1
|
Ahmad S, Drag MH, Salleh SM, Cai Z, Nielsen MO. Transcriptomics analysis of differentially expressed genes in subcutaneous and perirenal adipose tissue of sheep as affected by their pre- and early postnatal malnutrition histories. BMC Genomics 2021; 22:338. [PMID: 33975549 PMCID: PMC8114714 DOI: 10.1186/s12864-021-07672-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/30/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Early life malnutrition is known to target adipose tissue with varying impact depending on timing of the insult. This study aimed to identify differentially expressed genes in subcutaneous (SUB) and perirenal (PER) adipose tissue of 2.5-years old sheep to elucidate the biology underlying differential impacts of late gestation versus early postnatal malnutrition on functional development of adipose tissues. Adipose tissues were obtained from 37 adult sheep born as twins to dams fed either NORM (fulfilling energy and protein requirements), LOW (50% of NORM) or HIGH (110% of protein and 150% of energy requirements) diets in the last 6-weeks of gestation. From day 3 to 6 months of age, lambs were fed high-carbohydrate-high-fat (HCHF) or moderate low-fat (CONV) diets, and thereafter the same moderate low-fat diet. RESULTS The gene expression profile of SUB in the adult sheep was not affected by the pre- or early postnatal nutrition history. In PER, 993 and 186 differentially expressed genes (DEGs) were identified in LOW versus HIGH and NORM, respectively, but no DEG was found between HIGH and NORM. DEGs identified in the mismatched pre- and postnatal nutrition groups LOW-HCHF (101) and HIGH-HCHF (192) were largely downregulated compared to NORM-CONV. Out of 831 DEGs, 595 and 236 were up- and downregulated in HCHF versus CONV, respectively. The functional enrichment analyses revealed that transmembrane (ion) transport activities, motor activities related to cytoskeletal and spermatozoa function (microtubules and the cytoskeletal motor protein, dynein), and responsiveness to the (micro) environmental extracellular conditions, including endocrine and nervous stimuli were enriched in the DEGs of LOW versus HIGH and NORM. We confirmed that mismatched pre- and postnatal feeding was associated with long-term programming of adipose tissue remodeling and immunity-related pathways. In agreement with phenotypic measurements, early postnatal HCHF feeding targeted pathways involved in kidney cell differentiation, and mismatched LOW-HCHF sheep had specific impairments in cholesterol metabolism pathways. CONCLUSIONS Both pre- and postnatal malnutrition differentially programmed (patho-) physiological pathways with implications for adipose functional development associated with metabolic dysfunctions, and PER was a major target.
Collapse
Affiliation(s)
- Sharmila Ahmad
- Nutrition Research Unit, Department of Animal Science, Aarhus University, Blichers Alle 20, 8830 Tjele, Denmark
| | - Markus Hodal Drag
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Suraya Mohamad Salleh
- Department of Animal Science, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
- Department of Animal Nutrition and Management, Swedish University of Agricultural Sciences, 750 07 Uppsala, Sweden
| | - Zexi Cai
- Centre for Quantitative Genetics and Genomics, Aarhus University, Blichers Alle 20, 8830 Tjele, Denmark
| | - Mette Olaf Nielsen
- Nutrition Research Unit, Department of Animal Science, Aarhus University, Blichers Alle 20, 8830 Tjele, Denmark
| |
Collapse
|
2
|
Sato S, Mukai Y. Modulation of Chronic Inflammation by Quercetin: The Beneficial Effects on Obesity. J Inflamm Res 2020; 13:421-431. [PMID: 32848440 PMCID: PMC7425105 DOI: 10.2147/jir.s228361] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/22/2020] [Indexed: 12/18/2022] Open
Abstract
Obesity has become a major risk factor for the development of chronic diseases such as insulin resistance, type 2 diabetes mellitus, and cardiovascular disease. Moreover, obesity induces chronic inflammation in adipose tissue, liver, skeletal muscle, and the vascular system. Quercetin is the major representative of the flavonoid subclass of flavonols, which is ubiquitously contained within natural plants such as green tea, and vegetables, including onions and apples. Researchers have focused greater attention to the beneficial physiological roles of quercetin, which has anti-oxidative, anti-inflammatory, and anti-fibrotic effects on insulin resistance and atherosclerosis in obesity-related diseases. Also, the anti-inflammatory effects of quercetin on intestinal microbiota have been demonstrated in obesity. In addition, there is increasing evidence that quercetin is associated with epigenetic activities in cancer, and in maternal undernutrition during gestation and lactation. In this review, we focus on the chemical properties of quercetin, its dietary sources in obesity, and its anti-inflammatory effects on insulin resistance, atherosclerosis, intestinal microbiota, and maternal under-nutrition with epigenetic activity.
Collapse
Affiliation(s)
- Shin Sato
- Department of Nutrition, Aomori University of Health and Welfare, Aomori 030-8505, Japan
| | - Yuuka Mukai
- School of Nutrition and Dietetics, Faculty of Health and Social Work, Kanagawa University of Human Services, Kanagawa 238-8522, Japan
| |
Collapse
|
3
|
Sato S, Norikura T, Mukai Y. Maternal quercetin intake during lactation attenuates renal inflammation and modulates autophagy flux in high-fructose-diet-fed female rat offspring exposed to maternal malnutrition. Food Funct 2019; 10:5018-5031. [DOI: 10.1039/c9fo01134j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Quercetin intake during lactation causes long-term alterations in inflammation and autophagy flux in the kidneys of high-fructose-diet-fed adult female offspring exposed to maternal normal- or low-protein diets.
Collapse
Affiliation(s)
- Shin Sato
- Department of Nutrition
- Aomori University of Health and Welfare
- Aomori 030-8505
- Japan
| | - Toshio Norikura
- Department of Nutrition
- Aomori University of Health and Welfare
- Aomori 030-8505
- Japan
| | - Yuuka Mukai
- School of Nutrition and Dietetics
- Faculty of Health and Social Work
- Kanagawa University of Human Services
- Kanagawa 238-8522
- Japan
| |
Collapse
|
4
|
Kataoka S, Norikura T, Sato S. Maternal green tea polyphenol intake during lactation attenuates kidney injury in high-fat-diet-fed male offspring programmed by maternal protein restriction in rats. J Nutr Biochem 2018. [DOI: 10.1016/j.jnutbio.2018.01.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
5
|
Saroha V, Dellschaft NS, Keisler DH, Gardner DS, Budge H, Sebert SP, Symonds ME. Tissue cell stress response to obesity and its interaction with late gestation diet. Reprod Fertil Dev 2017; 30:430-441. [PMID: 28768569 DOI: 10.1071/rd16494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 07/08/2017] [Indexed: 12/31/2022] Open
Abstract
Intrauterine growth restriction in late pregnancy can contribute to adverse long-term metabolic health in the offspring. In the present study we used an animal (sheep) model of maternal dietary manipulation in late pregnancy, combined with exposure of the offspring to a low-activity, obesogenic environment after weaning, to characterise the effects on glucose homeostasis. Dizygotic twin-pregnant sheep were either fed to 60% of requirements (nutrient restriction (R)) or fed ad libitum (~140% of requirements (A)) from 110 days gestation until term (~147 days). After weaning (~3 months of age), the offspring were kept in either a standard (in order to remain lean) or low-activity, obesogenic environment. R mothers gained less weight and produced smaller offspring. As adults, obese offspring were heavier and fatter with reduced glucose tolerance, regardless of maternal diet. Molecular markers of stress and autophagy in liver and adipose tissue were increased with obesity, with gene expression of hepatic glucose-related protein 78 (Grp78) and omental activation transcription factor 6 (Atf6), Grp78 and ER stress degradation enhancer molecule 1 (Edem1) only being increased in R offspring. In conclusion, the adverse effect of juvenile-onset obesity on insulin-responsive tissues can be amplified by previous exposure to a suboptimal nutritional environment in utero, thereby contributing to earlier onset of insulin resistance.
Collapse
Affiliation(s)
- Vivek Saroha
- Early Life Research Unit, Academic Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - Neele S Dellschaft
- Early Life Research Unit, Academic Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - Duane H Keisler
- Department of Animal Science, University of Missouri, Columbia, MO 65211, USA
| | - David S Gardner
- School of Veterinary Medicine and Science, Sutton Bonington Campus, University of Nottingham, Nottingham LE12 5RD, UK
| | - Helen Budge
- Early Life Research Unit, Academic Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - Sylvain P Sebert
- Early Life Research Unit, Academic Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - Michael E Symonds
- Early Life Research Unit, Academic Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
6
|
Berti C, Cetin I, Agostoni C, Desoye G, Devlieger R, Emmett PM, Ensenauer R, Hauner H, Herrera E, Hoesli I, Krauss-Etschmann S, Olsen SF, Schaefer-Graf U, Schiessl B, Symonds ME, Koletzko B. Pregnancy and Infants' Outcome: Nutritional and Metabolic Implications. Crit Rev Food Sci Nutr 2016; 56:82-91. [PMID: 24628089 DOI: 10.1080/10408398.2012.745477] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pregnancy is a complex period of human growth, development, and imprinting. Nutrition and metabolism play a crucial role for the health and well-being of both mother and fetus, as well as for the long-term health of the offspring. Nevertheless, several biological and physiological mechanisms related to nutritive requirements together with their transfer and utilization across the placenta are still poorly understood. In February 2009, the Child Health Foundation invited leading experts of this field to a workshop to critically review and discuss current knowledge, with the aim to highlight priorities for future research. This paper summarizes our main conclusions with regards to maternal preconceptional body mass index, gestational weight gain, placental and fetal requirements in relation to adverse pregnancy and long-term outcomes of the fetus (nutritional programming). We conclude that there is an urgent need to develop further human investigations aimed at better understanding of the basis of biochemical mechanisms and pathophysiological events related to maternal-fetal nutrition and offspring health. An improved knowledge would help to optimize nutritional recommendations for pregnancy.
