151
|
Song A, Astbury S, Hoedl A, Nielsen B, Symonds ME, Bell RC. Lifetime Exposure to a Constant Environment Amplifies the Impact of a Fructose-Rich Diet on Glucose Homeostasis during Pregnancy. Nutrients 2017; 9:nu9040327. [PMID: 28346343 PMCID: PMC5409666 DOI: 10.3390/nu9040327] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/15/2017] [Accepted: 03/21/2017] [Indexed: 12/21/2022] Open
Abstract
The need to refine rodent models of human-related disease is now being recognized, in particular the rearing environment that can profoundly modulate metabolic regulation. Most studies on pregnancy and fetal development purchase and transport young females into the research facility, which after a short period of acclimation are investigated (Gen0). We demonstrate that female offspring (Gen1) show an exaggerated hyperinsulinemic response to pregnancy when fed a standard diet and with high fructose intake, which continues throughout pregnancy. Markers of maternal hepatic metabolism were differentially influenced, as the gene expression of acetyl-CoA-carboxylase was raised in Gen1 given fructose and controls, whereas glucose transporter 5 and fatty acid synthase expression were only raised with fructose. Gen1 rats weighed more than Gen0 throughout the study, although fructose feeding raised the percent body fat but not body weight. We show that long-term habituation to the living environment has a profound impact on the animal’s metabolic responses to nutritional intervention and pregnancy. This has important implications for interpreting many studies investigating the influence of maternal consumption of fructose on pregnancy outcomes and offspring to date.
Collapse
Affiliation(s)
- Aleida Song
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| | - Stuart Astbury
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada.
- Early Life Research Group, Academic Division of Child Health, Obstetrics & Gynaecology, and NIHR Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham NG7 2UH, UK.
| | - Abha Hoedl
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| | - Brent Nielsen
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| | - Michael E Symonds
- Early Life Research Group, Academic Division of Child Health, Obstetrics & Gynaecology, and NIHR Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham NG7 2UH, UK.
| | - Rhonda C Bell
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada.
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| |
Collapse
|
152
|
Sales VM, Ferguson-Smith AC, Patti ME. Epigenetic Mechanisms of Transmission of Metabolic Disease across Generations. Cell Metab 2017; 25:559-571. [PMID: 28273478 PMCID: PMC5404272 DOI: 10.1016/j.cmet.2017.02.016] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Both human and animal studies indicate that environmental exposures experienced during early life can robustly influence risk for adult disease. Moreover, environmental exposures experienced by parents during either intrauterine or postnatal life can also influence the health of their offspring, thus initiating a cycle of disease risk across generations. In this Perspective, we focus on epigenetic mechanisms in germ cells, including DNA methylation, histone modification, and non-coding RNAs, which collectively may provide a non-genetic molecular legacy of prior environmental exposures and influence transcriptional regulation, developmental trajectories, and adult disease risk in offspring.
Collapse
Affiliation(s)
- Vicencia Micheline Sales
- Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center and Harvard Medical School, One Joslin Place, Sixth Floor, Boston, MA 02215, USA
| | - Anne C Ferguson-Smith
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| | - Mary-Elizabeth Patti
- Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center and Harvard Medical School, One Joslin Place, Sixth Floor, Boston, MA 02215, USA.
