animal can affect the rate or quality of regeneration, and endocrine signals are likely the mediators of these effects. Endocrine signals acting directly on receptors expressed in the tissue or via neuroendocrine pathways can affect regeneration by regulating the immune response to injury, allocation of energetic resources, or by enhancing or inhibiting proliferation and differentiation pathways involved in regeneration. This review discusses the cumulative knowledge in the literature about endocrine regulation of regeneration and its importance in future research to advance biomedical research.
Collapse
Affiliation(s)
- Marietta R Easterling
- School of Biological Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington
| | - Kristin M Engbrecht
- School of Biological Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington
- Pacific Northwest National Laboratory, Richland, Washington
| | - Erica J Crespi
- School of Biological Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington
| |
Collapse
|
13
|
Westwater ML, Vilar-López R, Ziauddeen H, Verdejo-García A, Fletcher PC. Combined effects of age and BMI are related to altered cortical thickness in adolescence and adulthood. Dev Cogn Neurosci 2019; 40:100728. [PMID: 31751856 PMCID: PMC6913515 DOI: 10.1016/j.dcn.2019.100728] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 10/31/2019] [Accepted: 11/03/2019] [Indexed: 12/17/2022] Open
Abstract
Overweight and obesity are associated with functional and structural alterations in the brain, but how these associations change across critical developmental periods remains unknown. Here, we examined the relationship between age, body mass index (BMI) and cortical thickness (CT) in healthy adolescents (n = 70; 14-19 y) and adults (n = 75; 25-45 y). We also examined the relationship between adiposity, impulsivity, measured by delay discounting (DD), and CT of the inferior frontal gyrus (IFG), a region key to impulse control. A significant age-by-BMI interaction was observed in both adolescents and adults; however, the direction of this relationship differed between age groups. In adolescents, increased age-adjusted BMI Z-score attenuated age-related CT reductions globally and in frontal, temporal and occipital regions. In adults, increased BMI augmented age-related CT reductions, both globally and in bilateral parietal cortex. Although DD was unrelated to adiposity in both groups, increased DD and adiposity were both associated with reduced IFG thickness in adolescents and adults. Our findings suggest that the known age effects on CT in adolescence and adulthood are moderated by adiposity. The association between weight, cortical development and its functional implications would suggest that future studies of adolescent and adult brain development take adiposity into account.
Collapse
Affiliation(s)
- Margaret L Westwater
- Department of Psychiatry, University of Cambridge, Herchel Smith Building, Addenbrooke's Hospital, Cambridge CB2 0SZ, UK.
| | - Raquel Vilar-López
- Mind, Brain and Behavior Research Center, Universidad de Granada, Granada, Spain
| | - Hisham Ziauddeen
- Department of Psychiatry, University of Cambridge, Herchel Smith Building, Addenbrooke's Hospital, Cambridge CB2 0SZ, UK; Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Cambridgeshire and Peterborough Foundation Trust, Cambridge, CB21 5EF, UK
| | - Antonio Verdejo-García
- School of Psychological Sciences and Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Melbourne, Australia
| | - Paul C Fletcher
- Department of Psychiatry, University of Cambridge, Herchel Smith Building, Addenbrooke's Hospital, Cambridge CB2 0SZ, UK; Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Cambridgeshire and Peterborough Foundation Trust, Cambridge, CB21 5EF, UK
| |
Collapse
|
14
|
Denisova EI, Kozhevnikova VV, Bazhan NM, Makarova EN. Sex-specific effects of leptin administration to pregnant mice on the placentae and the metabolic phenotypes of offspring. FEBS Open Bio 2019; 10:96-106. [PMID: 31703240 PMCID: PMC6943234 DOI: 10.1002/2211-5463.12757] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/15/2019] [Accepted: 11/07/2019] [Indexed: 12/19/2022] Open
Abstract
Obesity during pregnancy has been shown to increase the risk of metabolic diseases in the offspring. However, the factors within the maternal milieu which affect offspring phenotypes and the underlying mechanisms remain unknown. The adipocyte hormone leptin plays a key role in regulating energy homeostasis and is known to participate in sex‐specific developmental programming. To examine the action of leptin on fetal growth, placental gene expression and postnatal offspring metabolism, we injected C57BL mice with leptin or saline on gestational day 12 and then measured body weights (BWs) of offspring fed on a standard or obesogenic diet, as well as mRNA expression levels of insulin‐like growth factors and glucose and amino acid transporters. Male and female offspring born to leptin‐treated mothers exhibited growth retardation before and a growth surge after weaning. Mature male offspring, but not female offspring, exhibited increased BWs on a standard diet. Leptin administration prevented the development of hyperglycaemia in the obese offspring of both sexes. The placentas of the male and female foetuses differed in size and gene expression, and leptin injection decreased the fetal weights of both sexes, the placental weights of the male foetuses and placental gene expression of the GLUT1 glucose transporter in female foetuses. The data suggest that mid‐pregnancy is an ontogenetic window for the sex‐specific programming effects of leptin, and these effects may be exerted via fetal sex‐specific placental responses to leptin administration.
Collapse
Affiliation(s)
- Elena I Denisova
- Laboratory of Physiological Genetics, Institute of Cytology and Genetics, the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Valeria V Kozhevnikova
- Laboratory of Physiological Genetics, Institute of Cytology and Genetics, the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Nadezhda M Bazhan
- Laboratory of Physiological Genetics, Institute of Cytology and Genetics, the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Department of Physiology, Novosibirsk State University, Novosibirsk, Russia
| | - Elena N Makarova
- Laboratory of Physiological Genetics, Institute of Cytology and Genetics, the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
15
|
Karakosta P, Margetaki K, Fthenou E, Kampouri M, Kyriklaki A, Koutra K, Chalkiadaki G, Roumeliotaki T, Vafeiadi M, Kogevinas M, Mantzoros C, Chatzi L. Cord Leptin is Associated with Neuropsychomotor Development in Childhood. Obesity (Silver Spring) 2019; 27:1693-1702. [PMID: 31479200 PMCID: PMC6756960 DOI: 10.1002/oby.22571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/06/2019] [Indexed: 11/10/2022]
Abstract
OBJECTIVE Leptin is critical for central nervous system development and maturation. This study aimed to evaluate the potential regulatory role of cord leptin in the neuropsychomotor development of children ages 18 months to 6 years. METHODS This study included 424 children from a prospective mother-child cohort (Rhea Study; Crete, Greece) with available cord leptin levels and data on neurodevelopmental outcomes at 18 months (Bayley Scales of Infant and Toddler Development, Third Edition), 4 years (McCarthy Scales of Children's Abilities), and 6 years (Raven's Coloured Progressive Matrices and Trail Making Test). Multivariable linear regression models were used to explore the associations. RESULTS Each 10-ng/mL increase in the cord leptin level was associated with increased scores on the gross motor scale at 18 months (β coefficient: 3.8; 95% CI: 0.0-7.5), with decreased scores in the general cognitive performance (β coefficient: -3.0; 95% CI: -5.5 to -0.4), perceptual performance (β coefficient: -3.4; 95% CI: -6.0 to -9.9), working memory (β coefficient: -3.1; 95% CI: -5.7 to -0.4), executive function (β coefficient -3.1; 95% CI: -5.7 to -0.5), and functions of the posterior cortex (β coefficient: -2.7; 95% CI: -5.2 to -0.1) scales at 4 years, and with a 3.7-unit decrease in the Raven's Coloured Progressive Matrices score at 6 years (β coefficient: -3.7; 95% CI: -6.9 to -0.5). CONCLUSIONS Increased cord leptin levels are associated with enhanced gross motor development at 18 months but decreased cognitive performance in early and middle childhood.
Collapse
Affiliation(s)
- Polyxeni Karakosta
- Department of Social Medicine, School of Medicine,
University of Crete, Heraklion, Greece
- Clinical Microbiology Laboratory, Attikon University
Hospital, National and Kapodistrian University of Athens, Greece
| | - Katerina Margetaki
- Department of Social Medicine, School of Medicine,
University of Crete, Heraklion, Greece
| | - Eleni Fthenou
- Qatar Biobank for Medical Research, Qatar Foundation for
Education, Science and Community, Doha, Doha, Qatar
| | - Mariza Kampouri
- Department of Social Medicine, School of Medicine,
University of Crete, Heraklion, Greece
| | - Andriani Kyriklaki
- Department of Social Medicine, School of Medicine,
University of Crete, Heraklion, Greece
| | - Katerina Koutra
- Department of Psychology, School of Social Sciences,
University of Crete, Rethymnon, Greece
| | - Georgia Chalkiadaki
- Department of Social Medicine, School of Medicine,
University of Crete, Heraklion, Greece
| | - Theano Roumeliotaki
- Department of Social Medicine, School of Medicine,
University of Crete, Heraklion, Greece
| | - Marina Vafeiadi
- Department of Social Medicine, School of Medicine,
University of Crete, Heraklion, Greece
| | - Manolis Kogevinas
- ISGlobal, Centre for Research in Environmental
Epidemiology (CREAL), Barcelona, Spain
| | - Christos Mantzoros
- Division of Endocrinology, Diabetes, and Metabolism,
Department of Medicine, Beth Israel, Deaconess Medical Center, Harvard Medical
School, Boston, Massachusetts, United States
| | - Lida Chatzi
- Department of Preventive Medicine, University of Southern
California, Los Angeles, CA, United States
| |
Collapse
|
16
|
Sjögren M, Soylu-Kucharz R, Dandunna U, Stan TL, Cavalera M, Sandelius Å, Zetterberg H, Björkqvist M. Leptin deficiency reverses high metabolic state and weight loss without affecting central pathology in the R6/2 mouse model of Huntington's disease. Neurobiol Dis 2019; 132:104560. [PMID: 31419548 DOI: 10.1016/j.nbd.2019.104560] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 06/13/2019] [Accepted: 07/30/2019] [Indexed: 11/18/2022] Open
Abstract
Body weight has been shown to be a predictor of clinical progression in Huntington's disease (HD). Alongside widespread neuronal pathology, both HD patients and the R6/2 mouse model of HD exhibit weight loss and increased energy expenditure, providing a rationale for targeting whole-body energy metabolism in HD. Leptin-deficient mice display low energy expenditure and increased body weight. We therefore hypothesized that normalizing energy metabolism in R6/2 mice, utilizing leptin- deficiency, would lead to a slower disease progression in the R6/2 mouse. In this study, we show that R6/2 mice on a leptin-deficient genetic background display increased body weight and increased fat mass compared to R6/2 mice, as well as wild type littermates. The increased body weight was accompanied by low energy expenditure, illustrated by a reduction in respiratory exchange rate. Leptin-deficient R6/2 mice had large white adipocytes with white adipocyte gene expression characteristics, in contrast to white adipose tissue in R6/2 mice, where white adipose tissue showed signs of browning. Leptin-deficient R6/2 mice did not exhibit improved neuropathological measures. Our results indicate that lowering energy metabolism in HD, by increasing fat mass and reducing respiratory exchange rate, is not sufficient to affect neuropathology. Further studies targeting energy metabolism in HD are warranted.