Collapse
Affiliation(s)
- C Berti
- a Unit of Obstetrics & Gynecology, Department of Biomedical and Clinical Sciences, Hospital 'L. Sacco', and Center for Fetal Research Giorgio Pardi , University of Milan , Milan , Italy
| | - I Cetin
- a Unit of Obstetrics & Gynecology, Department of Biomedical and Clinical Sciences, Hospital 'L. Sacco', and Center for Fetal Research Giorgio Pardi , University of Milan , Milan , Italy.,b Department of Clinical Sciences and Community Health, University of Milan, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - C Agostoni
- b Department of Clinical Sciences and Community Health, University of Milan, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - G Desoye
- c Department of Obstetrics & Gynaecology , Medical University of Graz , Graz , Austria
| | - R Devlieger
- d Department of Obstetrics & Gynaecology , University Hospitals K.U. Leuven , Leuven , Belgium
| | - P M Emmett
- e Centre for Child & Adolescent Health, School of Social & Community Medicine , University of Bristol , Bristol , United Kingdom
| | - R Ensenauer
- f Research Center, Dr. von Hauner Children's Hospital, Klinikum der Ludwig-Maximilians-Universität München , Munich , Germany
| | - H Hauner
- g Else Kroener-Fresenius-Centre for Nutritional Medicine, Klinikum rechts der Isar , Technical University of Munich , Munich , Germany
| | - E Herrera
- h Unit of Biochemistry & Molecular Biology, Universidad San Pablo CEU , Madrid , Spain
| | - I Hoesli
- i Department of Obstetrics and Gynaecology , University Hospital of Basel , Basel , Switzerland
| | - S Krauss-Etschmann
- j Comprehensive Pneumology Center, Ludwig Maximilians University Hospital , Munich , Germany.,k Helmholtz Zentrum München , Munich , Germany
| | - S F Olsen
- l Centre for Fetal Programming, Statens Serum Institut , Copenhagen , Denmark
| | - U Schaefer-Graf
- m Department of Obstetrics & Gynecology , Berlin Center for Diabetes & Pregnancy, St. Joseph Hospital , Berlin , Germany
| | - B Schiessl
- n Fetal Maternal Unit, Department of Obstetrics & Gynecology , University of Munich , Munich , Germany
| | - M E Symonds
- o The Early Life Nutrition Research Unit, Academic Child Health, School of Clinical Sciences, University Hospital of Nottingham , Nottingham , United Kingdom
| | - B Koletzko
- p Dr. von Hauner Children's Hospital , University of Munich Medical Centre , Munich , Germany
| |
Collapse
|
7
|
Ojha S, Fainberg HP, Wilson V, Pelella G, Castellanos M, May ST, Lotto AA, Sacks H, Symonds ME, Budge H. Gene pathway development in human epicardial adipose tissue during early life. JCI Insight 2016; 1:e87460. [PMID: 27699231 DOI: 10.1172/jci.insight.87460] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Studies in rodents and newborn humans demonstrate the influence of brown adipose tissue (BAT) in temperature control and energy balance and a critical role in the regulation of body weight. Here, we obtained samples of epicardial adipose tissue (EAT) from neonates, infants, and children in order to evaluate changes in their transcriptional landscape by applying a systems biology approach. Surprisingly, these analyses revealed that the transition to infancy is a critical stage for changes in the morphology of EAT and is reflected in unique gene expression patterns of a substantial proportion of thermogenic gene transcripts (~10%). Our results also indicated that the pattern of gene expression represents a distinct developmental stage, even after the rebound in abundance of thermogenic genes in later childhood. Using weighted gene coexpression network analyses, we found precise anthropometric-specific correlations with changes in gene expression and the decline of thermogenic capacity within EAT. In addition, these results indicate a sequential order of transcriptional events affecting cellular pathways, which could potentially explain the variation in the amount, or activity, of BAT in adulthood. Together, these results provide a resource to elucidate gene regulatory mechanisms underlying the progressive development of BAT during early life.
Collapse
Affiliation(s)
- Shalini Ojha
- Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, University Hospital, The University of Nottingham, Nottingham, United Kingdom
| | - Hernan P Fainberg
- Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, University Hospital, The University of Nottingham, Nottingham, United Kingdom
| | - Victoria Wilson
- Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, University Hospital, The University of Nottingham, Nottingham, United Kingdom
| | - Giuseppe Pelella
- University Hospitals Leicester NHS Trust, Leicester, United Kingdom
| | - Marcos Castellanos
- Nottingham Arabidopsis Stock Centre, School of Biosciences, The University of Nottingham, Nottingham, United Kingdom
| | - Sean T May
- Nottingham Arabidopsis Stock Centre, School of Biosciences, The University of Nottingham, Nottingham, United Kingdom
| | - Attilio A Lotto
- University Hospitals Leicester NHS Trust, Leicester, United Kingdom
| | - Harold Sacks
- VA Endocrinology and Diabetes Division, VA Greater Los Angeles Healthcare System, and Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Michael E Symonds
- Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, University Hospital, The University of Nottingham, Nottingham, United Kingdom
| | - Helen Budge
- Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, University Hospital, The University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
8
|
Symonds ME, Dellschaft N, Pope M, Birtwistle M, Alagal R, Keisler D, Budge H. Developmental programming, adiposity, and reproduction in ruminants. Theriogenology 2016; 86:120-9. [PMID: 27173959 DOI: 10.1016/j.theriogenology.2016.04.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 02/29/2016] [Accepted: 03/14/2016] [Indexed: 01/21/2023]
Abstract
Although sheep have been widely adopted as an animal model for examining the timing of nutritional interventions through pregnancy on the short- and long-term outcomes, only modest programming effects have been seen. This is due in part to the mismatch in numbers of twins and singletons between study groups as well as unequal numbers of males and females. Placental growth differs between singleton and twin pregnancies which can result in different body composition in the offspring. One tissue that is especially affected is adipose tissue which in the sheep fetus is primarily located around the kidneys and heart plus the sternal/neck region. Its main role is the rapid generation of heat due to activation of the brown adipose tissue-specific uncoupling protein 1 at birth. The fetal adipose tissue response to suboptimal maternal food intake at defined stages of development differs between the perirenal abdominal and pericardial depots, with the latter being more sensitive. Fetal adipose tissue growth may be mediated in part by changes in leptin status of the mother which are paralleled in the fetus. Then, over the first month of life plasma leptin is higher in females than males despite similar adiposity, when fat is the fastest growing tissue with the sternal/neck depot retaining uncoupling protein 1, whereas other depots do not. Future studies should take into account the respective effects of fetal number and sex to provide more detailed insights into the mechanisms by which adipose and related tissues can be programmed in utero.
Collapse
Affiliation(s)
- M E Symonds
- Early Life Research Unit, Academic Division of Child Health, Obstetrics & Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham, UK.
| | - N Dellschaft
- Early Life Research Unit, Academic Division of Child Health, Obstetrics & Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham, UK
| | - M Pope
- Early Life Research Unit, Academic Division of Child Health, Obstetrics & Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham, UK
| | - M Birtwistle
- Early Life Research Unit, Academic Division of Child Health, Obstetrics & Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham, UK
| | - R Alagal
- Early Life Research Unit, Academic Division of Child Health, Obstetrics & Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham, UK
| | - D Keisler
- Department of Animal Science, University of Missouri, Columbia, Missouri, USA
| | - H Budge
- Early Life Research Unit, Academic Division of Child Health, Obstetrics & Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham, UK
| |
Collapse
|
9
|
Palaniswamy S, Williams D, Järvelin MR, Sebert S. Vitamin D and the Promotion of Long-Term Metabolic Health from a Programming Perspective. Nutr Metab Insights 2016; 8:11-21. [PMID: 26843814 PMCID: PMC4737521 DOI: 10.4137/nmi.s29526] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/04/2015] [Accepted: 11/06/2015] [Indexed: 01/14/2023] Open
Abstract
Studies linking vitamin D and long-term metabolic health have generated much debate. Recommendations for the intake of vitamin D by the general public and by the health care professionals have been complicated by a number of inconsistencies in the literature. These caveats relate to the methodological approaches, differences in the populations (and the species) of study, and the definitions used for thresholds of vitamin D status. This review addresses current evidence available for assessing the potential programming of long-term metabolic health of offspring by maternal vitamin D status in pregnancy. It summarizes knowledge on the early origins of metabolic health and analyzes evidence for an association between the vitamin D status in pregnancy and maternal and fetal health status. In addition, we analyze the link between the regulation of inflammation and the vitamin D status in the general population to inform on the general mechanisms through which early vitamin D might affect the programming of long-term health. The evidence suggests an association between the vitamin D status in early life and the programming of long-term health. However, to the best of our knowledge, the current finding is insufficient to draw a final conclusion for evidence-based preventive actions. The data warrant replication in prospective studies and additional research substantiating the causal factors and pathways.
Collapse
Affiliation(s)
- Saranya Palaniswamy
- Center for Life-Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland.; Biocenter Oulu, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Dylan Williams
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, UK
| | - Marjo-Riitta Järvelin
- Center for Life-Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland.; Biocenter Oulu, Faculty of Medicine, University of Oulu, Oulu, Finland.; Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, UK.; MRC-PHE Centre for Environment & Health, School of Public Health, Imperial College, London, UK.; Unit of Primary Care, Oulu University Hospital, Oulu, Finland
| | - Sylvain Sebert
- Center for Life-Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland.; Biocenter Oulu, Faculty of Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
10
|
Maternal health and eating habits: metabolic consequences and impact on child health. Trends Mol Med 2015; 21:126-33. [PMID: 25662028 DOI: 10.1016/j.molmed.2014.12.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 12/15/2014] [Accepted: 12/16/2014] [Indexed: 12/21/2022]
Abstract
Apart from direct inheritance and the effects of a shared environment, maternal health, eating habits and diet can affect offspring health by developmental programming. Suboptimal maternal nutrition (i.e., either a reduction or an increase above requirement) or other insults experienced by the developing fetus can induce significant changes in adipose tissue and brain development, energy homeostasis, and the structure of vital organs. These can produce long-lasting adaptations that influence later energy balance, and increase the susceptibility of that individual to obesity and the components of the metabolic syndrome. Studies that elucidate the mechanisms behind these associations will have a positive impact on the health of the future adult population and may help to contain the obesity epidemic.