| |
Collapse
|
153
|
de Paula Simino LA, de Fante T, Figueiredo Fontana M, Oliveira Borges F, Torsoni MA, Milanski M, Velloso LA, Souza Torsoni A. Lipid overload during gestation and lactation can independently alter lipid homeostasis in offspring and promote metabolic impairment after new challenge to high-fat diet. Nutr Metab (Lond) 2017; 14:16. [PMID: 28239403 PMCID: PMC5319047 DOI: 10.1186/s12986-017-0168-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/06/2017] [Indexed: 02/06/2023] Open
Abstract
Background Nutritional status in early life is critically involved in the metabolic phenotype of offspring. However the changes triggered by maternal consumption of high-fat diet (HFD) in pre- or postnatal period should be better understood. Here we evaluated whether maternal HFD consumption during gestation and lactation could differently affect liver miR-122 and miR-370 expression leading to metabolic damages observed in offspring. Moreover, we investigate whether early overnutrition program offspring to more harmful response to HFD in later life. Methods Female mice were fed either a standard chow (SC) diet or a HFD three weeks before and during mating, gestation and/or lactation. Offspring were evaluated on the delivery day (d0), in a cross-fostering model at day 28 (d28) and in adult life, after a re-challenge with a HFD (d82). Results In vitro analysis using liver cell line showed that palmitate could induced decrease in miR-122 and increase in miR-370 expression. Newborn pups (d0) from obese dams showed a decrease in lipid oxidation markers (Cpt1a and Acadvl), an increase in triacylglycerol synthesis markers (Agpat and Gpam), as well as lower miR-122 and higher miR-370 hepatic content that was inversely correlated to maternal serum NEFA and TAG. Pups fostered to SC dams presented an increase in body weight and Agpat/Gpam expression at d28 compared to pups fostered to HFD dams and an inverse correlation was observed between miR-122 hepatic expression and offspring serum TAG. In adult life (d82), the reintroduction of HFD resulted in higher body weight gain and hepatic lipid content. These effects were accompanied by impairment in lipid and glucose metabolism, demonstrated by reduced Cpt1a/Acadvl and increased Agpat/Gpam expression, lower glucose tolerance and insulin sensitivity. Conclusion Our data suggest that both gestational and lactation overnutrition results in metabolic changes that can permanently alter lipid homeostasis in offspring. The presence of fatty acids in maternal blood and milk seem to be responsible for modulating the expression of miR-122 and miR-370, which are involved in liver metabolism. These alterations significantly increase susceptibility to obesity and ectopic lipid accumulation and lead to a more harmful response to HFD in offspring. Electronic supplementary material The online version of this article (doi:10.1186/s12986-017-0168-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laís Angélica de Paula Simino
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Thaís de Fante
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Marina Figueiredo Fontana
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Fernanda Oliveira Borges
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Márcio Alberto Torsoni
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Marciane Milanski
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Lício Augusto Velloso
- Laboratory of Cell Signaling, Faculty of Medical Sciences, University Of Campinas - UNICAMP, Campinas, São Paulo Brazil
| | - Adriana Souza Torsoni
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| |
Collapse
|
154
|
Wesolowski SR, El Kasmi KC, Jonscher KR, Friedman JE. Developmental origins of NAFLD: a womb with a clue. Nat Rev Gastroenterol Hepatol 2017; 14:81-96. [PMID: 27780972 PMCID: PMC5725959 DOI: 10.1038/nrgastro.2016.160] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Changes in the maternal environment leading to an altered intrauterine milieu can result in subtle insults to the fetus, promoting increased lifetime disease risk and/or disease acceleration in childhood and later in life. Particularly worrisome is that the prevalence of NAFLD is rapidly increasing among children and adults, and is being diagnosed at increasingly younger ages, pointing towards an early-life origin. A wealth of evidence, in humans and non-human primates, suggests that maternal nutrition affects the placenta and fetal tissues, leading to persistent changes in hepatic metabolism, mitochondrial function, the intestinal microbiota, liver macrophage activation and susceptibility to NASH postnatally. Deleterious exposures in utero include fetal hypoxia, increased nutrient supply, inflammation and altered gut microbiota that might produce metabolic clues, including fatty acids, metabolites, endotoxins, bile acids and cytokines, which prime the infant liver for NAFLD in a persistent manner and increase susceptibility to NASH. Mechanistic links to early disease pathways might involve shifts in lipid metabolism, mitochondrial dysfunction, pioneering gut microorganisms, macrophage programming and epigenetic changes that alter the liver microenvironment, favouring liver injury. In this Review, we discuss how maternal, fetal, neonatal and infant exposures provide developmental clues and mechanisms to help explain NAFLD acceleration and increased disease prevalence. Mechanisms identified in clinical and preclinical models suggest important opportunities for prevention and intervention that could slow down the growing epidemic of NAFLD in the next generation.