Collapse
Affiliation(s)
- Marie Sjögren
- Wallenberg Neuroscience Center, Brain Disease Biomarker Unit, Department of Experimental Medical Sciences, Lund University, Lund, Sweden.
| | - Rana Soylu-Kucharz
- Wallenberg Neuroscience Center, Brain Disease Biomarker Unit, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Unali Dandunna
- Wallenberg Neuroscience Center, Brain Disease Biomarker Unit, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Tiberiu Loredan Stan
- Wallenberg Neuroscience Center, Brain Disease Biomarker Unit, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Michele Cavalera
- Department of Clinical Sciences, Cardiovascular Research, Translational Studies, Lund University, Malmö, Sweden
| | - Åsa Sandelius
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom; UK Dementia Research Institute at UCL, London, United Kingdom
| | - Maria Björkqvist
- Wallenberg Neuroscience Center, Brain Disease Biomarker Unit, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
17
|
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
|
18
|
Yay A, Onder GO, Ozdamar S, Bahadir A, Aytekin M, Baran M. The Effects of Leptin on Rat Brain Development; An Experimental Study. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-018-09803-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
19
|
Li N, Arbuckle TE, Muckle G, Lanphear BP, Boivin M, Chen A, Dodds L, Fraser WD, Ouellet E, Séguin JR, Velez MP, Yolton K, Braun JM. Associations of cord blood leptin and adiponectin with children's cognitive abilities. Psychoneuroendocrinology 2019; 99:257-264. [PMID: 30390444 PMCID: PMC6239208 DOI: 10.1016/j.psyneuen.2018.10.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022]
Abstract
Background Adipocytokines may play a role in fetal programming of neurodevelopment. We aimed to investigate the associations between cord blood adipocytokine concentrations and children's intelligence test scores. Methods We used data from two ongoing pregnancy cohorts in North America: the Maternal-Infant Research on Environmental Chemicals (MIREC, n = 429) and Health Outcomes and Measures of the Environment (HOME, n = 183) Studies. Umbilical cord blood adipocytokine concentrations were measured using enzyme-linked immunosorbent assays. We assessed children's Intelligence Quotient (IQ) and its components using the Wechsler Preschool and Primary Scales of Intelligence-III or Wechsler Intelligence Scale for Children-IV. We used linear regression and linear mixed models to estimate associations between log2-transformed adipocytokine concentrations and children's IQ after adjusting for sociodemographic, perinatal, and child factors. Results After adjusting for covariates, cord blood adiponectin was positively associated with children's full-scale IQ scores at age 3 years in the MIREC Study (β = 1.4, 95% confidence interval [CI]: 0.2, 2.5) and at ages 5 and 8 years in the HOME Study (β = 1.7, CI: -0.1, 3.5). Adiponectin was positively associated with performance IQ in both studies (MIREC: β = 2.0, CI: 0.7, 3.3; HOME: β = 2.2, CI: 0.5, 3.9). Adiponectin was positively associated with working memory composite scores at age 8 in the HOME Study (β = 3.1, CI: 1.0, 5.2). Leptin was not associated with children's IQ in either study. Conclusions Cord blood adiponectin was associated with higher full-scale and performance IQ and working memory composite scores in children. Future studies are needed to explore the mechanisms underlying these associations.
Collapse
Affiliation(s)
- Nan Li
- Department of Epidemiology, Brown University, Providence, RI, United States.
| | - Tye E Arbuckle
- Population Studies Division, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario, Canada
| | - Gina Muckle
- School of Psychology, Laval University, Ville de Québec, Québec, Canada
| | - Bruce P Lanphear
- Faculty of Health Sciences, Simon Fraser University, British Columbia, Canada; Child and Family Research Institute, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Michel Boivin
- School of Psychology, Laval University, Ville de Québec, Québec, Canada
| | - Aimin Chen
- Division of Epidemiology, Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Linda Dodds
- Perinatal Epidemiology Research Unit, IWK Health Center, Halifax, Canada
| | - William D Fraser
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Mother and Child University Hospital Center, Montreal, Québec, Canada; Centre de recherche du CHUS (CHU de Sherbrooke), University of Sherbrooke, Sherbrooke, Québec, Canada
| | - Emmanuel Ouellet
- CHU de Québec-Université Laval Research Center, Ville de Québec, Québec, Canada
| | - Jean R Séguin
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Mother and Child University Hospital Center, Montreal, Québec, Canada; Department of Psychiatry, University of Montréal, Montréal, Québec, Canada
| | - Maria P Velez
- Department of Obstetrics and Gynecology, Queen's University, Kingston, Ontario, Canada
| | - Kimberly Yolton
- Department of Pediatrics, Division of General and Community Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Joseph M Braun
- Department of Epidemiology, Brown University, Providence, RI, United States
| |
Collapse
|
20
|
Bender MC, Sifuentes CJ, Denver RJ. Leptin Induces Mitosis and Activates the Canonical Wnt/β-Catenin Signaling Pathway in Neurogenic Regions of Xenopus Tadpole Brain. Front Endocrinol (Lausanne) 2017; 8:99. [PMID: 28533765 PMCID: PMC5421298 DOI: 10.3389/fendo.2017.00099] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/20/2017] [Indexed: 12/16/2022] Open
Abstract
In addition to its well-known role as an adipostat in adult mammals, leptin has diverse physiological and developmental actions in vertebrates. Leptin has been shown to promote development of hypothalamic circuits and to induce mitosis in different brain areas of mammals. We investigated the ontogeny of leptin mRNA, leptin actions on cell proliferation in the brain, and gene expression in the preoptic area/hypothalamus of tadpoles of Xenopus laevis. The level of leptin mRNA was low in premetamorphic tadpoles, but increased strongly at the beginning of metamorphosis and peaked at metamorphic climax. This increase in leptin mRNA at the onset of metamorphosis correlated with increased cell proliferation in the neurogenic zones of tadpole brain. We found that intracerebroventricular (i.c.v.) injection of recombinant Xenopus leptin (rxLeptin) in premetamorphic tadpoles strongly increased cell proliferation in neurogenic zones throughout the tadpole brain. We conducted gene expression profiling of genes induced at 2 h following i.c.v. injection of rxLeptin. This analysis identified 2,322 genes induced and 1,493 genes repressed by rxLeptin. The most enriched Kyoto Encyclopedia of Genes and Genomes term was the canonical Wnt/β-catenin pathway. Using electroporation-mediated gene transfer into tadpole brain of a reporter vector responsive to the canonical Wnt/β-catenin signaling pathway, we found that i.c.v. rxLeptin injection activated Wnt/β-catenin-dependent transcriptional activity. Our findings show that leptin acts on the premetamorphic tadpole brain to induce cell proliferation, possibly acting via the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Melissa Cui Bender
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Christopher J. Sifuentes
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Robert J. Denver
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
21
|
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: 71] [Impact Index Per Article: 8.9] [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
|
22
|
Association between obesity-related biomarkers and cognitive and motor development in infants. Behav Brain Res 2017; 325:12-16. [PMID: 28238825 DOI: 10.1016/j.bbr.2017.02.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/15/2017] [Accepted: 02/18/2017] [Indexed: 01/25/2023]
Abstract
BACKGROUND This study aimed to verify the association between obesity-related biomarkers and cognitive and motor development in infants between 6 and 24 months of age. METHODS A cross-sectional study was conducted with 50 infants and plasma levels of leptin, adiponectin, resistin, soluble tumor necrosis factor receptors 1 and 2 (sTNFR1 and sTNFR2), chemokines, brain-derived neurotrophic factor (BDNF), serum cortisol and redox status were measured. The Bayley-III test was utilized to evaluate cognitive and motor development, and multiple linear stepwise regression models were performed to verify the association between selected biomarkers and cognitive and motor development. RESULTS A significant association was found among plasma leptin and sTNFR1 levels with cognitive composite scores, and these two independents variables together explained 37% of the variability of cognitive composite scores (p=0.001). Only plasma sTNFR1 levels were associated and explained 24% of the variability of motor composite scores (p=0.003). CONCLUSIONS Plasma levels of sTNFR1 were associated with the increase in cognitive and motor development scores in infants between 6 and 24 months of age through a mechanism not directly related to excess body weight. Moreover, increase in plasma levels of leptin reduced the cognitive development in this age range.
Collapse
|
23
|
Fernandez-Martos CM, Atkinson RAK, Chuah MI, King AE, Vickers JC. Combination treatment with leptin and pioglitazone in a mouse model of Alzheimer's disease. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2016; 3:92-106. [PMID: 29067321 PMCID: PMC5651376 DOI: 10.1016/j.trci.2016.11.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Combination therapy approaches may be necessary to address the many facets of pathologic change in the brain in Alzheimer's disease (AD). The drugs leptin and pioglitazone have previously been shown individually to have neuroprotective and anti-inflammatory actions, respectively, in animal models. METHODS We studied the impact of combined leptin and pioglitazone treatment in 6-month-old APP/PS1 (APPswe/PSEN1dE9) transgenic AD mouse model. RESULTS We report that an acute 2-week treatment with combined leptin and pioglitazone resulted in a reduction of spatial memory deficits (Y maze) and brain β-amyloid levels (soluble β-amyloid and amyloid plaque burden) relative to vehicle-treated animals. Combination treatment was also associated with amelioration in plaque-associated neuritic pathology and synapse loss, and also a significantly reduced neocortical glial response. DISCUSSION Combination therapy with leptin and pioglitazone ameliorates pathologic changes in APP/PS1 mice and may represent a potential treatment approach for AD.