Collapse
|
11
|
Dellschaft NS, Alexandre-Gouabau MC, Gardner DS, Antignac JP, Keisler DH, Budge H, Symonds ME, Sebert SP. Effect of pre- and postnatal growth and post-weaning activity on glucose metabolism in the offspring. J Endocrinol 2015; 224:171-82. [PMID: 25416820 DOI: 10.1530/joe-14-0600] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Maternal caloric restriction during late gestation reduces birth weight, but whether long-term adverse metabolic outcomes of intra-uterine growth retardation (IUGR) are dependent on either accelerated postnatal growth or exposure to an obesogenic environment after weaning is not established. We induced IUGR in twin-pregnant sheep using a 40% maternal caloric restriction commencing from 110 days of gestation until term (∼147 days), compared with mothers fed to 100% of requirements. Offspring were reared either as singletons to accelerate postnatal growth or as twins to achieve standard growth. To promote an adverse phenotype in young adulthood, after weaning, offspring were reared under a low-activity obesogenic environment with the exception of a subgroup of IUGR offspring, reared as twins, maintained in a standard activity environment. We assessed glucose tolerance together with leptin and cortisol responses to feeding in young adulthood when the hypothalamus was sampled for assessment of genes regulating appetite control, energy and endocrine sensitivity. Caloric restriction reduced maternal plasma glucose, raised non-esterified fatty acids, and changed the metabolomic profile, but had no effect on insulin, leptin, or cortisol. IUGR offspring whose postnatal growth was enhanced and were obese showed insulin and leptin resistance plus raised cortisol. This was accompanied by increased hypothalamic gene expression for energy and glucocorticoid sensitivity. These long-term adaptations were reduced but not normalized in IUGR offspring whose postnatal growth was not accelerated and remained lean in a standard post-weaning environment. IUGR results in an adverse metabolic phenotype, especially when postnatal growth is enhanced and offspring progress to juvenile-onset obesity.
Collapse
Affiliation(s)
- Neele S Dellschaft
- Early Life Research UnitAcademic Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham NG7 2UH, UKINRA and University of NantesUMR-1280 Physiologie des Adaptations Nutritionnelles, CHU Hôtel Dieu, 44093 Nantes cedex 1, FranceSchool of Veterinary Medicine and ScienceThe University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKInstitute of Health SciencesCentre for Life-Course Epidemiology, and Biocentre Oulu, University of Oulu, Aapistie 5B, 90014 Oulu, FinlandOnirisLaboratoire d'Etude des Résidus et Contaminants dans les Aliments, LUNAM Université, USC INRA 1329, Nantes, FranceDepartment of Animal ScienceUniversity of Missouri, Columbia, Missouri 65211, USA Early Life Research UnitAcademic Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham NG7 2UH, UKINRA and University of NantesUMR-1280 Physiologie des Adaptations Nutritionnelles, CHU Hôtel Dieu, 44093 Nantes cedex 1, FranceSchool of Veterinary Medicine and ScienceThe University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKInstitute of Health SciencesCentre for Life-Course Epidemiology, and Biocentre Oulu, University of Oulu, Aapistie 5B, 90014 Oulu, FinlandOnirisLaboratoire d'Etude des Résidus et Contaminants dans les Aliments, LUNAM Université, USC INRA 1329, Nantes, FranceDepartment of Animal ScienceUniversity of Missouri, Columbia, Missouri 65211, USA
| | - Marie-Cecile Alexandre-Gouabau
- Early Life Research UnitAcademic Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham NG7 2UH, UKINRA and University of NantesUMR-1280 Physiologie des Adaptations Nutritionnelles, CHU Hôtel Dieu, 44093 Nantes cedex 1, FranceSchool of Veterinary Medicine and ScienceThe University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKInstitute of Health SciencesCentre for Life-Course Epidemiology, and Biocentre Oulu, University of Oulu, Aapistie 5B, 90014 Oulu, FinlandOnirisLaboratoire d'Etude des Résidus et Contaminants dans les Aliments, LUNAM Université, USC INRA 1329, Nantes, FranceDepartment of Animal ScienceUniversity of Missouri, Columbia, Missouri 65211, USA
| | - David S Gardner
- Early Life Research UnitAcademic Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham NG7 2UH, UKINRA and University of NantesUMR-1280 Physiologie des Adaptations Nutritionnelles, CHU Hôtel Dieu, 44093 Nantes cedex 1, FranceSchool of Veterinary Medicine and ScienceThe University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKInstitute of Health SciencesCentre for Life-Course Epidemiology, and Biocentre Oulu, University of Oulu, Aapistie 5B, 90014 Oulu, FinlandOnirisLaboratoire d'Etude des Résidus et Contaminants dans les Aliments, LUNAM Université, USC INRA 1329, Nantes, FranceDepartment of Animal ScienceUniversity of Missouri, Columbia, Missouri 65211, USA
| | - Jean-Philippe Antignac
- Early Life Research UnitAcademic Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham NG7 2UH, UKINRA and University of NantesUMR-1280 Physiologie des Adaptations Nutritionnelles, CHU Hôtel Dieu, 44093 Nantes cedex 1, FranceSchool of Veterinary Medicine and ScienceThe University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKInstitute of Health SciencesCentre for Life-Course Epidemiology, and Biocentre Oulu, University of Oulu, Aapistie 5B, 90014 Oulu, FinlandOnirisLaboratoire d'Etude des Résidus et Contaminants dans les Aliments, LUNAM Université, USC INRA 1329, Nantes, FranceDepartment of Animal ScienceUniversity of Missouri, Columbia, Missouri 65211, USA
| | - Duane H Keisler
- Early Life Research UnitAcademic Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham NG7 2UH, UKINRA and University of NantesUMR-1280 Physiologie des Adaptations Nutritionnelles, CHU Hôtel Dieu, 44093 Nantes cedex 1, FranceSchool of Veterinary Medicine and ScienceThe University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKInstitute of Health SciencesCentre for Life-Course Epidemiology, and Biocentre Oulu, University of Oulu, Aapistie 5B, 90014 Oulu, FinlandOnirisLaboratoire d'Etude des Résidus et Contaminants dans les Aliments, LUNAM Université, USC INRA 1329, Nantes, FranceDepartment of Animal ScienceUniversity of Missouri, Columbia, Missouri 65211, USA
| | - Helen Budge
- Early Life Research UnitAcademic Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham NG7 2UH, UKINRA and University of NantesUMR-1280 Physiologie des Adaptations Nutritionnelles, CHU Hôtel Dieu, 44093 Nantes cedex 1, FranceSchool of Veterinary Medicine and ScienceThe University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKInstitute of Health SciencesCentre for Life-Course Epidemiology, and Biocentre Oulu, University of Oulu, Aapistie 5B, 90014 Oulu, FinlandOnirisLaboratoire d'Etude des Résidus et Contaminants dans les Aliments, LUNAM Université, USC INRA 1329, Nantes, FranceDepartment of Animal ScienceUniversity of Missouri, Columbia, Missouri 65211, USA
| | - Michael E Symonds
- Early Life Research UnitAcademic Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham NG7 2UH, UKINRA and University of NantesUMR-1280 Physiologie des Adaptations Nutritionnelles, CHU Hôtel Dieu, 44093 Nantes cedex 1, FranceSchool of Veterinary Medicine and ScienceThe University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKInstitute of Health SciencesCentre for Life-Course Epidemiology, and Biocentre Oulu, University of Oulu, Aapistie 5B, 90014 Oulu, FinlandOnirisLaboratoire d'Etude des Résidus et Contaminants dans les Aliments, LUNAM Université, USC INRA 1329, Nantes, FranceDepartment of Animal ScienceUniversity of Missouri, Columbia, Missouri 65211, USA
| | - Sylvain P Sebert
- Early Life Research UnitAcademic Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham NG7 2UH, UKINRA and University of NantesUMR-1280 Physiologie des Adaptations Nutritionnelles, CHU Hôtel Dieu, 44093 Nantes cedex 1, FranceSchool of Veterinary Medicine and ScienceThe University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKInstitute of Health SciencesCentre for Life-Course Epidemiology, and Biocentre Oulu, University of Oulu, Aapistie 5B, 90014 Oulu, FinlandOnirisLaboratoire d'Etude des Résidus et Contaminants dans les Aliments, LUNAM Université, USC INRA 1329, Nantes, FranceDepartment of Animal ScienceUniversity of Missouri, Columbia, Missouri 65211, USA Early Life Research UnitAcademic Division of Child Health, Obstetrics and Gynaecology, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham NG7 2UH, UKINRA and University of NantesUMR-1280 Physiologie des Adaptations Nutritionnelles, CHU Hôtel Dieu, 44093 Nantes cedex 1, FranceSchool of Veterinary Medicine and ScienceThe University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKInstitute of Health SciencesCentre for Life-Course Epidemiology, and Biocentre Oulu, University of Oulu, Aapistie 5B, 90014 Oulu, FinlandOnirisLaboratoire d'Etude des Résidus et Contaminants dans les Aliments, LUNAM Université, USC INRA 1329, Nantes, FranceDepartment of Animal ScienceUniversity of Missouri, Columbia, Missouri 65211, USA
| |
Collapse
|
12
|
Ojha S, Saroha V, Symonds ME, Budge H. Excess nutrient supply in early life and its later metabolic consequences. Clin Exp Pharmacol Physiol 2014; 40:817-23. [PMID: 23350968 DOI: 10.1111/1440-1681.12061] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 01/17/2013] [Accepted: 01/20/2013] [Indexed: 12/20/2022]
Abstract
Suboptimal nutrition in early life, both in utero and during infancy, is linked to increased risk of adult obesity and its associated adverse metabolic health problems. Excess nutrient supply during early life can lead to metabolic programming in the offspring. Such overnutrition can occur in the offspring of obese mothers, the offspring of mothers who gain excess weight during gestation, infants of diabetic mothers and infants who undergo rapid growth, particularly weight gain, during early infancy. Postnatal overnutrition is particularly detrimental for infants who are born small for gestational age, who are overfed to attain 'catch-up growth'. Potential mechanisms underlying metabolic programming that results from excess nutrition during early life include resetting of hypothalamic energy sensing and appetite regulation, altered adipose tissue insulin sensitivity and impaired brown adipose tissue function. More detailed understanding of the mechanisms involved in metabolic programming could enable the development of therapeutic strategies for ameliorating its ill effects. Research in this field could potentially identify optimal and appropriate preventative interventions for a burgeoning population at risk of increased mortality and morbidity from obesity and its concomitant metabolic conditions.