Collapse
Affiliation(s)
| | - Karim C. El Kasmi
- Department of Pediatrics, Section of Gastroenterology, Hepatology and Nutrition, University of Colorado
| | | | - Jacob E. Friedman
- Department of Pediatrics, Section of Neonatology, University of Colorado,Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado, Anschutz Medical Campus, 12801 East 17th Avenue, MS 8106, Aurora, Colorado 80045, USA
| |
Collapse
|
155
|
de Paula Simino LA, de Fante T, Figueiredo Fontana M, Oliveira Borges F, Torsoni MA, Milanski M, Velloso LA, Souza Torsoni A. Lipid overload during gestation and lactation can independently alter lipid homeostasis in offspring and promote metabolic impairment after new challenge to high-fat diet. Nutr Metab (Lond) 2017. [PMID: 28239403 DOI: 10.1186/sl2986-017-0168-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Nutritional status in early life is critically involved in the metabolic phenotype of offspring. However the changes triggered by maternal consumption of high-fat diet (HFD) in pre- or postnatal period should be better understood. Here we evaluated whether maternal HFD consumption during gestation and lactation could differently affect liver miR-122 and miR-370 expression leading to metabolic damages observed in offspring. Moreover, we investigate whether early overnutrition program offspring to more harmful response to HFD in later life. METHODS Female mice were fed either a standard chow (SC) diet or a HFD three weeks before and during mating, gestation and/or lactation. Offspring were evaluated on the delivery day (d0), in a cross-fostering model at day 28 (d28) and in adult life, after a re-challenge with a HFD (d82). RESULTS In vitro analysis using liver cell line showed that palmitate could induced decrease in miR-122 and increase in miR-370 expression. Newborn pups (d0) from obese dams showed a decrease in lipid oxidation markers (Cpt1a and Acadvl), an increase in triacylglycerol synthesis markers (Agpat and Gpam), as well as lower miR-122 and higher miR-370 hepatic content that was inversely correlated to maternal serum NEFA and TAG. Pups fostered to SC dams presented an increase in body weight and Agpat/Gpam expression at d28 compared to pups fostered to HFD dams and an inverse correlation was observed between miR-122 hepatic expression and offspring serum TAG. In adult life (d82), the reintroduction of HFD resulted in higher body weight gain and hepatic lipid content. These effects were accompanied by impairment in lipid and glucose metabolism, demonstrated by reduced Cpt1a/Acadvl and increased Agpat/Gpam expression, lower glucose tolerance and insulin sensitivity. CONCLUSION Our data suggest that both gestational and lactation overnutrition results in metabolic changes that can permanently alter lipid homeostasis in offspring. The presence of fatty acids in maternal blood and milk seem to be responsible for modulating the expression of miR-122 and miR-370, which are involved in liver metabolism. These alterations significantly increase susceptibility to obesity and ectopic lipid accumulation and lead to a more harmful response to HFD in offspring.
Collapse
Affiliation(s)
- Laís Angélica de Paula Simino
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Thaís de Fante
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Marina Figueiredo Fontana
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Fernanda Oliveira Borges
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Márcio Alberto Torsoni
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Marciane Milanski
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Lício Augusto Velloso
- Laboratory of Cell Signaling, Faculty of Medical Sciences, University Of Campinas - UNICAMP, Campinas, São Paulo Brazil
| | - Adriana Souza Torsoni
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| |
Collapse
|
156
|
Reynolds CM, Segovia SA, Vickers MH. Experimental Models of Maternal Obesity and Neuroendocrine Programming of Metabolic Disorders in Offspring. Front Endocrinol (Lausanne) 2017; 8:245. [PMID: 28993758 PMCID: PMC5622157 DOI: 10.3389/fendo.2017.00245] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 09/06/2017] [Indexed: 12/17/2022] Open
Abstract
Evidence from epidemiological, clinical, and experimental studies have clearly shown that disease risk in later life is increased following a poor early life environment, a process preferentially termed developmental programming. In particular, this work clearly highlights the importance of the nutritional environment during early development with alterations in maternal nutrition, including both under- and overnutrition, increasing the risk for a range of cardiometabolic and neurobehavioral disorders in adult offspring characterized by both adipokine resistance and obesity. Although the mechanistic basis for such developmental programming is not yet fully defined, a common feature derived from experimental animal models is that of alterations in the wiring of the neuroendocrine pathways that control energy balance and appetite regulation during early stages of developmental plasticity. The adipokine leptin has also received significant attention with clear experimental evidence that normal regulation of leptin levels during the early life period is critical for the normal development of tissues and related signaling pathways that are involved in metabolic and cardiovascular homeostasis. There is also increasing evidence that alterations in the epigenome and other underlying mechanisms including an altered gut-brain axis may contribute to lasting cardiometabolic dysfunction in offspring. Ongoing studies that further define the mechanisms between these associations will allow for identification of early risk markers and implementation of strategies around interventions that will have obvious beneficial implications in breaking a programmed transgenerational cycle of metabolic disorders.