Collapse
Affiliation(s)
- Carmen M Fernandez-Martos
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Hobart, Tasmania, Australia
| | - Rachel A K Atkinson
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Hobart, Tasmania, Australia
| | - Meng I Chuah
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Hobart, Tasmania, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Hobart, Tasmania, Australia
| | - James C Vickers
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
24
|
Pugh SJ, Hutcheon JA, Richardson GA, Brooks MM, Himes KP, Day NL, Bodnar LM. Gestational weight gain, prepregnancy body mass index and offspring attention-deficit hyperactivity disorder symptoms and behaviour at age 10. BJOG 2016; 123:2094-2103. [PMID: 26996156 PMCID: PMC5031500 DOI: 10.1111/1471-0528.13909] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2015] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To assess offspring attention-deficit hyperactivity disorder (ADHD) symptoms and emotional/behavioural impairments at age 10 years in relation to maternal gestational weight gain (GWG) and prepregnancy body mass index (BMI). DESIGN AND SETTING Longitudinal birth cohort from Magee-Womens Hospital, Pittsburgh, Pennsylvania (enrolled 1983-86). POPULATION Mother-infant dyads (n = 511) were followed through pregnancy to 10 years. METHODS Self-reported total GWG was converted to gestational-age-standardised z-scores. Multivariable linear and negative binomial regressions were used to estimate effects of GWG and BMI on outcomes. MAIN OUTCOME MEASURES Child ADHD symptoms were assessed with the Conners' Continuous Performance Test. Child behaviour was assessed by parent and teacher ratings on the Child Behaviour Checklist (CBCL) and Teacher Report Form, respectively. RESULTS The mean (SD) total GWG (kg) was 14.5 (5.9), and 10% of women had a pregravid BMI ≥30 kg/m2 . Prepregnancy obesity (BMI of 30 kg/m2 ) was associated with increased offspring problem behaviours including internalising behaviours (adjusted β 3.3 points, 95% CI 1.7-4.9), externalising behaviours (adjusted β 2.9 points, 95% CI 1.4-4.6), and attention problems (adjusted β 2.3 points, 95% CI 1.1-3.4) on the CBCL, compared with normal weight mothers (BMI of 22 kg/m2 ). There were nonsignificant trends towards increased offspring impulsivity with low GWG among lean mothers (adjusted incidence rate ratio 1.2, 95% CI 0.9-1.5) and high GWG among overweight mothers (adjusted incidence rate ratio 1.7, 95% CI 0.9-2.8), but additional outcomes did not differ by GWG z-score. Results were not meaningfully different after excluding high-substance users. CONCLUSIONS In a low-income and high-risk sample, we observed a small increase in child behaviour problems among children of obese mothers, which could have an impact on child behaviour in the population. TWEETABLE ABSTRACT Maternal obesity is associated with a small increase in child behaviour problems.
Collapse
Affiliation(s)
- S J Pugh
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - J A Hutcheon
- Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, BC, Canada
| | - G A Richardson
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - M M Brooks
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - K P Himes
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Magee-Womens Research Institute, Pittsburgh, PA, USA
| | - N L Day
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - L M Bodnar
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
25
|
Younes-Rapozo V, Moura EG, Manhães AC, Pinheiro CR, Carvalho JC, Barradas PC, de Oliveira E, Lisboa PC. Neonatal Nicotine Exposure Leads to Hypothalamic Gliosis in Adult Overweight Rats. J Neuroendocrinol 2015; 27:887-98. [PMID: 26453898 DOI: 10.1111/jne.12328] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/23/2015] [Accepted: 10/06/2015] [Indexed: 01/22/2023]
Abstract
Astrocytes and microglia, the immune competent cells of central nercous system, can be activated in response to metabolic signals such as obesity and hyperleptinaemia. In rats, maternal exposure to nicotine during lactation leads to central obesity, hyperleptinaemia, leptin resistance and alterations in hypothalamic neuropeptides in the offspring during adulthood. In the present study, we studied the activation of astrocytes and microglia, as well as the pattern of inflammatory mediators, in adult offspring of this experimental model. On postnatal day 2 (P2), osmotic minipumps releasing nicotine (NIC) (-6 mg/kg/day) or saline for 14 days were s.c. implanted in dams. Male offspring were killed on P180 and hypothalamic immunohistochemistry, retroperitoneal white adipose tissue (WAT) polymerase chain reaction analysis and multiplex analysis for plasma inflammatory mediators were carried out. At P180, NIC astrocyte cell number was higher in the arcuate nucleus (ARC) (medial: +82%; lateral: +110%), in the paraventricular nucleus (PVN) (+144%) and in the lateral hypothalamus (+121%). NIC glial fibrillary acidic protein fibre density was higher in the lateral ARC (+178%) and in the PVN (+183%). Interleukin-6 was not affected in the hypothalamus. NIC monocyte chemotactic protein 1 was only higher in the periventricular nucleus (+287%). NIC microglia (iba-1-positive) cell number was higher (+68%) only in the PVN, as was the chemokine (C-X3-C motif) receptor 1 density (+93%). NIC interleukin-10 was lower in the WAT (-58%) and plasma (-50%). Thus, offspring of mothers exposed to nicotine during lactation present hypothalamic astrogliosis at adulthood and microgliosis in the PVN.
Collapse
Affiliation(s)
- V Younes-Rapozo
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - E G Moura
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - A C Manhães
- Laboratório de Neurofisiologia, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - C R Pinheiro
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - J C Carvalho
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - P C Barradas
- Laboratório de Neurobiologia, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - E de Oliveira
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - P C Lisboa
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
26
|
Mela V, Díaz F, Lopez-Rodriguez AB, Vázquez MJ, Gertler A, Argente J, Tena-Sempere M, Viveros MP, Chowen JA. Blockage of the Neonatal Leptin Surge Affects the Gene Expression of Growth Factors, Glial Proteins, and Neuropeptides Involved in the Control of Metabolism and Reproduction in Peripubertal Male and Female Rats. Endocrinology 2015; 156:2571-81. [PMID: 25856428 DOI: 10.1210/en.2014-1981] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Leptin (Lep) is important in the development of neuroendocrine circuits involved in metabolic control. Because both Lep and metabolism influence pubertal development, we hypothesized that early changes in Lep signaling could also modulate hypothalamic (HT) systems involved in reproduction. We previously demonstrated that a single injection of a Lep antagonist (Antag) on postnatal day (PND)9, coincident with the neonatal Lep peak, induced sexually dimorphic modifications in trophic factors and markers of cell turnover and neuronal maturation in the HT on PND13. Here, our aim was to investigate whether the alterations induced by Lep antagonism persist into puberty. Accordingly, male and female rats were treated with a pegylated super Lep Antag from PND5 to PND9 and killed just before the normal appearance of external signs of puberty (PND33 in females and PND43 in males). There was no effect on body weight, but in males food intake increased, subcutaneous adipose tissue decreased and HT neuropeptide Y and Agouti-related peptide mRNA levels were reduced, with no effect in females. In both sexes, the Antag increased HT mRNA levels of the kisspeptin receptor, G protein-coupled recepter 54 (Gpr54). Expression of the Lep receptor, trophic factors, and glial markers were differently affected in the HT of peripubertal males and females. Lep production in adipose tissue was decreased in Antag-treated rats of both sexes, with production of other cytokines being differentially regulated between sexes. In conclusion, in addition to the long-term effects on metabolism, changes in neonatal Lep levels modifies factors involved in reproduction that could possibly affect sexual maturation.