Collapse
Affiliation(s)
- Shalini Ojha
- The Early Life Nutrition Research Unit, Academic Division of Child Health, School of Clinical Sciences, University Hospital, The University of Nottingham, Nottingham, UK
| | | | | | | |
Collapse
|
13
|
Bloor ID, Symonds ME. Sexual dimorphism in white and brown adipose tissue with obesity and inflammation. Horm Behav 2014; 66:95-103. [PMID: 24589990 DOI: 10.1016/j.yhbeh.2014.02.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 12/24/2022]
Abstract
This article is part of a Special Issue "Energy Balance". Obesity and its associated comorbidities remain at epidemic levels globally and show no signs of abatement in either adult or child populations. White adipose tissue has long been established as an endocrine signalling organ possessing both metabolic and immune functions. This role can become dysregulated following excess adiposity caused by adipocyte hypertrophy and hyperplasia. In contrast, brown adipose tissue (BAT) is only present in comparatively small amounts in the body but can significantly impact on heat production, and thus could prevent excess white adiposity. Obesity and associated risk factors for adverse metabolic health are not only linked with enlarged fat mass but also are dependent on its anatomical deposition. In addition, numerous studies have revealed a disparity in white adipose tissue deposition prior to and during the development of obesity between the sexes. Females therefore tend to develop a greater abundance of femoral and gluteal subcutaneous fat whereas males exhibit more central adiposity. In females, lower body subcutaneous adipose tissue depots appear to possess a greater capacity for lipid storage, enhanced lipolytic flux and hyperplastic tissue remodelling compared to visceral adipocytes. These differences are acknowledged to contribute to the poorer metabolic and inflammatory profiles observed in males. Importantly, the converse outcomes between sexes disappear after the menopause, suggesting a role for sex hormones within the onset of metabolic complications with obesity. This review further considers how BAT impacts upon on the relationship between excess adiposity, gender, inflammation and endocrine signalling and could thus ultimately be a target to prevent obesity.
Collapse
Affiliation(s)
- Ian D Bloor
- Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, The University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Michael E Symonds
- Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, The University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
| |
Collapse
|
14
|
Mathias PCF, Elmhiri G, de Oliveira JC, Delayre-Orthez C, Barella LF, Tófolo LP, Fabricio GS, Chango A, Abdennebi-Najar L. Maternal diet, bioactive molecules, and exercising as reprogramming tools of metabolic programming. Eur J Nutr 2014; 53:711-22. [DOI: 10.1007/s00394-014-0654-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 01/12/2014] [Indexed: 12/21/2022]
|
15
|
Symonds ME, Sebert SP, Budge H. Dangers of dieting: what advice should be given to obese expectant mothers? ACTA ACUST UNITED AC 2014. [DOI: 10.1586/eog.09.68] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
16
|
Fainberg HP, Sharkey D, Sebert S, Wilson V, Pope M, Budge H, Symonds ME. Suboptimal maternal nutrition during early fetal kidney development specifically promotes renal lipid accumulation following juvenile obesity in the offspring. Reprod Fertil Dev 2014; 25:728-36. [PMID: 22951182 DOI: 10.1071/rd12037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 06/06/2012] [Indexed: 11/23/2022] Open
Abstract
Reduced maternal food intake between early-to-mid gestation results in tissue-specific adaptations in the offspring following juvenile-onset obesity that are indicative of insulin resistance. The aim of the present study was to establish the extent to which renal ectopic lipid accumulation, as opposed to other markers of renal stress, such as iron deposition and apoptosis, is enhanced in obese offspring born to mothers nutrient restricted (NR) throughout early fetal kidney development. Pregnant sheep were fed either 100% (control) or NR (i.e. fed 50% of their total metabolisable energy requirement from 30-80 days gestation and 100% at all other times). At weaning, offspring were made obese and, at approximately 1 year, kidneys were sampled. Triglyceride content, HIF-1α gene expression and the protein abundance of the outer-membrane transporter voltage-dependent anion-selective channel protein (VDAC)-I on the kidney cortex were increased in obese offspring born to NR mothers compared with those born to controls, which exhibited increased iron accumulation within the tubular epithelial cells and increased gene expression of the death receptor Fas. In conclusion, suboptimal maternal nutrition coincident with early fetal kidney development results in enhanced renal lipid deposition following juvenile obesity and could accelerate the onset of the adverse metabolic, rather than cardiovascular, symptoms accompanying the metabolic syndrome.
Collapse
Affiliation(s)
- H P Fainberg
- Early Life Nutrition Research Unit, Academic Child Health, School of Medicine, University Hospital, Nottingham NG7 2UH, UK
| | | | | | | | | | | | | |
Collapse
|
17
|
Pope M, Budge H, Symonds ME. The developmental transition of ovine adipose tissue through early life. Acta Physiol (Oxf) 2014; 210:20-30. [PMID: 23351024 DOI: 10.1111/apha.12053] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 11/14/2012] [Accepted: 12/13/2012] [Indexed: 11/30/2022]
Abstract
AIM Hypothermia induced by cold exposure at birth is prevented in sheep by the rapid onset of non-shivering thermogenesis in brown adipose tissue (BAT). Changes in adipose tissue composition in early life are therefore essential for survival but also influence adiposity in later life and were thus examined in detail during early development. METHODS Changes in adipose composition were investigated by immunohistochemistry and qRT-PCR between the period from the first appearance of adipose in the mid gestation foetus, through birth and up to 1 month of age. RESULTS We identified four distinct phases of development, each associated with pronounced changes in tissue histology and in distribution of the BAT specific uncoupling protein (UCP)1. At mid gestation, perirenal adipose tissue exhibited a dense proliferative, structure marked by high expression of KI-67 but with no UCP1 or visible lipid droplets. By late gestation large quantities of UCP1 were present, lipid storage was evident and expression of BAT-related genes were abundant (e.g. prolactin and β3 receptors). Subsequently, within 12 h of birth, the depot was largely depleted of lipid and expression of genes such as UCP1, PGC1α, CIDEA peaked. By 30 days UCP1 was undetectable and the depot contained large lipid droplets; however, genes characteristic of BAT (e.g. PRDM16 and BMP7) and most characteristic of white adipose tissue (e.g. leptin and RIP140) were still abundant. CONCLUSION Adipose tissue undergoes profound compositional changes in early life, of which an increased understanding could offer potential interventions to retain BAT in later life.
Collapse
Affiliation(s)
- M. Pope
- Early Life Nutrition Research Unit, Academic Division of Child Health; School of Medicine, University Hospital, The University of Nottingham; Nottingham UK
| | - H. Budge
- Early Life Nutrition Research Unit, Academic Division of Child Health; School of Medicine, University Hospital, The University of Nottingham; Nottingham UK
| | - M. E. Symonds
- Early Life Nutrition Research Unit, Academic Division of Child Health; School of Medicine, University Hospital, The University of Nottingham; Nottingham UK
| |
Collapse
|
18
|
Ojha S, Robinson L, Symonds ME, Budge H. Suboptimal maternal nutrition affects offspring health in adult life. Early Hum Dev 2013; 89:909-13. [PMID: 24080391 DOI: 10.1016/j.earlhumdev.2013.08.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/26/2013] [Indexed: 01/24/2023]
Abstract
Suboptimal maternal nutrition during pregnancy is prevalent and compromises fetal development. Physiological and metabolic adaptations made by the fetus to an inadequate, or excess, maternal nutritional environment, may promote immediate survival but are lasting, conferring significantly increased risks of ill health in childhood and adulthood. In addition, such fetal adaptations are particularly detrimental when nutrient supply is no longer constrained in contemporary nutrient rich environments. Given the prevalence of suboptimal maternal nutritional environments during fetal development, effective prevention, early detection and therapeutic interventions to reduce the increased risks on population health must be a health priority. Therefore, the mechanisms of these lasting in utero adaptations are highly relevant to establishing how exposure to a suboptimal nutritional environment impacts on the health of current generations living in an environment challenged by excess nutrition.
Collapse
Affiliation(s)
- Shalini Ojha
- The Early Life Research Unit, Academic Child Health, School of Medicine, The University of Nottingham, Nottingham, NG7 2UH, UK.
| | | | | | | |
Collapse
|
19
|
Bloor ID, Sébert SP, Saroha V, Gardner DS, Keisler DH, Budge H, Symonds ME, Mahajan RP. Sex differences in metabolic and adipose tissue responses to juvenile-onset obesity in sheep. Endocrinology 2013; 154:3622-31. [PMID: 23885012 DOI: 10.1210/en.2013-1207] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Sex is a major factor determining adipose tissue distribution and the subsequent adverse effects of obesity-related disease including type 2 diabetes. The role of gender on juvenile obesity and the accompanying metabolic and inflammatory responses is not well established. Using an ovine model of juvenile onset obesity induced by reduced physical activity, we examined the effect of gender on metabolic, circulatory, and related inflammatory and energy-sensing profiles of the major adipose tissue depots. Despite a similar increase in fat mass with obesity between genders, males demonstrated a higher storage capacity of lipids within perirenal-abdominal adipocytes and exhibited raised insulin. In contrast, obese females became hypercortisolemic, a response that was positively correlated with central fat mass. Analysis of gene expression in perirenal-abdominal adipose tissue demonstrated the stimulation of inflammatory markers in males, but not females, with obesity. Obese females displayed increased expression of genes involved in the glucocorticoid axis and energy sensing in perirenal-abdominal, but not omental, adipose tissue, indicating a depot-specific mechanism that may be protective from the adverse effects of metabolic dysfunction and inflammation. In conclusion, young males are at a greater risk than females to the onset of comorbidities associated with juvenile-onset obesity. These sex-specific differences in cortisol and adipose tissue could explain the earlier onset of the metabolic-related diseases in males compared with females after obesity.
Collapse
Affiliation(s)
- Ian D Bloor
- Academic Division of Child Health School of Clinical Sciences, E Floor, East Block, Queen's Medical Centre, University Hospital, The University of Nottingham, Nottingham NG7 2UH, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Fowden AL, Jellyman JK, Valenzuela OA, Forhead AJ. Nutritional Programming of Intrauterine Development: A Concept Applicable to the Horse? J Equine Vet Sci 2013. [DOI: 10.1016/j.jevs.2013.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
21
|
Nutritional regulation of fetal growth and implications for productive life in ruminants. Animal 2012; 4:1075-83. [PMID: 22444610 DOI: 10.1017/s1751731110000479] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The maternal nutritional and metabolic environment is critical in determining not only the reproductive success but also the long-term health and viability of the offspring. Changes in maternal diet at defined stages of gestation coincident with different stages of development can have pronounced effects on organ and tissue function in later life. This includes adipose tissue for which differential effects are observed between brown and white adipose tissues. One early, critical window of organ development in the ruminant relates to the period covering uterine attachment, or implantation, and rapid placental growth. During this period, there is pronounced cell division within developing organelles in many fetal tissues, leading to their structural development. In sheep, a 50% global reduction in caloric intake over this specific period profoundly affects placental growth and morphology, resulting in reduced placentome weight. This occurs in conjunction with a lower capacity to inactivate maternal cortisol through the enzyme 11β-hydroxysteroid dehydrogenase type 2 in response to a decrease in maternal plasma cortisol in early gestation. The birth weight of the offspring is, however, unaffected by this dietary manipulation and, although they possess more fat, this adaptation does not persist into adulthood when they become equally obese as those born to control fed mothers. Subsequently, after birth, further changes in fat development occur which impact on both glucocorticoid action and inflammatory responses. These adaptations can include changes in the relative populations of both brown and white adipocytes for which prolactin acting through its receptor appears to have a prominent role. Earlier when in utero nutrient restricted (i.e. between early-to-mid gestation) offspring are exposed to an obesogenic postnatal environment; they exhibit an exaggerated insulin response, which is accompanied by a range of amplified and thus, adverse, physiological or metabolic responses to obesity. These types of adaptations are in marked contrast to the effect of late gestational nutrient restriction, which results in reduced fat mass at birth. As young adults, however, fat mass is increased and, although basal insulin is unaffected, these offspring are insulin resistant. In conclusion, changes in nutrient supply to either the mother and/or her fetus can have profound effects on a range of metabolically important tissues. These have the potential to either exacerbate, or protect from, the adverse effects of later obesity and accompanying complications in the resulting offspring.
Collapse
|
22
|
Bloor ID, Sebert SP, Mahajan RP, Symonds ME. The influence of sex on early stage markers of kidney dysfunction in response to juvenile obesity. Hypertension 2012; 60:991-7. [PMID: 22949537 DOI: 10.1161/hypertensionaha.112.195412] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Changes within the kidney in response to obesity are critical in determining the magnitude of later dysfunction. However, the cause of this process in response to juvenile onset obesity and how it can be determined by sex is poorly understood. We therefore examined the effect of juvenile obesity induced by exposure to a restricted activity environment from weaning until early adulthood on the molecular responses within the kidney together with glomerular area and nucleated cell number. This was stratified by sex and was undertaken in a sheep model of early obesity. Despite a similar magnitude of increase in fat mass with obesity onset between sexes, adverse effects on glomerular area and cell number together with raised gene expression within the kidney only occurred in males. Irrespective of obesity, gene expression of C-C motif receptor 2 was higher, and interleukin-6 lower, in male kidneys compared with female kidneys. The effects of sex on molecular differences within the kidney were amplified with obesity, which had no effect on any gene studied in females but had an enhanced response in males. Obese males therefore showed increased gene expression of a range of markers relating to the glucocorticoid axis, inflammation, and lipid sensing. In conclusion, young females were protected from adverse renal effects of obesity, which results in very little inflammatory or related responses. Our findings emphasize the critical importance of sex specificity in disease pathogenesis. An increased understanding of the specific mechanisms will have important implications for therapeutic strategies aimed at preventing adverse consequences of obesity.
Collapse
Affiliation(s)
- Ian D Bloor
- Early Life Nutrition Research Unit, Academic Division of Child Health, School of Medicine, E Floor, East Block, Queen's Medical Centre, University Hospital, Nottingham, United Kingdom
| | | | | | | |
Collapse
|
23
|
Symonds ME, Pope M, Sharkey D, Budge H. Adipose tissue and fetal programming. Diabetologia 2012; 55:1597-606. [PMID: 22402988 DOI: 10.1007/s00125-012-2505-5] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 01/12/2012] [Indexed: 10/28/2022]
Abstract
Adipose tissue function changes with development. In the newborn, brown adipose tissue (BAT) is essential for ensuring effective adaptation to the extrauterine environment, and its growth during gestation is largely dependent on glucose supply from the mother to the fetus. The amount, location and type of adipose tissue deposited can also determine fetal glucose homeostasis. Adipose tissue first appears at around mid-gestation. Total adipose mass then increases through late gestation, when it comprises a mixture of white and brown adipocytes. BAT possesses a unique uncoupling protein, UCP1, which is responsible for the rapid generation of large amounts of heat at birth. Then, during postnatal life some, but not all, depots are replaced by white fat. This process can be utilised to investigate the physiological conversion of brown to white fat, and how it is re-programmed by nutritional changes in pre- and postnatal environments. A reduction in early BAT deposition may perpetuate through the life cycle, thereby suppressing energy expenditure and ultimately promoting obesity. Normal fat development profiles in the offspring are modified by changes in maternal diet at defined stages of pregnancy, ultimately leading to adverse long-term outcomes. For example, excess macrophage accumulation and the onset of insulin resistance occur in an adipose tissue depot-specific manner in offspring born to mothers fed a suboptimal diet from early to mid-gestation. In conclusion, the growth of the different fetal adipose tissue depots varies according to maternal diet and, if challenged in later life, this can contribute to insulin resistance and impaired glucose homeostasis.
Collapse
Affiliation(s)
- M E Symonds
- The Early Life Nutrition Research Unit, Academic Division of Child Health, School of Clinical Sciences, University Hospital, Nottingham, NG7 2UH, UK.
| | | | | | | |
Collapse
|
24
|
Sebert S, Sharkey D, Budge H, Symonds ME. The early programming of metabolic health: is epigenetic setting the missing link? Am J Clin Nutr 2011; 94:1953S-1958S. [PMID: 21543542 DOI: 10.3945/ajcn.110.001040] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Adult health is dependent, in part, on maternal nutrition and growth during early life, which may independently affect insulin sensitivity, body composition, and overall energy homeostasis. Since the publication of the "thrifty phenotype hypothesis" by Hales and Barker (Diabetologia 1992;35:595-601), animal experiments have focused on establishing the mechanisms involved, which include changes in fetal cortisol, insulin, and leptin secretion or sensitivity. Intrauterine growth retardation can be induced by either prolonged modest changes in maternal diet or by more severe changes in uterine blood supply near to term. These contrasting challenges result in different amounts of cellular stress in the offspring. In addition, shifts in the transcriptional activity of DNA may produce sustained metabolic adaptations. Within tissues and organs that control metabolic homeostasis (eg, hypothalamus, adipose tissue, stomach, skeletal muscle, and heart), a range of phenotypes can be induced by sustained changes in maternal diet via modulation of genes that control DNA methylation and by histone acetylation, which suggests epigenetic programming. We now need to understand how changes in maternal diet affect DNA and how they are conserved on exposure to oxidative stress. A main challenge will be to establish how the dietary environment interacts with the programmed phenotype to trigger the development of metabolic disease. This may aid in the establishment of nutrigenomic strategies to prevent the metabolic syndrome.
Collapse
Affiliation(s)
- Sylvain Sebert
- Early Life Nutrition Research Unit, Academic Division of Child Health, and Nottingham Respiratory Medicine Biomedical Research Unit, School of Clinical Sciences, University Hospital Nottingham, Nottingham, United Kingdom
| | | | | | | |
Collapse
|
25
|
Koletzko B, Symonds ME, Olsen SF. Programming research: where are we and where do we go from here? Am J Clin Nutr 2011; 94:2036S-2043S. [PMID: 22089444 DOI: 10.3945/ajcn.111.018903] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Convincing evidence has accumulated to show that both pre- and postnatal nutrition preprogram long-term health, well-being, and performance until adulthood and old age. There is a very large potential in the application of this knowledge to promote public health. One of the prerequisites for translational application is to strengthen the scientific evidence. More extensive knowledge is needed (eg, on effect sizes of early life programming in contemporary populations, on specific nutritional exposures, on sensitive time periods in early life, on precise underlying mechanisms, and on potential effect differences in subgroups characterized by, eg, genetic predisposition or sex). Future programming research should aim at filling the existing gaps in scientific knowledge, consider the entire lifespan, address socioeconomic issues, and foster innovation. Research should aim at results suitable for translational application (eg, by leading to health-promoting policies and evidence-based dietary recommendations in the perinatal period). International collaboration and a close research partnership of academia, industry, and small and medium enterprises may strengthen research and innovative potential enhancing the likelihood of translational application. The scientific know-how and methodology available today allow us to take major steps forward in the near future; hence, research on nutritional programming deserves high priority.