Collapse
Affiliation(s)
- Virginia Mela
- Department of Physiology (Animal Physiology II) (V.M., A.B.L.-R., M.-P.V.), Faculty of Biology, Universidad Complutense, 28040 Madrid, Spain; Department of Endocrinology (F.D., J.A., J.A.C.), Hospital Infantil Universitario Niño Jesús, Department of Pediatrics, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa, 28009 Madrid, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de Obesidad y Nutrición (F.D., M.J.V., J.A., M.T.-S., J.A.C.), Instituto Carlos III, 28903 Madrid, Spain; Department of Cell Biology, Physiology, and Immunology (M.J.V., M.T.-S.), University of Córdoba and Instituto Maimónides de Investigación Biomédica, Hospital Universitario Reina Sofia, 14004 Córdoba, Spain; and The Robert H. Smith Faculty of Agriculture, Food and Environment (A.G.), The Hebrew University of Jerusalem, Rehovot, Israel 76100
| | - Francisca Díaz
- Department of Physiology (Animal Physiology II) (V.M., A.B.L.-R., M.-P.V.), Faculty of Biology, Universidad Complutense, 28040 Madrid, Spain; Department of Endocrinology (F.D., J.A., J.A.C.), Hospital Infantil Universitario Niño Jesús, Department of Pediatrics, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa, 28009 Madrid, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de Obesidad y Nutrición (F.D., M.J.V., J.A., M.T.-S., J.A.C.), Instituto Carlos III, 28903 Madrid, Spain; Department of Cell Biology, Physiology, and Immunology (M.J.V., M.T.-S.), University of Córdoba and Instituto Maimónides de Investigación Biomédica, Hospital Universitario Reina Sofia, 14004 Córdoba, Spain; and The Robert H. Smith Faculty of Agriculture, Food and Environment (A.G.), The Hebrew University of Jerusalem, Rehovot, Israel 76100
| | - Ana Belen Lopez-Rodriguez
- Department of Physiology (Animal Physiology II) (V.M., A.B.L.-R., M.-P.V.), Faculty of Biology, Universidad Complutense, 28040 Madrid, Spain; Department of Endocrinology (F.D., J.A., J.A.C.), Hospital Infantil Universitario Niño Jesús, Department of Pediatrics, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa, 28009 Madrid, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de Obesidad y Nutrición (F.D., M.J.V., J.A., M.T.-S., J.A.C.), Instituto Carlos III, 28903 Madrid, Spain; Department of Cell Biology, Physiology, and Immunology (M.J.V., M.T.-S.), University of Córdoba and Instituto Maimónides de Investigación Biomédica, Hospital Universitario Reina Sofia, 14004 Córdoba, Spain; and The Robert H. Smith Faculty of Agriculture, Food and Environment (A.G.), The Hebrew University of Jerusalem, Rehovot, Israel 76100
| | - María Jesús Vázquez
- Department of Physiology (Animal Physiology II) (V.M., A.B.L.-R., M.-P.V.), Faculty of Biology, Universidad Complutense, 28040 Madrid, Spain; Department of Endocrinology (F.D., J.A., J.A.C.), Hospital Infantil Universitario Niño Jesús, Department of Pediatrics, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa, 28009 Madrid, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de Obesidad y Nutrición (F.D., M.J.V., J.A., M.T.-S., J.A.C.), Instituto Carlos III, 28903 Madrid, Spain; Department of Cell Biology, Physiology, and Immunology (M.J.V., M.T.-S.), University of Córdoba and Instituto Maimónides de Investigación Biomédica, Hospital Universitario Reina Sofia, 14004 Córdoba, Spain; and The Robert H. Smith Faculty of Agriculture, Food and Environment (A.G.), The Hebrew University of Jerusalem, Rehovot, Israel 76100
| | - Arieh Gertler
- Department of Physiology (Animal Physiology II) (V.M., A.B.L.-R., M.-P.V.), Faculty of Biology, Universidad Complutense, 28040 Madrid, Spain; Department of Endocrinology (F.D., J.A., J.A.C.), Hospital Infantil Universitario Niño Jesús, Department of Pediatrics, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa, 28009 Madrid, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de Obesidad y Nutrición (F.D., M.J.V., J.A., M.T.-S., J.A.C.), Instituto Carlos III, 28903 Madrid, Spain; Department of Cell Biology, Physiology, and Immunology (M.J.V., M.T.-S.), University of Córdoba and Instituto Maimónides de Investigación Biomédica, Hospital Universitario Reina Sofia, 14004 Córdoba, Spain; and The Robert H. Smith Faculty of Agriculture, Food and Environment (A.G.), The Hebrew University of Jerusalem, Rehovot, Israel 76100
| | - Jesús Argente
- Department of Physiology (Animal Physiology II) (V.M., A.B.L.-R., M.-P.V.), Faculty of Biology, Universidad Complutense, 28040 Madrid, Spain; Department of Endocrinology (F.D., J.A., J.A.C.), Hospital Infantil Universitario Niño Jesús, Department of Pediatrics, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa, 28009 Madrid, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de Obesidad y Nutrición (F.D., M.J.V., J.A., M.T.-S., J.A.C.), Instituto Carlos III, 28903 Madrid, Spain; Department of Cell Biology, Physiology, and Immunology (M.J.V., M.T.-S.), University of Córdoba and Instituto Maimónides de Investigación Biomédica, Hospital Universitario Reina Sofia, 14004 Córdoba, Spain; and The Robert H. Smith Faculty of Agriculture, Food and Environment (A.G.), The Hebrew University of Jerusalem, Rehovot, Israel 76100
| | - Manuel Tena-Sempere
- Department of Physiology (Animal Physiology II) (V.M., A.B.L.-R., M.-P.V.), Faculty of Biology, Universidad Complutense, 28040 Madrid, Spain; Department of Endocrinology (F.D., J.A., J.A.C.), Hospital Infantil Universitario Niño Jesús, Department of Pediatrics, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa, 28009 Madrid, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de Obesidad y Nutrición (F.D., M.J.V., J.A., M.T.-S., J.A.C.), Instituto Carlos III, 28903 Madrid, Spain; Department of Cell Biology, Physiology, and Immunology (M.J.V., M.T.-S.), University of Córdoba and Instituto Maimónides de Investigación Biomédica, Hospital Universitario Reina Sofia, 14004 Córdoba, Spain; and The Robert H. Smith Faculty of Agriculture, Food and Environment (A.G.), The Hebrew University of Jerusalem, Rehovot, Israel 76100
| | - María-Paz Viveros
- Department of Physiology (Animal Physiology II) (V.M., A.B.L.-R., M.-P.V.), Faculty of Biology, Universidad Complutense, 28040 Madrid, Spain; Department of Endocrinology (F.D., J.A., J.A.C.), Hospital Infantil Universitario Niño Jesús, Department of Pediatrics, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa, 28009 Madrid, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de Obesidad y Nutrición (F.D., M.J.V., J.A., M.T.-S., J.A.C.), Instituto Carlos III, 28903 Madrid, Spain; Department of Cell Biology, Physiology, and Immunology (M.J.V., M.T.-S.), University of Córdoba and Instituto Maimónides de Investigación Biomédica, Hospital Universitario Reina Sofia, 14004 Córdoba, Spain; and The Robert H. Smith Faculty of Agriculture, Food and Environment (A.G.), The Hebrew University of Jerusalem, Rehovot, Israel 76100
| | - Julie A Chowen
- Department of Physiology (Animal Physiology II) (V.M., A.B.L.-R., M.-P.V.), Faculty of Biology, Universidad Complutense, 28040 Madrid, Spain; Department of Endocrinology (F.D., J.A., J.A.C.), Hospital Infantil Universitario Niño Jesús, Department of Pediatrics, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa, 28009 Madrid, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de Obesidad y Nutrición (F.D., M.J.V., J.A., M.T.-S., J.A.C.), Instituto Carlos III, 28903 Madrid, Spain; Department of Cell Biology, Physiology, and Immunology (M.J.V., M.T.-S.), University of Córdoba and Instituto Maimónides de Investigación Biomédica, Hospital Universitario Reina Sofia, 14004 Córdoba, Spain; and The Robert H. Smith Faculty of Agriculture, Food and Environment (A.G.), The Hebrew University of Jerusalem, Rehovot, Israel 76100
| |
Collapse
|
27
|
Lemas DJ, Brinton JT, Shapiro ALB, Glueck DH, Friedman JE, Dabelea D. Associations of maternal weight status prior and during pregnancy with neonatal cardiometabolic markers at birth: the Healthy Start study. Int J Obes (Lond) 2015; 39:1437-42. [PMID: 26055075 PMCID: PMC4596750 DOI: 10.1038/ijo.2015.109] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 04/09/2015] [Accepted: 05/31/2015] [Indexed: 02/05/2023]
Abstract
Background Maternal obesity increases adult offspring risk for cardiovascular disease; however the role of offspring adiposity in mediating this association remains poorly characterized. Objective To investigate the associations of maternal pre-pregnant body mass index (maternal BMI) and gestational weight gain (GWG) with neonatal cardio-metabolic markers independent of fetal growth and neonatal adiposity. Methods A total of 753 maternal-infant pairs from the Healthy Start study, a large multi-ethnic pre-birth observational cohort were used. Neonatal cardio-metabolic markers included cord blood glucose, insulin, glucose-to-insulin ratio (Glu/Ins), total and high-density lipoprotein cholesterol (HDL-c), triglycerides, free fatty acids and leptin. Maternal BMI was abstracted from medical records or self-reported. GWG was calculated as the difference between the first pre-pregnant weight and the last weight measurement before delivery. Neonatal adiposity (percent fat mass) was measured within 72 hours of delivery using whole body air displacement plethysmography. Results In covariate adjusted models, maternal BMI was positively associated with cord blood insulin (p=0.01) and leptin (p<0.001) levels and inversely associated with cord blood HDL-c (p=0.05) and Glu/Ins (p=0.003). Adjustment for fetal growth or neonatal adiposity attenuated the effect of maternal BMI on neonatal insulin, rendering the association non-significant. However, maternal BMI remained associated with higher leptin (p<0.0011), lower HDL-c (p=0.02) and Glu/Ins (p=0.05), independent of neonatal adiposity. GWG was positively associated with neonatal insulin (p=0.02), glucose (p=0.03) and leptin levels (p<0.001) and negatively associated with Glu/Ins (p=0.006). After adjusting for neonatal adiposity, GWG remained associated with higher neonatal glucose (p=0.02) and leptin levels (p=0.02) and lower Glu/Ins (p=0.048). Conclusions Maternal weight prior and/or during pregnancy is associated with neonatal cardio-metabolic makers including leptin, glucose, and HDL-c at delivery, independent of neonatal adiposity. Our results suggest that intrauterine exposure to maternal obesity influences metabolic processes beyond fetal growth and fat accretion.
Collapse
Affiliation(s)
- D J Lemas
- Department of Pediatrics, Section of Neonatology, University of Colorado Denver, Aurora, CO, USA
| | - J T Brinton
- Department of Medicine, University of Colorado Denver, Denver, CO, USA
| | - A L B Shapiro
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Aurora, CO, USA
| | - D H Glueck
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - J E Friedman
- Department of Pediatrics, Section of Neonatology, University of Colorado Denver, Aurora, CO, USA
| | - D Dabelea
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
28
|
Alexeev EE, Lönnerdal B, Griffin IJ. Effects of postnatal growth restriction and subsequent catch-up growth on neurodevelopment and glucose homeostasis in rats. BMC PHYSIOLOGY 2015; 15:3. [PMID: 26040642 PMCID: PMC4455975 DOI: 10.1186/s12899-015-0017-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 05/26/2015] [Indexed: 11/10/2022]
Abstract
Background There is increasing evidence that poor growth of preterm infants is a risk factor for poor long-term development, while the effects of early postnatal growth restriction are not well known. We utilized a rat model to examine the consequences of different patterns of postnatal growth and hypothesized that early growth failure leads to impaired development and insulin resistance. Rat pups were separated at birth into normal (N, n = 10) or restricted intake (R, n = 16) litters. At d11, R pups were re-randomized into litters of 6 (R-6), 10 (R-10) or 16 (R-16) pups/dam. N pups remained in litters of 10 pups/dam (N-10). Memory and learning were examined through T-maze test. Insulin sensitivity was measured by i.p. insulin tolerance test and glucose tolerance test. Results By d10, N pups weighed 20 % more than R pups (p < 0.001). By d15, the R-6 group caught up to the N-10 group in weight, the R-10 group showed partial catch-up growth and the R-16 group showed no catch-up growth. All R groups showed poorer scores in developmental testing when compared with the N-10 group during T-Maze test (p < 0.05). Although R-16 were more insulin sensitive than R-6 and R-10, all R groups were more glucose tolerant than N-10. Conclusion In rats, differences in postnatal growth restriction leads to changes in development and in insulin sensitivity. These results may contribute to better elucidating the causes of poor developmental outcomes in human preterm infants.