Collapse
Affiliation(s)
- Berthold Koletzko
- Division of Metabolic and Nutritional Medicine, Department of Pediatrics, Dr von Hauner Children's Hospital, University of Munich Medical Center, Munich, Germany.
| | | | | | | | | |
Collapse
|
26
|
Abstract
Metabolic syndrome is reaching epidemic proportions, particularly in developing countries. In this review, we explore the concept-based on the developmental-origin-of-health-and-disease hypothesis-that reprogramming during critical times of fetal life can lead to metabolic syndrome in adulthood. Specifically, we summarize the epidemiological evidence linking prenatal stress, manifested by low birth weight, to metabolic syndrome and its individual components. We also review animal studies that suggest potential mechanisms for the long-term effects of fetal reprogramming, including the cellular response to stress and both organ- and hormone-specific alterations induced by stress. Although metabolic syndrome in adulthood is undoubtedly caused by multiple factors, including modifiable behavior, fetal life may provide a critical window in which individuals are predisposed to metabolic syndrome later in life.
Collapse
Affiliation(s)
- Paolo Rinaudo
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, California 94115, USA.
| | | |
Collapse
|
27
|
Cardel M, Dulin-Keita A, Casazza K. Contributors to Pediatric Obesity in Adolescence: More than just Energy Imbalance. ACTA ACUST UNITED AC 2011; 3:17-26. [PMID: 24563683 DOI: 10.2174/1876823701103010017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Disentangling the etiology of pediatric obesity continues to challenge researchers. Due to rapid growth and development, changes in the hormonal milieu, increased autonomy in feeding practices and greater interactions with environmental factors, adolescence is a particularly important period for the determination of body composition trajectories and the relationship to current and future obesity outcomes. A plethora of studies have focused on excess energy consumption and physical inactivity as they relate to weight and fat gain in adolescence. Although these "Big Two" have an impact, the increasing trends in pediatric obesity are not accounted for solely by increased energy intake and decreased physical activity. Indeed, under similar conditions of energy balance, inter-individual variation in fat accumulation has been consistently noted. It is becoming more evident that additional factors may contribute independently and/or synergistically to the increase in obesity. Such factors include (but are not limited to) metabolic programming in utero and in early childhood, the hormonal environment, endocrine disruptors, parental feeding practices, and the built environment. Our objective, therefore, is to investigate possible factors, particularly in adolescence that contributes to the increase in pediatric obesity beyond "The Big Two".
Collapse
Affiliation(s)
- Michelle Cardel
- Department of Nutrition Sciences and Clinical Nutrition Research Center, University of Alabama at Birmingham, Birmingham, AL 35294-3360, USA
| | - Akilah Dulin-Keita
- Department of Nutrition Sciences and Clinical Nutrition Research Center, University of Alabama at Birmingham, Birmingham, AL 35294-3360, USA
| | - Krista Casazza
- Department of Nutrition Sciences and Clinical Nutrition Research Center, University of Alabama at Birmingham, Birmingham, AL 35294-3360, USA
| |
Collapse
|
28
|
The conflicting effects of maternal nutrient restriction and early-life obesity on renal health. Proc Nutr Soc 2011; 70:268-75. [DOI: 10.1017/s0029665110004921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Epidemiological and animal studies have demonstrated that early-life nutrition alters the metabolic responses and generates structural changes in complex tissues, such as the kidneys, which may lead to a reduction in the offspring lifespan. Independently, obesity induces a spontaneous low-grade chronic inflammatory response by modulating several of the major metabolic pathways that ultimately compromise long-term renal health. However, the combined effects of maternal nutrition and early-life obesity in the development of renal diseases are far from conclusive. Previous results, using the ovine model, demonstrated that the combination of a reduction in fetal nutrition and juvenile obesity induced a series of adaptations associated with severe metabolic syndrome in the heart and adipose tissue. Surprisingly, exposure to an obesogenic environment in the kidney of those offspring produced an apparent reduction in glomerulosclerosis in relation to age- and weight-matched controls. However, this reduction in cellular apoptosis was accompanied by a rise in glomerular filtration rate and blood pressure of equal intensity when compared with obese controls. The intention of this review is to explain the adaptive responses observed in this model, based on insights into the mechanism of renal fetal programming, and their potential interactions with some of the metabolic changes produced by obesity.
Collapse
|
29
|
Hyatt MA, Gardner DS, Sebert S, Wilson V, Davidson N, Nigmatullina Y, Chan LLY, Budge H, Symonds ME. Suboptimal maternal nutrition, during early fetal liver development, promotes lipid accumulation in the liver of obese offspring. Reproduction 2010; 141:119-26. [PMID: 21045167 PMCID: PMC3001618 DOI: 10.1530/rep-10-0325] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Maternal nutrition during the period of early organ development can modulate the offspring's ability to metabolise excess fat as young adults when exposed to an obesogenic environment. This study examined the hypothesis that exposing offspring to nutrient restriction coincident with early hepatogenesis would result in endocrine and metabolic adaptations that subsequently lead to increased ectopic lipid accumulation within the liver. Pregnant sheep were fed either 50 or 100% of total metabolisable energy requirements from 30 to 80 days gestation and 100% thereafter. At weaning, offspring were made obese, and at ∼1 year of age livers were sampled. Lipid infiltration and molecular indices of gluconeogenesis, lipid metabolism and mitochondrial function were measured. Although hepatic triglyceride accumulation was not affected by obesity per se, it was nearly doubled in obese offspring born to nutrient-restricted mothers. This adaptation was accompanied by elevated gene expression for peroxisome proliferator-activated receptor γ (PPARG) and its co-activator PGC1α, which may be indicative of changes in the rate of hepatic fatty acid oxidation. In contrast, maternal diet had no influence on the stimulatory effect of obesity on gene expression for a range of proteins involved in glucose metabolism and energy balance including glucokinase, glucocorticoid receptors and uncoupling protein 2. Similarly, although gene expressions for the insulin and IGF1 receptors were suppressed by obesity they were not influenced by the prenatal nutritional environment. In conclusion, excess hepatic lipid accumulation with juvenile obesity is promoted by suboptimal nutrition coincident with early development of the fetal liver.
Collapse
Affiliation(s)
- M A Hyatt
- Early Life Nutrition Research Unit, Academic Child Health Respiratory Biomedical Research Unit, School of Clinical Sciences, Queen's Medical Centre, University Hospital, Nottingham, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Giraudo SQ, Della-Fera MA, Proctor L, Wickwire K, Ambati S, Baile CA. Maternal high fat feeding and gestational dietary restriction: effects on offspring body weight, food intake and hypothalamic gene expression over three generations in mice. Pharmacol Biochem Behav 2010; 97:121-9. [PMID: 20430050 DOI: 10.1016/j.pbb.2010.04.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 03/22/2010] [Accepted: 04/19/2010] [Indexed: 10/19/2022]
Abstract
Excessive gestational weight gain and maternal obesity have both been associated with increased incidence of obesity and metabolic disorder in offspring in both humans and animal models. The objectives of this study were to determine (1) whether mild gestational food restriction during the third trimester (GFR) would alter food intake and growth parameters of offspring, (2) whether effects of GFR depended on diet (high fat [HF] vs chow), (3) whether effects of excessive gestational weight gain (WG) would become magnified across generations, and (4) whether diet and GFR would alter hypothalamic gene expression in adult offspring. Three generations of female C57BL/6 mice were fed chow or HF diet, mated at 11 weeks of age and assigned to ad libitum feeding or 25% GFR. Offspring were fed the same diet as their mothers. Results showed (1) maternal gestational WG was positively correlated with offspring WG. (2) HF offspring weighed less (p<0.01) at weaning (WWT) but gained more during the 8 weeks after weaning than chow-fed offspring (p<0.05), resulting in higher final body weights (BW) (p<0.01). (3) HF males from GFR mothers had higher WWT (p<0.05), but subsequent WG and final BW were less (p<0.05) compared to males from ad lib mothers. (4) In the HF group, GFR also resulted in decreased FI (p<0.05) and FE (p<0.07) in offspring, compared to offspring from ad lib mothers. (5) In generation 3, hypothalamic expression of tyrosine hydroxylase was lower in HF males from GFR mothers compared to HF males from ad lib mothers (p<0.05). In conclusion, gender and maternal GFR had independent effects on growth and FI, and hypothalamic gene expression was dependent on both gender and maternal GFR in HF offspring. Even mild food restriction of obese mothers during pregnancy may have beneficial effects in reducing the risk or degree of obesity in offspring.
Collapse
Affiliation(s)
- Silvia Q Giraudo
- Department of Foods & Nutrition, University of Georgia, Athens, GA 30602-3622, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Hyatt MA, Keisler DH, Budge H, Symonds ME. Maternal parity and its effect on adipose tissue deposition and endocrine sensitivity in the postnatal sheep. J Endocrinol 2010; 204:173-9. [PMID: 19934248 PMCID: PMC2807923 DOI: 10.1677/joe-09-0358] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Maternal parity influences size at birth, postnatal growth and body composition with firstborn infants being more likely to be smaller with increased fat mass, suggesting that adiposity is set in early life. The precise effect of parity on fat mass and its endocrine sensitivity remains unclear and was, therefore, investigated in the present study. We utilised an established sheep model in which perirenal-abdominal fat mass (the major fat depot in the neonatal sheep) increases approximately 10-fold over the first month of life and focussed on the impact of parity on glucocorticoid sensitivity and adipokine expression in the adipocyte. Twin-bearing sheep of similar body weight and adiposity that consumed identical diets were utilised, and maternal blood samples were taken at 130 days of gestation. One offspring from each twin pair was sampled at 1 day of age, coincident with the time of maximal recruitment of uncoupling protein 1 (UCP1), whilst its sibling was sampled at 1 month, when UCP1 had disappeared. Plasma leptin was lower in nulliparous mothers than in multiparous mothers, and offspring of nulliparous mothers possessed more adipose tissue with increased mRNA abundance of leptin, glucocorticoid receptor and UCP2, adaptations that persisted up to 1 month of age when gene expression for interleukin-6 and adiponectin was also raised. The increase in fat mass associated with firstborn status is therefore accompanied by a resetting of the leptin and glucocorticoid axis within the adipocyte. Our findings emphasise the importance of parity in determining adipose tissue development and that firstborn offspring have an increased capacity for adipogenesis which may be critical in determining later adiposity.
Collapse
Affiliation(s)
- M A Hyatt
- Early Life Nutrition Research Unit, Academic Child Health, Division of Human DevelopmentSchool of Clinical Sciences, University HospitalE Floor East Block, Derby Road, Nottingham, NG7 2UHUK
| | - D H Keisler
- Department of Animal SciencesUniversity of MissouriColumbia, Missouri, 65201USA
| | - H Budge
- Early Life Nutrition Research Unit, Academic Child Health, Division of Human DevelopmentSchool of Clinical Sciences, University HospitalE Floor East Block, Derby Road, Nottingham, NG7 2UHUK
- Respiratory Biomedical Research UnitSchool of Clinical Sciences, University Hospital, University of NottinghamE Floor East Block, Derby Road, Nottingham, NG7 2UHUK
| | - M E Symonds
- Early Life Nutrition Research Unit, Academic Child Health, Division of Human DevelopmentSchool of Clinical Sciences, University HospitalE Floor East Block, Derby Road, Nottingham, NG7 2UHUK
- Respiratory Biomedical Research UnitSchool of Clinical Sciences, University Hospital, University of NottinghamE Floor East Block, Derby Road, Nottingham, NG7 2UHUK
- (Correspondence should be addressed to M E Symonds; )
| |
Collapse
|
32
|
Sébert SP, Hyatt MA, Chan LLY, Yiallourides M, Fainberg HP, Patel N, Sharkey D, Stephenson T, Rhind SM, Bell RC, Budge H, Gardner DS, Symonds ME. Influence of prenatal nutrition and obesity on tissue specific fat mass and obesity-associated (FTO) gene expression. Reproduction 2010; 139:265-74. [DOI: 10.1530/rep-09-0173] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The recent discovery of an association between body composition, energy intake and the fat mass and obesity-associated (FTO) gene represents a promising new therapeutic target in obesity prevention. In a well, pre-established large animal model, we investigated the regulation ofFTOgene expression under conditions either leading to obesity or increased risk of obesity related disorders: i) a sedentary ‘Western’ lifestyle and ii) prenatal exposure to nutrient restriction. Pregnant sheep were either fed to fully meet their nutritional requirements throughout gestation or 50% of this amount from early-to-mid gestation. Following weaning, offspring were either made obese through exposure to a sedentary obesogenic environment or remained lean. A significant positive relationship between placentalFTOgene expression and fetal weight was found at 110 days gestation. In both the newborn and adult offspring, the hypothalamus was the major site ofFTOgene expression. HypothalamicFTOgene expression was upregulated by obesity and was further increased by prenatal nutrient restriction. Importantly, we found a strong negative relationship between the hypothalamicFTOgene expression and food intake in lean animals only that may imply FTO as a novel controller of energy intake. In contrast,FTOgene expression in the heart was downregulated in obese offspring born to nutrient restricted mothers. In addition,FTOgene expression was unaffected by obesity or prenatal diet in insulin-dependent tissues, where it changed with age possibly reflecting adaptations in cellular energetic activity. These findings extend information gained from human epidemiology and provide new insights into the regulation ofin vivoenergy metabolism to prevent obesity.
Collapse
|
33
|
Abstract
The primary markers of the metabolic syndrome are central obesity, insulin resistance and hypertension. In this review, we consider the effect of changes in maternal nutrition during critical windows in fetal development on an individual's subsequent predisposition to the metabolic syndrome. The fetal origins of obesity, cardiovascular disease and insulin resistance have been investigated in a wide range of epidemiological and animal studies; these investigations highlight adaptations made by the nutritionally manipulated fetus that aim to maintain energy homeostasis to ensure survival. One consequence of such developmental plasticity may be a long term re-setting of cellular energy homeostasis, most probably via epigenetic modification of genes involved in a number of key regulatory pathways. For example, reduced maternal-fetal nutrition during early gestation to midgestation affects adipose tissue development and adiposity of the fetus by setting an increased number of adipocyte precursor cells. Importantly, clinically relevant adaptations to nutritional challenges in utero may only manifest as primary components of the metabolic syndrome if followed by a period of accelerated growth early in the postnatal period and/or if offspring become obese.
Collapse
Affiliation(s)
- Michael E Symonds
- Early Life Nutrition Research Unit, Academic Child Health, Division of Human Development, School of Clinical Sciences, University Hospital, Nottingham, UK.
| | | | | | | |
Collapse
|
34
|
Rhodes P, Craigon J, Gray C, Rhind SM, Loughna PT, Gardner DS. Adult-onset obesity reveals prenatal programming of glucose-insulin sensitivity in male sheep nutrient restricted during late gestation. PLoS One 2009; 4:e7393. [PMID: 19826474 PMCID: PMC2756957 DOI: 10.1371/journal.pone.0007393] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Accepted: 09/06/2009] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Obesity invokes a range of metabolic disturbances, but the transition from a poor to excessive nutritional environment may exacerbate adult metabolic dysfunction. The current study investigated global maternal nutrient restriction during early or late gestation on glucose tolerance and insulin sensitivity in the adult offspring when lean and obese. METHODS/PRINCIPAL FINDINGS Pregnant sheep received adequate (1.0M; CE, n = 6) or energy restricted (0.7M) diet during early (1-65 days; LEE, n = 6) or late (65-128 days; LEL, n = 7) gestation (term approximately 147 days). Subsequent offspring remained on pasture until 1.5 years when all received glucose and insulin tolerance tests (GTT & ITT) and body composition determination by dual energy x-ray absorptiometry (DXA). All animals were then exposed to an obesogenic environment for 6-7 months and all protocols repeated. Prenatal dietary treatment had no effect on birth weight or on metabolic endpoints when animals were 'lean' (1.5 years). Obesity revealed generalised metabolic 'inflexibility' and insulin resistance; characterised by blunted excursions of plasma NEFA and increased insulin(AUC) (from 133 to 341 [s.e.d. 26] ng.ml(-1).120 mins) during a GTT, respectively. For LEL vs. CE, the peak in plasma insulin when obese was greater (7.8 vs. 4.7 [s.e.d. 1.1] ng.ml(-1)) and was exacerbated by offspring sex (i.e. 9.8 vs. 4.4 [s.e.d. 1.16] ng.ml(-1); LEL male vs. CE male, respectively). Acquisition of obesity also significantly influenced the plasma lipid and protein profile to suggest, overall, greater net lipogenesis and reduced protein metabolism. CONCLUSIONS This study indicates generalised metabolic dysfunction with adult-onset obesity which also exacerbates and 'reveals' programming of glucose-insulin sensitivity in male offspring prenatally exposed to maternal undernutrition during late gestation. Taken together, the data suggest that metabolic function appears little compromised in young prenatally 'programmed' animals so long as weight is adequately controlled. Nutritional excess in adulthood exacerbates any programmed phenotype, indicating greater vigilance over weight control is required for those individuals exposed to nutritional thrift during gestation.
Collapse
Affiliation(s)
- Philip Rhodes
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, United Kingdom
| | - Jim Craigon
- School of Biosciences, University of Nottingham, Sutton Bonington, United Kingdom
| | - Clint Gray
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, United Kingdom
| | - Stuart M. Rhind
- Macaulay Land Use Research Institute, Craigiebuckler, Aberdeen, United Kingdom
| | - Paul T. Loughna
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, United Kingdom
| | - David S. Gardner
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, United Kingdom
| |
Collapse
|
35
|
Sharkey D, Gardner DS, Symonds ME, Budge H. Maternal nutrient restriction during early fetal kidney development attenuates the renal innate inflammatory response in obese young adult offspring. Am J Physiol Renal Physiol 2009; 297:F1199-207. [PMID: 19759269 DOI: 10.1152/ajprenal.00303.2009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Obesity is an independent risk factor for developing chronic kidney disease. Toll-like receptor 4 (TLR4), interleukin (IL)-18, and uncoupling protein 2 (UCP2) are important components of the innate immune system mediating inflammatory renal damage. Early to midgestation maternal nutrient restriction appears to protect the kidney from the deleterious effects of early onset obesity, although the mechanisms remain unclear. We examined the combined effects of gestational maternal nutrient restriction during early fetal kidney development and early onset obesity on the renal innate immune response in offspring. Pregnant sheep were randomly assigned to a normal (control, 100%) or nutrient-restricted (NR, 50%) diet from days 30 to 80 gestation and 100% thereafter. Offspring were killed humanely at 7 days or, following rearing in an obesogenic environment, at 1 yr of age, and renal tissues were collected. IL-18 and TLR4 expression were strongly correlated irrespective of intervention. Seven-day NR offspring had significantly lower relative renal mass and IL-18 mRNA expression. At 1 yr of age, obesity resulted in increased mRNA abundance of TLR4, IL-18, and UCP2, coupled with tubular atrophy and greater immunohistological staining of glomerular IL-6 and medullary tumor necrosis factor (TNF)-alpha. NR obese offspring had a marked reduction of TLR4 abundance and renal IL-6 staining. In conclusion, maternal nutrient restriction during early fetal kidney development attenuates the effects of early onset obesity-related nephropathy, in part, through the downregulation of the innate inflammatory response. A better understanding of maternal nutrition and the in utero nutritional environment may offer therapeutic strategies aimed at reducing the burden of later kidney disease.
Collapse
Affiliation(s)
- Don Sharkey
- Early Life Nutrition Research Group, Academic Child Health, University of Nottingham, Nottingham, United Kingdom
| | | | | | | |
Collapse
|
36
|
Abstract
The emerging role of adipose tissue as a dynamic endocrine organ with an extent of anatomical and physiological plasticity has generated numerous studies linking early-life events with long-term alterations in adipose tissue structure and function. Coupled with increasing rates of human obesity, which cannot be explained without some genetic component, the role of early programming of adipose tissue may provide an insight into potential mechanisms. The developmental origins of health and disease hypothesis investigates the potential association between a compromised fetal and postnatal environment and later disease, such as obesity and type 2 diabetes, in the offspring. A number of animal models have been developed to examine potential mechanisms that drive these physiological changes, including rodent and large-mammal models that provide mechanistic insights into the epidemiological findings. In utero challenges such as under- or over-provision of nutrients, placental insufficiency and glucocorticoid infusion, as well as postnatal nutritional challenges, can all result in the long-term programming of adipose tissue abundance and function. A range of hormones, enzymes, transcription factors and other metabolic signalling molecules have been implicated in adverse adipose tissue development, including leptin, glucocorticoids, members of the PPAR family, fatty acid-binding proteins and adipokines. The long-term structural and physiological consequences associated with these molecular and cellular changes are less well described. The experimental models, potential mechanisms and regulators of the early programming of adipose tissue in large mammalian species will be summarised in the present review.