Collapse
Affiliation(s)
- Erica E Alexeev
- Department of Nutrition, University of California, Davis, CA, 95616, USA.
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, CA, 95616, USA.
| | - Ian J Griffin
- Department of Pediatrics, University of California, Davis Medical Center, Sacramento, CA, 95817, USA.
| |
Collapse
|
29
|
Gali Ramamoorthy T, Begum G, Harno E, White A. Developmental programming of hypothalamic neuronal circuits: impact on energy balance control. Front Neurosci 2015; 9:126. [PMID: 25954145 PMCID: PMC4404811 DOI: 10.3389/fnins.2015.00126] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 03/26/2015] [Indexed: 01/08/2023] Open
Abstract
The prevalence of obesity in adults and children has increased globally at an alarming rate. Mounting evidence from both epidemiological studies and animal models indicates that adult obesity and associated metabolic disorders can be programmed by intrauterine and early postnatal environment- a phenomenon known as "fetal programming of adult disease." Data from nutritional intervention studies in animals including maternal under- and over-nutrition support the developmental origins of obesity and metabolic syndrome. The hypothalamic neuronal circuits located in the arcuate nucleus controlling appetite and energy expenditure are set early in life and are perturbed by maternal nutritional insults. In this review, we focus on the effects of maternal nutrition in programming permanent changes in these hypothalamic circuits, with experimental evidence from animal models of maternal under- and over-nutrition. We discuss the epigenetic modifications which regulate hypothalamic gene expression as potential molecular mechanisms linking maternal diet during pregnancy to the offspring's risk of obesity at a later age. Understanding these mechanisms in key metabolic genes may provide insights into the development of preventative intervention strategies.
Collapse
Affiliation(s)
| | - Ghazala Begum
- School of Clinical and Experimental Medicine, University of Birmingham Birmingham, UK
| | - Erika Harno
- Faculty of Life Sciences, University of Manchester Manchester, UK
| | - Anne White
- Faculty of Life Sciences, University of Manchester Manchester, UK ; Faculty of Medical and Human Sciences, Centre for Endocrinology and Diabetes, University of Manchester Manchester, UK
| |
Collapse
|
30
|
Antoniou EE, Fowler T, Reed K, Southwood TR, McCleery JP, Zeegers MP. Maternal pre-pregnancy weight and externalising behaviour problems in preschool children: a UK-based twin study. BMJ Open 2014; 4:e005974. [PMID: 25314961 PMCID: PMC4202011 DOI: 10.1136/bmjopen-2014-005974] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE To estimate the heritability of child behaviour problems and investigate the association between maternal pre-pregnancy overweight and child behaviour problems in a genetically sensitive design. DESIGN Observational cross-sectional study. SETTING The Twins and Multiple Births Association Heritability Study (TAMBAHS) is an online UK-wide volunteer-based study investigating the development of twins from birth until 5 years of age. PARTICIPANTS A total of 443 (16% of the initial registered members) mothers answered questions on pre-pregnancy weight and their twins' internalising and externalising problems using the Child Behavior Checklist and correcting for important covariates including gestational age, twins' birth weight, age and sex, mother's educational level and smoking (before, during and after pregnancy). PRIMARY OUTCOMES The heritability of behaviour problems and their association with maternal pre-pregnancy weight. RESULTS The genetic analysis suggested that genetic and common environmental factors account for most of the variation in externalising disorders (an ACE model was the most parsimonious with genetic factors (A) explaining 46% (95% CI 33% to 60%) of the variance, common environment (C) explaining 42% (95% CI 27% to 54%) and non-shared environmental factors (E) explaining 13% (95% CI 10% to 16%) of the variance. For internalising problems, a CE model was the most parsimonious model with the common environment explaining 51% (95% CI 44% to 58%) of the variance and non-shared environment explaining 49% (95% CI 42% to 56%) of the variance. Moreover, the regression analysis results suggested that children of overweight mothers showed a trend (OR=1.10, 95% CI 0.58% to 2.06) towards being more aggressive and exhibit externalising behaviours compared to children of normal weight mothers. CONCLUSIONS Maternal pre-pregnancy weight may play a role in children's aggressive behaviour.
Collapse
Affiliation(s)
- Evangelia E Antoniou
- Department of Clinical Psychological Science, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Tom Fowler
- Department of Public Health, Heart of Birmingham Teaching PCT, Birmingham, UK
| | - Keith Reed
- The Twins and Multiple Births Association (TAMBA), UK
| | | | - Joseph P McCleery
- School of Psychology, University of Birmingham, UK
- Center for Autism Research, Children's Hospital of Philadelphia, Philadelphia, PA USA
| | - Maurice P Zeegers
- Department of Complex Genetics, Cluster of Genetics and Cell Biology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
31
|
Meyer LR, Zhu V, Miller A, Roghair RD. Growth restriction, leptin, and the programming of adult behavior in mice. Behav Brain Res 2014; 275:131-5. [PMID: 25196633 DOI: 10.1016/j.bbr.2014.08.054] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 08/26/2014] [Accepted: 08/27/2014] [Indexed: 11/16/2022]
Abstract
Prematurity and neonatal growth restriction (GR) are risk factors for autism and attention deficit hyperactivity disorder (ADHD). Leptin production is suppressed during periods of undernutrition, and we have shown that isolated neonatal leptin deficiency leads to adult hyperactivity while neonatal leptin supplementation normalizes the brain morphology of GR mice. We hypothesized that neonatal leptin would prevent the development of GR-associated behavioral abnormalities. From postnatal day 4-14, C57BL/6 mice were randomized to daily injections of saline or leptin (80ng/g), and GR was identified by a weanling weight below the tenth percentile. The behavioral phenotypes of GR and control mice were assessed beginning at 4 months. Within the tripartite chamber, GR mice had significantly impaired social interaction. Baseline escape times from the Barnes maze were faster for GR mice (65+/-6s vs 87+/-7s for controls, p<0.05), but GR mice exhibited regression in their escape times on days 2 and 3 (56% regressed vs 22% of control saline mice, p<0.05). Compared to controls, GR mice entered the open arms of the elevated plus maze more often and stayed there longer (72+/-10s vs 36+/-5s, p<0.01). Neonatal leptin supplementation normalized the behavior of GR mice across all behavioral assays. In conclusion, GR alters the social interactions, learning and activity of mice, and supplementation with the neurotrophic hormone leptin mitigates these effects. We speculate neonatal leptin deficiency may contribute to the adverse neurodevelopmental outcomes associated with postnatal growth restriction, and postnatal leptin therapy may be protective.
Collapse
Affiliation(s)
- Lauritz R Meyer
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States of America.
| | - Vivian Zhu
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States of America
| | - Alise Miller
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States of America
| | - Robert D Roghair
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States of America
| |
Collapse
|
32
|
Guimond D, Diabira D, Porcher C, Bader F, Ferrand N, Zhu M, Appleyard SM, Wayman GA, Gaiarsa JL. Leptin potentiates GABAergic synaptic transmission in the developing rodent hippocampus. Front Cell Neurosci 2014; 8:235. [PMID: 25177272 PMCID: PMC4133691 DOI: 10.3389/fncel.2014.00235] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 07/26/2014] [Indexed: 12/17/2022] Open
Abstract
It is becoming increasingly clear that leptin is not only a hormone regulating energy homeostasis but also a neurotrophic factor impacting a number of brain regions, including the hippocampus. Although leptin promotes the development of GABAergic transmission in the hypothalamus, little is known about its action on the GABAergic system in the hippocampus. Here we show that leptin modulates GABAergic transmission onto developing CA3 pyramidal cells of newborn rats. Specifically, leptin induces a long-lasting potentiation (LLP-GABAA) of miniature GABAA receptor-mediated postsynaptic current (GABAA-PSC) frequency. Leptin also increases the amplitude of evoked GABAA-PSCs in a subset of neurons along with a decrease in the coefficient of variation and no change in the paired-pulse ratio, pointing to an increased recruitment of functional synapses. Adding pharmacological blockers to the recording pipette showed that the leptin-induced LLP-GABAA requires postsynaptic calcium released from internal stores, as well as postsynaptic MAPK/ERK kinases 1 and/or 2 (MEK1/2), phosphoinositide 3 kinase (PI3K) and calcium-calmodulin kinase kinase (CaMKK). Finally, study of CA3 pyramidal cells in leptin-deficient ob/ob mice revealed a reduction in the basal frequency of miniature GABAA-PSCs compared to wild type littermates. In addition, presynaptic GAD65 immunostaining was reduced in the CA3 stratum pyramidale of mutant animals, both results converging to suggest a decreased number of functional GABAergic synapses in ob/ob mice. Overall, these results show that leptin potentiates and promotes the development of GABAergic synaptic transmission in the developing hippocampus likely via an increase in the number of functional synapses, and provide insights into the intracellular pathways mediating this effect. This study further extends the scope of leptin's neurotrophic action to a key regulator of hippocampal development and function, namely GABAergic transmission.
Collapse
Affiliation(s)
- Damien Guimond
- Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France ; Unité 901, Institut National de la Santé et de la Recherche Médicale Marseille, France ; Institut de Neurobiologie de la Méditerranée Marseille, France ; Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University Pullman, WA, USA
| | - Diabe Diabira
- Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France ; Unité 901, Institut National de la Santé et de la Recherche Médicale Marseille, France ; Institut de Neurobiologie de la Méditerranée Marseille, France
| | - Christophe Porcher
- Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France ; Unité 901, Institut National de la Santé et de la Recherche Médicale Marseille, France ; Institut de Neurobiologie de la Méditerranée Marseille, France
| | - Francesca Bader
- Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France ; Unité 901, Institut National de la Santé et de la Recherche Médicale Marseille, France ; Institut de Neurobiologie de la Méditerranée Marseille, France
| | - Nadine Ferrand
- Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France ; Unité 901, Institut National de la Santé et de la Recherche Médicale Marseille, France ; Institut de Neurobiologie de la Méditerranée Marseille, France
| | - Mingyan Zhu
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University Pullman, WA, USA
| | - Suzanne M Appleyard
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University Pullman, WA, USA
| | - Gary A Wayman
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University Pullman, WA, USA
| | - Jean-Luc Gaiarsa
- Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France ; Unité 901, Institut National de la Santé et de la Recherche Médicale Marseille, France ; Institut de Neurobiologie de la Méditerranée Marseille, France
| |
Collapse
|
33
|
Lester BM, Conradt E, Marsit CJ. Are epigenetic changes in the intrauterine environment related to newborn neurobehavior? Epigenomics 2014; 6:175-8. [PMID: 24811786 PMCID: PMC4407197 DOI: 10.2217/epi.14.9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Barry M Lester
- Department of Pediatrics, Brown Center for the Study of Children at Risk, Warren Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA and Department of Psychiatry & Human Behavior, Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Elisabeth Conradt
- Department of Pediatrics, Brown Center for the Study of Children at Risk, Warren Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Carmen J Marsit
- Departments of Pharmacology & Toxicology and Community & Family Medicine, Section Biostatistics and Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| |
Collapse
|
34
|
Lesseur C, Armstrong DA, Murphy MA, Appleton AA, Koestler DC, Paquette AG, Lester BM, Marsit CJ. Sex-specific associations between placental leptin promoter DNA methylation and infant neurobehavior. Psychoneuroendocrinology 2014; 40:1-9. [PMID: 24485470 PMCID: PMC3912462 DOI: 10.1016/j.psyneuen.2013.10.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 09/27/2013] [Accepted: 10/21/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND Leptin (LEP) is a hormone central for energy homeostasis and has been implicated in neurodevelopment. This adipokine is produced by the placenta and is epigenetically regulated by promoter DNA methylation. Recent evidence has suggested a role for LEP in behavioral development. In this study, we investigated associations between profiles of human newborn neurobehavior and placental LEP DNA methylation. METHODS We determined LEP promoter methylation in 444 placental samples from healthy term infants and measured LEP gene expression in a random subset of these samples. Infant neurobehavior was assessed with the NICU Network Neurobehavioral Scales (NNNS) and we examined the relationship between LEP promoter methylation and profiles of infant neurobehavior derived from these scores generated using a hierarchical model-based clustering method. RESULTS LEP methylation is negatively correlated with gene expression only in placentas from male infants (r=-0.6, P=0.006). A 10% increase in LEP DNA methylation was associated with membership in a profile of infant neurobehavior marked by increased lethargy and hypotonicity (OR=1.9; 95% CI: 1.07-3.4), and consistently with reduced risk of membership in a profile characterized by decreased lethargy and hypotonicity (OR=0.54; 95% CI: 0.3-0.94) only in male infants (n=223). No statistically significant associations were observed amongst female infants. DISCUSSION These results suggest that increased placental LEP DNA methylation, related to reduced expression, may play a role in human newborn neurodevelopment, particularly in reactivity to various stimuli, but that these effects may be sexually dimorphic.
Collapse
Affiliation(s)
- Corina Lesseur
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, 7650 Remsen, Hanover, NH 03755, USA.
| | - David A. Armstrong
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, 7650 Remsen, Hanover, NH 03755, USA
| | - Megan A. Murphy
- Section of Biostatistics and Epidemiology, Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth and Norris Cotton Cancer Center, 1 Medical Center Drive, 7927 Rubin Building, Lebanon, NH 03756, USA
| | - Allison A. Appleton
- Section of Biostatistics and Epidemiology, Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth and Norris Cotton Cancer Center, 1 Medical Center Drive, 7927 Rubin Building, Lebanon, NH 03756, USA
| | - Devin C. Koestler
- Section of Biostatistics and Epidemiology, Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth and Norris Cotton Cancer Center, 1 Medical Center Drive, 7927 Rubin Building, Lebanon, NH 03756, USA
| | - Alison G. Paquette
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, 7650 Remsen, Hanover, NH 03755, USA
| | - Barry M. Lester
- The Brown Center for the Study of Children at Risk, Warren Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, RI 02903, USA
| | - Carmen J. Marsit
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, 7650 Remsen, Hanover, NH 03755, USA,Section of Biostatistics and Epidemiology, Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth and Norris Cotton Cancer Center, 1 Medical Center Drive, 7927 Rubin Building, Lebanon, NH 03756, USA,Corresponding author at: Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, 7650 Remsen, Hanover, NH 03755, USA. Tel.: +1 603 6501825; fax: +1 603 6501129. (C. Lesseur), (C.J. Marsit)
| |
Collapse
|
35
|
Makarova EN, Chepeleva EV, Panchenko PE, Bazhan NM. Influence of abnormally high leptin levels during pregnancy on metabolic phenotypes in progeny mice. Am J Physiol Regul Integr Comp Physiol 2013; 305:R1268-80. [PMID: 24089373 DOI: 10.1152/ajpregu.00162.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Maternal obesity increases the risk of obesity in offspring, and obesity is accompanied by an increase in blood leptin levels. The "yellow" mutation at the mouse agouti locus (A(y)) increases blood leptin levels in C57BL preobese pregnant mice without affecting other metabolic characteristics. We investigated the influence of the A(y) mutation or leptin injection at the end of pregnancy in C57BL mice on metabolic phenotypes and the susceptibility to diet-induced obesity (DIO) in offspring. In both C57BL-A(y) and leptin-treated mice, the maternal effect was more pronounced in male offspring. Compared with males born to control mothers, males born to A(y) mothers displayed equal food intake (FI) but decreased body weight (BW) gain after weaning, equal glucose tolerance, and enhanced FI-to-BW ratios on the standard diet but the same FI and BW on the high-fat diet. Males born to A(y) mothers were less responsive to the anorectic effect of exogenous leptin and less resistant to fasting (were not hyperphagic and gained less weight during refeeding after food deprivation) compared with males born to control mothers. However, all progeny displayed equal hypothalamic expression of Agouti gene-related protein (AgRP), neuropeptide Y (NPY), and proopiomelanocortin (POMC) and equal plasma leptin and glucose levels after food deprivation. Leptin injections in C57BL mice on day 17 of pregnancy decreased BW in both male and female offspring but inhibited FI and DIO only in male offspring. Our results show that hyperleptinemia during pregnancy has sex-specific long-term effects on energy balance regulation in progeny and does not predispose offspring to developing obesity.
Collapse
Affiliation(s)
- Elena N Makarova
- Laboratory of Physiological Genetics, Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | | | | | | |
Collapse
|
36
|
Stachowiak EK, Oommen S, Vasu VT, Srinivasan M, Stachowiak M, Gohil K, Patel MS. Maternal obesity affects gene expression and cellular development in fetal brains. Nutr Neurosci 2013; 16:96-103. [DOI: 10.1179/1476830512y.0000000035] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
37
|
Beck P, Urbano FJ, Williams DK, Garcia-Rill E. Effects of leptin on pedunculopontine nucleus (PPN) neurons. J Neural Transm (Vienna) 2013; 120:1027-38. [PMID: 23263542 PMCID: PMC3618992 DOI: 10.1007/s00702-012-0957-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 12/10/2012] [Indexed: 12/24/2022]
Abstract
Leptin, a hormone that regulates appetite and energy expenditure, is increased in obese individuals, although these individuals often exhibit leptin resistance. Obesity is characterized by sleep/wake disturbances, such as excessive daytime sleepiness, increased REM sleep, increased nighttime arousals, and decreased percentage of total sleep time. Several studies have shown that short sleep duration is highly correlated with decreased leptin levels in both animal and human models. Arousal and rapid eye movement (REM) sleep are regulated by the cholinergic arm of the reticular activating system, the pedunculopontine nucleus (PPN). The goal of this project was to determine the role of leptin in the PPN, and thus in obesity-related sleep disorders. Whole-cell patch-clamp recordings were conducted on PPN neurons in 9- to 17-day-old rat brainstem slices. Leptin decreased action potential (AP) amplitude, AP frequency, and h-current (I(H)). These findings suggest that leptin causes a blockade of Na⁺ channels. Therefore, we conducted an experiment to test the effects of leptin on Na⁺ conductance. To determine the average voltage dependence of this conductance, results from each cell were equally weighted by expressing conductance as a fraction of the maximum conductance in each cell. I Na amplitude was decreased in a dose-dependent manner, suggesting a direct effect of leptin on these channels. The average decrease in Na⁺ conductance by leptin was ~40 %. We hypothesize that leptin normally decreases activity in the PPN by reducing I(H) and I(Na) currents, and that in states of leptin dysregulation (i.e., leptin resistance) this effect may be blunted, therefore causing increased arousal and REM sleep drive, and ultimately leading to sleep-related disorders.
Collapse
Affiliation(s)
- Paige Beck
- Center for Translational Neuroscience, Dept. Neurobiology & Dev. Sci., University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - D. Keith Williams
- Center for Translational Neuroscience, Dept. Neurobiology & Dev. Sci., University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Edgar Garcia-Rill
- Center for Translational Neuroscience, Dept. Neurobiology & Dev. Sci., University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
38
|
Sferruzzi-Perri AN, Vaughan OR, Forhead AJ, Fowden AL. Hormonal and nutritional drivers of intrauterine growth. Curr Opin Clin Nutr Metab Care 2013; 16:298-309. [PMID: 23340010 DOI: 10.1097/mco.0b013e32835e3643] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
PURPOSE OF REVIEW Size at birth is critical in determining life expectancy with both small and large neonates at risk of shortened life spans. This review examines the hormonal and nutritional drivers of intrauterine growth with emphasis on the role of foetal hormones as nutritional signals in utero. RECENT FINDINGS Nutrients drive intrauterine growth by providing substrate for tissue accretion, whereas hormones regulate nutrient distribution between foetal oxidative metabolism and mass accumulation. The main hormonal drivers of intrauterine growth are insulin, insulin-like growth factors and thyroid hormones. Together with leptin and cortisol, these hormones control cellular nutrient uptake and the balance between accretion and differentiation in regulating tissue growth. They also act indirectly via the placenta to alter the materno-foetal supply of nutrients and oxygen. By responding to nutrient and oxygen availability, foetal hormones optimize the survival and growth of the foetus with respect to its genetic potential, particularly during adverse conditions. However, changes in the intrauterine growth of individual tissues may alter their function permanently. SUMMARY In both normal and compromised pregnancies, intrauterine growth is determined by multiple hormonal and nutritional drivers which interact to produce a specific pattern of intrauterine development with potential lifelong consequences for health.