Collapse
|
37
|
The impact of diet during early life and its contribution to later disease: critical checkpoints in development and their long-term consequences for metabolic health. Proc Nutr Soc 2009; 68:416-21. [DOI: 10.1017/s0029665109990152] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Changes in maternal diet at different stages of reproduction can have pronounced influences on the health and well-being of the resulting offspring, especially following exposure to an obesogenic environment. The mechanisms mediating adaptations in development of the embryo, placenta, fetus and newborn include changes in the maternal metabolic environment. These changes include reductions in a range of maternal counter-regulatory hormones such as cortisol, leptin and insulin. In the sheep, for example, targeted maternal nutrient restriction coincident with the period of maximal placental growth has pronounced effects on the development of the kidney and adipose tissue. As a consequence, the response of these tissues varies greatly following adolescent-onset obesity and ultimately results in these offspring exhibiting all the symptoms of the metabolic syndrome earlier in young adult life. Leptin administration to the offspring after birth can have some long-term differential effects, although much higher amounts are required to cause a response in small compared with large animal models. At the same time, the responsiveness of the offspring is gender dependent, which may relate to the differences in leptin sensitivity around the time of birth. Increasing maternal food intake during pregnancy, either globally or of individual nutrients, has little positive impact on birth weight but does impact on liver development. The challenge now is to establish which components of the maternal diet can be sustainably modified in order to optimise the maternal endocrine environment through pregnancy, thus ensuring feto–placental growth is appropriate in relation to an individual's gender and body composition.
Collapse
|
38
|
Sharkey D, Symonds ME, Budge H. Adipose tissue inflammation: developmental ontogeny and consequences of gestational nutrient restriction in offspring. Endocrinology 2009; 150:3913-20. [PMID: 19423760 DOI: 10.1210/en.2008-1784] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Increasing adiposity predisposes to the development of the metabolic syndrome, in part, through adipose tissue dysregulation and inflammation. In addition, offspring nutrient-restricted (NR) in utero can exhibit an increased risk of early-onset insulin resistance and obesity, although the mechanisms remain unclear. We aimed to: 1) define adipose tissue ontogeny of key proinflammatory and endoplasmic reticulum stress gene expression from late fetal to early adult life and 2) examine the impact on these genes in gestational nutrient restriction. Pregnant sheep were fed 100% (control) or 50% (NR) of their nutritional requirements between early to mid (28-80 d, term approximately 147 d) or late (110-147 d) gestation. In control offspring, toll-like receptor 4 (TLR4), and the macrophage marker CD68, peaked at 30 d of life before declining. IL-18 peaked at 6 months of age, whereas the endoplasmic reticulum chaperone glucose-regulated protein 78 peaked at birth and subsequently declined through postnatal life. TLR4 and CD68 positively correlated with relative adipose tissue mass and with each other. Early to midgestational NR offspring had decreased abundance of IL-18 at 6 months of age. In late gestational NR offspring, CD68 was significantly lower at birth, a pattern that reversed in juvenile offspring, coupled with increased TLR4 abundance. In conclusion, the in utero nutritional environment can alter the adipose tissue inflammatory profile in offspring. This may contribute to the increased risk of insulin resistance or obesity, dependent on the timing of nutrient restriction. Establishing the optimal maternal diet during pregnancy could reduce the burden of later adult disease in the offspring.
Collapse
Affiliation(s)
- Don Sharkey
- Centre for Reproduction and Early Life, Institute of Clinical Research, University of Nottingham NG7 2UH, United Kingdom
| | | | | |
Collapse
|
39
|
Yiallourides M, Sebert SP, Wilson V, Sharkey D, Rhind SM, Symonds ME, Budge H. The differential effects of the timing of maternal nutrient restriction in the ovine placenta on glucocorticoid sensitivity, uncoupling protein 2, peroxisome proliferator-activated receptor-gamma and cell proliferation. Reproduction 2009; 138:601-8. [PMID: 19525364 DOI: 10.1530/rep-09-0043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Nutrient restriction (NR) during critical windows of pregnancy has differential effects on placento-fetal growth and development. Our study, therefore, investigated developmental and metabolic adaptations within the ovine placenta following NR at different critical windows during the first 110 days of gestation (term=147 days). Thus, the effects of NR on cell proliferation, glucocorticoid sensitivity, IGF1 and 2 receptor, peroxisome proliferator-activated receptor gamma (PPARG), and uncoupling protein (UCP)2 gene expression in the placenta were examined. Singleton bearing sheep (n=4-8 per group) were fed either 100% of their total metabolizable energy requirements throughout the study or 50% of this amount between 0-30, 31-65, 66-110, and 0-110 days gestation. A significant reduction in cell proliferation and increased gene expression for the glucocorticoid and IGF2 receptors, PPARG, and UCP2 were detected in placentae sampled from mothers who were nutrient restricted between days 66 and 110 of gestation, only, relative to controls. This window of gestation coincides with the maximum placental growth and the start of exponential growth of the fetus when there are substantially increased metabolic demands on the placenta compared with earlier in gestation. Consequently, increased glucocorticoid sensitivity and suppressed IGF2 action could contribute to a switch in the placenta from proliferation to differentiation, thereby improving its nutrient transfer capacity. Upregulation of PPARG and UCP2 would promote placental fatty acid metabolism thereby limiting glucose utilization. These compensatory placental responses may serve to maintain fetal growth but could result in adverse adaptations such as the early onset of the metabolic syndrome in later life.
Collapse
Affiliation(s)
- M Yiallourides
- Early Life Nutrition Research Unit, Academic Child Health, School of Clinical Sciences, University Hospital, Nottingham NG72UH, UK
| | | | | | | | | | | | | |
Collapse
|
40
|
Session on ‘Obesity’ Adipose tissue development, nutrition in early life and its impact on later obesity. Proc Nutr Soc 2009; 68:321-6. [DOI: 10.1017/s0029665109001402] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
It is now apparent that one key factor determining the current obesity epidemic within the developed world is the extent to which adipose tissue growth and function can be reset in early life. Adipose tissue can be either brown or white, with brown fat being characterised as possessing a unique uncoupling protein (uncoupling protein 1) that enables the rapid generation of heat by non-shivering thermogenesis. In large mammals this function is recruited at approximately the time of birth, after which brown fat is lost, not normally reappearing again throughout the life cycle. The origin and developmental regulation of brown fat in large mammals is therefore very different from that of small mammals in which brown fat is retained throughout the life cycle and may have the same origin as muscle cells. In contrast, white adipose tissue increases in mass after birth, paralleled by a rise in glucocorticoid action and macrophage accumulation. This process can be reset by changes in the maternal nutritional environment, with the magnitude of response being further determined by the timing at which such a challenge is imposed. Importantly, the long-term response within white adipocytes can occur in the absence of any change in total fat mass. The present review therefore emphasises the need to further understand the developmental regulation of the function of fat through the life cycle in order to optimise appropriate and sustainable intervention strategies necessary not only to prevent obesity in the first place but also to reverse excess fat mass in obese individuals.
Collapse
|
41
|
Chan LLY, Sébert SP, Hyatt MA, Stephenson T, Budge H, Symonds ME, Gardner DS. Effect of maternal nutrient restriction from early to midgestation on cardiac function and metabolism after adolescent-onset obesity. Am J Physiol Regul Integr Comp Physiol 2009; 296:R1455-63. [PMID: 19244582 DOI: 10.1152/ajpregu.91019.2008] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Maternal nutrient restriction (NR) from early to midgestation has marked effects on endocrine sensitivity and organ function of the resulting offspring. We hypothesized that early NR may reset the expression profile of genes central to myocardial energy metabolism, influencing ectopic lipid deposition and cardiac function in the obese adult offspring. NR offspring were exposed to an "obesogenic" environment, and their cardiac function and molecular indexes of myocardial energy metabolism were assessed to explore the hypothesis that an obese individual's risk of heart disease may be modified after maternal NR. Pregnant sheep were fed 100% (control) or 50% (NR) energy requirement from days 30 to 80 of gestation and 100% energy requirement thereafter. At weaning, offspring were exposed to an obesogenic environment or remained lean. At approximately 1 yr of age, the hemodynamic response of these offspring to hypotension, together with left ventricular expression profiles of fatty acid-binding protein 3 (FABP3), peroxisome proliferator-activated receptor-gamma (PPARgamma) and its coactivator (PGC)-1alpha, acetyl-CoA carboxylase (ACC), AMP-activated protein kinase (AMPK)-alpha(2), and voltage-dependent anion channel 1 (VDAC1), was determined. Obesity produced left ventricular hypertrophy in all animals, with increased ectopic (myocardial) lipid in NR offspring. Obesity per se significantly reduced myocardial transcript expression of PGC-1alpha, AMPKalpha(2), VDAC1, and ACC and increased expression of PPARgamma and FABP3. However, although NR animals were similarly obese, their transcript expression of ACC, PPARgamma, and FABP3 was similar to that of lean animals, indicating altered cardiac energy metabolism. Indeed, blunted tachycardia and an amplified inotropic response to hypotension characterized cardiac function in obese NR offspring. The results suggest that maternal NR during early organogenesis can precipitate an altered myocardial response to hypotension and increased myocardial lipid deposition in the adult offspring after adolescent-onset obesity, potentially rendering these individuals more at risk of early heart failure as they age.
Collapse
Affiliation(s)
- L L Y Chan
- School of Veterinary Medicine & Science, University of Nottingham, Sutton Bonington, Leicestershire LE12 5RD, UK
| | | | | | | | | | | | | |
Collapse
|