Collapse
Affiliation(s)
- Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| | | | | | | |
Collapse
|
39
|
Steculorum SM, Vogt MC, Brüning JC. Perinatal programming of metabolic diseases: role of insulin in the development of hypothalamic neurocircuits. Endocrinol Metab Clin North Am 2013; 42:149-64. [PMID: 23391245 DOI: 10.1016/j.ecl.2012.10.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It is increasingly accepted that the metabolic future of an individual can be programmed as early as at developmental stages. For instance, offspring of diabetic mothers have a greater risk of becoming obese and diabetic later in life. Animal studies have demonstrated that hyperinsulinemia and/or hyperglycemia during perinatal life permanently impair the organization and long-term function of hypothalamic networks that control appetite and glucose homeostasis. This review summarizes the main findings regarding the key regulatory roles of perinatal insulin and glucose levels on hypothalamic development and on long-term programming of metabolic diseases reported in different rodent models.
Collapse
Affiliation(s)
- Sophie M Steculorum
- Department of Mouse Genetics and Metabolism, University Hospital Cologne and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Köln 50674, Germany
| | | | | |
Collapse
|
40
|
Abstract
Research into the control of energy balance has tended to focus on discrete brain regions, such as the brainstem, medulla, arcuate nucleus of the hypothalamus, and neocortex. Recently, a larger picture has begun to emerge in which the coordinated communication between these areas is proving to be critical to appropriate regulation of metabolism. By serving as a center for such communication, the paraventricular nucleus of the hypothalamus (PVH) is perhaps the most important brain nucleus regulating the physiological response to energetic challenges. Here we review recent advances in the understanding of the circuitry and function of the PVH.
Collapse
Affiliation(s)
- Jennifer W. Hill
- Department of Physiology and Pharmacology, Center for Diabetes and Endocrine Research, University of Toledo Medical Center, Obstetrics-Gynecology, University of Toledo, USA
| |
Collapse
|
41
|
Picó C, Palou M, Priego T, Sánchez J, Palou A. Metabolic programming of obesity by energy restriction during the perinatal period: different outcomes depending on gender and period, type and severity of restriction. Front Physiol 2012. [PMID: 23189059 PMCID: PMC3504314 DOI: 10.3389/fphys.2012.00436] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Epidemiological studies in humans and controlled intervention studies in animals have shown that nutritional programming in early periods of life is a phenomenon that affects metabolic and physiological functions throughout life. The phenotypes of health or disease are hence the result of the interaction between genetic and environmental factors, starting right from conception. In this sense, gestation and lactation are disclosed as critical periods. Continuous food restriction during these stages may lead to permanent adaptations with lasting effects on the metabolism of the offspring and may influence the propensity to develop different chronic diseases associated with obesity. However, the different outcomes of these adaptations on later health may depend on factors such as the type, duration, period, and severity of the exposure to energy restriction conditions, and they are, in part, gender specific. A better understanding of the factors and mechanisms involved in metabolic programming, and their effects, may contribute significantly to the prevention of obesity, which is considered to be one of the major health concerns of our time. Here, the different outcomes of maternal food restriction during gestation and lactation in the metabolic health of offspring, as well as potential mechanisms underlying these effects are reviewed.
Collapse
Affiliation(s)
- Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics), University of the Balearic Islands, and CIBER de Fisiopatología de la Obesidad y Nutrición Spain
| | | | | | | | | |
Collapse
|
42
|
Abstract
Considerable epidemiological, experimental and clinical data have amassed showing that the risk of developing disease in later life is dependent upon early life conditions. In particular, altered maternal nutrition, including undernutrition and overnutrition, can lead to metabolic disorders in offspring characterised by obesity and leptin resistance. The adipokine leptin has received significant interest as a potential programming factor; alterations in the profile of leptin in early life are associated with altered susceptibility to obesity and metabolic disorders in adulthood. Maintenance of a critical leptin level during early development facilitates the normal maturation of tissues and signalling pathways involved in metabolic homeostasis. A period of relative hypo- or hyperleptinemia during this window of development will induce some of the metabolic adaptations which underlie developmental programming. However, it remains unclear whether leptin alone is a critical factor for the programming of obesity. At least in animal experimental studies, developmental programming is potentially reversible by manipulating the concentration of circulating leptin during a critical window of developmental plasticity and offers an exciting new approach for therapeutic intervention.
Collapse
Affiliation(s)
- M H Vickers
- Liggins Institute and The National Research Centre for Growth and Development, University of Auckland, Grafton, Auckland, New
| | | |
Collapse
|
43
|
D'Ippolito S, Tersigni C, Scambia G, Di Simone N. Adipokines, an adipose tissue and placental product with biological functions during pregnancy. Biofactors 2012; 38:14-23. [PMID: 22287297 DOI: 10.1002/biof.201] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 12/28/2011] [Indexed: 01/03/2023]
Abstract
Latter half of pregnancy is characterized by a "physiological diabetogenic state" since changes in insulin-sensitivity have been well documented. These changes ensure continuous supply of nutrients to the growing fetus. In the last years the role of adipocyte-derived signaling molecules, collectively known as adipokines has been object of different in vitro and in vivo studies. Of interest, adipokines and/or their receptors are expressed in the placental tissue which, therefore, can contribute to development of maternal insulin-resistance and, as a consequence, fetal growth. Leptin, adiponectin, and resistin represent the most well studied adipokines and, with the exception of adiponectin, their serum and placental levels increase as pregnancy progresses. High levels of adipokines have also been detected in umbilical plasma hence suggesting a possible role on fetal development and metabolism; however, it remains still unclear if such adipokines can directly stimulate fetal tissues development acting as growth factors. In addition to their well known metabolic effects, we also reported studies describing the role of adipokines in promoting proliferation and invasiveness of trophoblast cells and affecting local angiogenic processes. These observations strongly suggest that adipokines, by alternatively interfering with placental development, may affect pregnancy outcome and fetal growth. However, further studies are needed to better understand the local regulation of their expression. © 2012 International Union of Biochemistry and Molecular Biology, Inc.
Collapse
Affiliation(s)
- Silvia D'Ippolito
- Department of Obstetrics and Gynecology, Policlinico A. Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | | |
Collapse
|
44
|
Roitbak T, Bragina O, Padilla JL, Pickett GG. The role of microRNAs in neural stem cell-supported endothelial morphogenesis. Vasc Cell 2011; 3:25. [PMID: 22071092 PMCID: PMC3226576 DOI: 10.1186/2045-824x-3-25] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 11/09/2011] [Indexed: 12/28/2022] Open
Abstract
Functional signaling between neural stem/progenitor cells (NSPCs) and brain endothelial cells (ECs) is essential to the coordination of organized responses during initial embryonic development and also during tissue repair, which occurs following brain injury. In this study, we investigated the molecular mechanisms underlying this functional signaling, using primary mouse brain ECs and NSPCs from embryonic mouse brain. EC/NSPC co-culture experiments have revealed that neural progenitors secrete factors supporting angiogenesis, which induce noticeable changes in endothelial morphology. We demonstrate that NSPCs influence the expression of mTOR and TGF-β signaling pathway components implicated in the regulation of angiogenesis. Endothelial morphogenesis, an essential component of vascular development, is a complex process involving gene activation and the upregulation of specific cell signaling pathways. Recently identified small molecules, called microRNAs (miRNAs), regulate the expression of genes and proteins in many tissues, including brain and vasculature. We found that NSPCs induced considerable changes in the expression of at least 24 miRNAs and 13 genes in ECs. Three NSPC-regulated EC miRNAs were identified as the potential primary mediators of this NSPC/EC interaction. We found that the specific inhibition, or overexpression, of miRNAs miR-155, miR-100, and miR-let-7i subsequently altered the expression of major components of the mTOR, TGF-β and IGF-1R signaling pathways in ECs. Overexpression of these miRNAs in ECs suppressed, while inhibition activated, the in vitro formation of capillary-like structures, a process representative of EC morphogenesis. In addition, we demonstrate that inhibition of FGF, VEGF, and TGF-β receptor signaling abolished NSPC-promoted changes in the endothelial miRNA profiles. Our findings demonstrate that NSPCs induce changes in the miRNA expression of ECs, which are capable of activating angiogenesis by modulating distinct cell signaling pathways.
Collapse
Affiliation(s)
- Tamara Roitbak
- Department of Neurosugery, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
| | | | | | | |
Collapse
|
45
|
Froehlich TE, Anixt JS, Loe IM, Chirdkiatgumchai V, Kuan L, Gilman RC. Update on environmental risk factors for attention-deficit/hyperactivity disorder. Curr Psychiatry Rep 2011; 13:333-44. [PMID: 21779823 PMCID: PMC3277258 DOI: 10.1007/s11920-011-0221-3] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a prevalent neurobehavioral disorder affecting 5% to 10% of children. Although considered to be a highly familial disorder, ADHD heritability estimates of 60% to 80% highlight the considerable role that environmental factors may still play in disorder susceptibility. Proposed ADHD environmental risk factors include prenatal substance exposures, heavy metal and chemical exposures, nutritional factors, and lifestyle/psychosocial factors. This paper reviews the literature published in 2010 investigating the association between environmental risk factors and ADHD or related symptomatology. Sources of risk factor exposure and the proposed mechanism by which each exposure is linked to ADHD-related neurobehavioral changes are also reported. Methodologic limitations of the current literature are discussed, and guidelines for future study are proposed. An improved understanding of the role that environmental factors play in ADHD etiology is critical to future ADHD prevention efforts.
Collapse
Affiliation(s)
- Tanya E. Froehlich
- Cincinnati Children’s Hospital, 3333 Burnet Avenue, MLC 4002, Cincinnati, OH 45229, USA
| | - Julia S. Anixt
- Cincinnati Children’s Hospital, 3333 Burnet Avenue, MLC 4002, Cincinnati, OH 45229, USA
| | - Irene M. Loe
- Stanford University School of Medicine, 750 Welch Road, Suite 315, Palo Alto, CA 94304, USA
| | | | - Lisa Kuan
- Cincinnati Children’s Hospital, 3333 Burnet Avenue, MLC 4002, Cincinnati, OH 45229, USA
| | - Richard C. Gilman
- Cincinnati Children’s Hospital, 3333 Burnet Avenue, MLC 4002, Cincinnati, OH 45229, USA
| |
Collapse
|
46
|
Denver RJ, Bonett RM, Boorse GC. Evolution of leptin structure and function. Neuroendocrinology 2011; 94:21-38. [PMID: 21677426 DOI: 10.1159/000328435] [Citation(s) in RCA: 146] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 04/11/2011] [Indexed: 12/15/2022]
Abstract
Leptin, the protein product of the obese(ob or Lep) gene, is a hormone synthesized by adipocytes that signals available energy reserves to the brain, and thereby influences development, growth, metabolism and reproduction. In mammals, leptin functions as an adiposity signal: circulating leptin fluctuates in proportion to fat mass, and it acts on the hypothalamus to suppress food intake. Orthologs of mammalian Lep genes were recently isolated from several fish and two amphibian species, and here we report the identification of two Lep genes in a reptile, the lizard Anolis carolinensis. While vertebrate leptins show large divergence in their primary amino acid sequence, they form similar tertiary structures, and may have similar potencies when tested in vitro on heterologous leptin receptors (LepRs). Leptin binds to LepRs on the plasma membrane, activating several intracellular signaling pathways. Vertebrate LepRs signal via the Janus kinase (Jak) and signal transducer and activator of transcription (STAT) pathway. Three tyrosine residues located within the LepR cytoplasmic domain are phosphorylated by Jak2 and are required for activation of SH2-containing tyrosine phosphatase-2, STAT5 and STAT3 signaling. These tyrosines are conserved from fishes to mammals, demonstrating their critical role in signaling by the LepR. Leptin is anorexigenic in representatives of all vertebrate classes, suggesting that its role in energy balance is ancient and has been evolutionarily conserved. In addition to its integral role as a regulator of appetite and energy balance, leptin exerts pleiotropic actions in development, physiology and behavior.
Collapse
Affiliation(s)
- Robert J Denver
- Department of Molecular, Cellular and Developmental Biology, The University of Michigan, Ann Arbor, USA. rdenver @ umich.edu
| | | | | |
Collapse
|
47
|
Hashimoto R, Udagawa J, Kagohashi Y, Matsumoto A, Hatta T, Otani H. Direct and indirect effects of neuropeptide Y and neurotrophin 3 on myelination in the neonatal brains. Brain Res 2010; 1373:55-66. [PMID: 21167823 DOI: 10.1016/j.brainres.2010.12.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2010] [Revised: 12/06/2010] [Accepted: 12/09/2010] [Indexed: 11/29/2022]
Abstract
Neuropeptide Y (NPY) is expressed in the developing central nervous system, however, its role in the brain development remains unclear. In this study, C57/B6 mice were intraperitoneally administered 1 nmol/capita/day of NPY, 10 nmol/capita/day of an NPY-receptor 1-specific antagonist (Y1R-A), or NPY and Y1R-A simultaneously (NPY+Y1R-A) from postnatal day (P) 7 to P14. Recombinant NPY reached the P14 cerebrum in 1 hour. These treatments didn't significantly affect body weight gain or P14 brain weight. The ratio of myelinated axons to total axons in the parietal cerebrum was significantly higher in the NPY group than in the control group. The expression of myelin basic protein (MBP)-mRNA in the cerebrum was significantly higher in the NPY group than in the control group and was significantly lower in the NPY+Y1R-A group than in the NPY group, while it was significantly higher in the NPY+Y1R-A group than in the control group. In cultured oligodendroglioma-derived B12 cells, NPY didn't influence the MBP-mRNA expression, while neurotrophin 3 (NT3) increased MBP mRNA via receptor-type tyrosine kinase type C (Trk C). NPY administration significantly increased NT3-mRNA expression in the P14 cerebrum as deduced by quantitative real-time PCR. The change in phosphorylated Trk C (P-Trk C) was proportional to that of the NT3-mRNA expression, and the proportion of P-Trk C was higher in the NPY group than in the control group. These results suggest that NPY, partially via Y1R, induces NT3 which, via Trk C phosphorylation, accelerates myelination by oligodendrocytes in the mouse brain during the neonatal period.
Collapse
Affiliation(s)
- Ryuju Hashimoto
- Department of Developmental Biology, Faculty of Medicine, Shimane University, Izumo, Japan.
| | | | | | | | | | | |
Collapse
|
48
|
Otani H, Udagawa J, Hatta T, Kagohashi Y, Hashimoto R, Matsumoto A, Satow F, Nimura M. Individual variation in organ histogenesis as a causative factor in the developmental origins of health and disease: unnoticed congenital anomalies? Congenit Anom (Kyoto) 2010; 50:205-11. [PMID: 20831656 DOI: 10.1111/j.1741-4520.2010.00295.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Morphological studies of congenital anomalies have mainly focused on abnormal shape (i.e. malformation) and thus on disturbed organogenesis. However, in regard to postnatal functions of organs that develop through branching mechanisms, organ size is another important morphological feature. These organs consist of a large number of structural and functional units, such as nephrons in the kidney, and the total number of these units, that is approximately proportional to the organ size, has been shown to vary widely among individuals. Organ-specific cells are differentiated and organized to form structural units and realize organ-specific functions during the histogenetic period (i.e. from mid-gestation to the early postnatal period). The total number of units is attained at the end of histogenesis and determines the total functional capacity, including the functional reserve of the organ, and thus may be related to predispositions to postnatal organ-based diseases, because the functional reserve decreases during the course of life and eventually become short of the minimum requirement of each organ. Therefore, it may be hypothesized that a smaller number of units of organs at the end of histogenesis is one of the predisposing factors for postnatal diseases (i.e. a form of unnoticed but late-manifested congenital anomalies), in this era of extended longevity. However, the mechanisms that control the total number of units in each organ during histogenesis and the possible relationship among the numbers of units in different organs remain unknown. Here, we review our trials based on the above hypothesis in order to (1) mathematically analyze the morphometric data of the different organs in fetuses to elucidate relationship among developing organs, (2) analyze the developing neuro-immuno-endocrine network as a series of mechanisms to systemically correlate the histogenesis of multiple organs, and (3) examine the maternal environment, including dietary fat, as a factor to influence histogenesis and thus the predisposition to type 1 diabetes.
Collapse
Affiliation(s)
- Hiroki Otani
- Department of Developmental Biology, Faculty of Medicine, Shimane University, Izumo, Shimane, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Coupé B, Amarger V, Grit I, Benani A, Parnet P. Nutritional programming affects hypothalamic organization and early response to leptin. Endocrinology 2010; 151:702-13. [PMID: 20016030 DOI: 10.1210/en.2009-0893] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Nutritional programming, taking place in utero or early after birth, is closely linked with metabolic and appetite disorders in adulthood. Following the hypothesis that nutritional programming impacts hypothalamic neuronal organization, we report on discrepancies of multiple molecular and cellular early events that take place in the hypothalamus of rats submitted to intrauterine growth restriction (IUGR). Expression screening performed on hypothalami from IUGR rats at birth and at postnatal d 12 identified changes in gene expression of neurodevelopmental process (cell differentiation and cytoskeleton organization). Additionally, a slight reduction of agouti-related protein and a strong reduction of alpha-MSH-immunoreactive efferent fibers were demonstrated in the paraventricular nucleus of IUGR rats. Rapid catch-up growth of IUGR rats, 5 d after birth, had a positive effect on neurodevelopmental factors and on neuronal projections emanating from the arcuate nucleus. The molecular and cellular anomalies detected in IUGR rats can be related to the reduced and delayed plasma leptin surge from d 0-16 when compared with control and IUGR rats with catch-up growth. However, the ability of leptin to activate intracellular signaling in arcuate nucleus neurons was not reduced in IUGR rats. Other mechanism such as epigenetic regulation of the major appetite-regulating neuropeptides genes was analyzed in parallel with their mRNA expression during postnatal development. This study reveals the importance of an early catch-up growth that reduces abnormal organization of hypothalamic pathways involved in energy homeostasis, whereas protein restriction, maintained during postnatal development leads to an important immaturity of the hypothalamus.
Collapse
Affiliation(s)
- Bérengère Coupé
- Institut National de la Recherche Agronomique, Unité Mixte de Recherche 1280 Physiologie des Adaptations Nutritionnelles, Université de Nantes, Nantes, France
| | | | | | | | | |
Collapse
|
50
|
Rodriguez A. Maternal pre-pregnancy obesity and risk for inattention and negative emotionality in children. J Child Psychol Psychiatry 2010; 51:134-43. [PMID: 19674195 DOI: 10.1111/j.1469-7610.2009.02133.x] [Citation(s) in RCA: 184] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE This study aimed to replicate and extend previous work showing an association between maternal pre-pregnancy adiposity and risk for attention deficit hyperactivity disorder (ADHD) symptoms in children. METHODS A Swedish population-based prospective pregnancy-offspring cohort was followed up when children were 5 years old (N = 1,714). Mothers and kindergarten teachers rated children's ADHD symptoms, presence and duration of problems, and emotionality. Dichotomized outcomes examined difficulties of clinical relevance (top 15% of the distribution). Analyses adjusted for pregnancy (maternal smoking, depressive symptoms, life events, education, age, family structure), birth outcomes (birth weight, gestational age, infant sex) and concurrent variables (family structure, maternal depressive symptoms, parental ADHD symptoms, and child overweight) in an attempt to rule out confounding. RESULTS Maternal pre-pregnancy overweight and obesity predicted high inattention symptom scores and obesity was associated with a two-fold increase in risk of difficulties with emotion intensity and emotion regulation according to teacher reports. Means of maternal ratings were unrelated to pre-pregnancy body mass index (BMI). Presence and duration of problems were associated with both maternal over and underweight according to teachers. CONCLUSIONS Despite discrepancies between maternal and teacher reports, these results provide further evidence that maternal pre-pregnancy overweight and obesity are associated with child inattention symptoms and extend previous work by establishing a link between obesity and emotional difficulties. Maternal adiposity at the time of conception may be instrumental in programming child mental health, as prenatal brain development depends on maternal energy supply. Possible mechanisms include disturbed maternal metabolic function. If maternal pre-pregnancy obesity is a causal risk factor, the potential for prevention is great.
Collapse
Affiliation(s)
- Alina Rodriguez
- Department of Psychology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|