Collapse
Affiliation(s)
| | | | - Mark H. Vickers
- Liggins Institute, University of Auckland, Auckland, New Zealand
- *Correspondence: Mark H. Vickers,
| |
Collapse
|
157
|
Thompson JR, Valleau JC, Barling AN, Franco JG, DeCapo M, Bagley JL, Sullivan EL. Exposure to a High-Fat Diet during Early Development Programs Behavior and Impairs the Central Serotonergic System in Juvenile Non-Human Primates. Front Endocrinol (Lausanne) 2017; 8:164. [PMID: 28785241 PMCID: PMC5519527 DOI: 10.3389/fendo.2017.00164] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 06/27/2017] [Indexed: 12/29/2022] Open
Abstract
Perinatal exposure to maternal obesity and high-fat diet (HFD) consumption not only poses metabolic risks to offspring but also impacts brain development and mental health. Using a non-human primate model, we observed a persistent increase in anxiety in juvenile offspring exposed to a maternal HFD. Postweaning HFD consumption also increased anxiety and independently increased stereotypic behaviors. These behavioral changes were associated with modified cortisol stress response and impairments in the development of the central serotonin synthesis, with altered tryptophan hydroxylase-2 mRNA expression in the dorsal and median raphe. Postweaning HFD consumption decreased serotonergic immunoreactivity in area 10 of the prefrontal cortex. These results suggest that perinatal exposure to HFD consumption programs development of the brain and endocrine system, leading to behavioral impairments associated with mental health and neurodevelopmental disorders. Also, an early nutritional intervention (consumption of the control diet at weaning) was not sufficient to ameliorate many of the behavioral changes, such as increased anxiety, that were induced by maternal HFD consumption. Given the level of dietary fat consumption and maternal obesity in developed nations these findings have important implications for the mental health of future generations.
Collapse
Affiliation(s)
- Jacqueline R. Thompson
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Jeanette C. Valleau
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Ashley N. Barling
- Department of Biology, University of Portland, Portland, OR, United States
| | - Juliana G. Franco
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Madison DeCapo
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Jennifer L. Bagley
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Elinor L. Sullivan
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR, United States
- Department of Biology, University of Portland, Portland, OR, United States
- *Correspondence: Elinor L. Sullivan,
| |
Collapse
|
158
|
Jonscher KR, Stewart MS, Alfonso-Garcia A, DeFelice BC, Wang XX, Luo Y, Levi M, Heerwagen MJR, Janssen RC, de la Houssaye BA, Wiitala E, Florey G, Jonscher RL, Potma EO, Fiehn O, Friedman JE. Early PQQ supplementation has persistent long-term protective effects on developmental programming of hepatic lipotoxicity and inflammation in obese mice. FASEB J 2016; 31:1434-1448. [PMID: 28007783 DOI: 10.1096/fj.201600906r] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 12/12/2016] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is widespread in adults and children. Early exposure to maternal obesity or Western-style diet (WD) increases steatosis and oxidative stress in fetal liver and is associated with lifetime disease risk in the offspring. Pyrroloquinoline quinone (PQQ) is a natural antioxidant found in soil, enriched in human breast milk, and essential for development in mammals. We investigated whether a supplemental dose of PQQ, provided prenatally in a mouse model of diet-induced obesity during pregnancy, could protect obese offspring from progression of NAFLD. PQQ treatment given pre- and postnatally in WD-fed offspring had no effect on weight gain but increased metabolic flexibility while reducing body fat and liver lipids, compared with untreated obese offspring. Indices of NAFLD, including hepatic ceramide levels, oxidative stress, and expression of proinflammatory genes (Nos2, Nlrp3, Il6, and Ptgs2), were decreased in WD PQQ-fed mice, concomitant with increased expression of fatty acid oxidation genes and decreased Pparg expression. Notably, these changes persisted even after PQQ withdrawal at weaning. Our results suggest that supplementation with PQQ, particularly during pregnancy and lactation, protects offspring from WD-induced developmental programming of hepatic lipotoxicity and may help slow the advancing epidemic of NAFLD in the next generation.-Jonscher, K. R., Stewart, M. S., Alfonso-Garcia, A., DeFelice, B. C., Wang, X. X., Luo, Y., Levi, M., Heerwagen, M. J. R., Janssen, R. C., de la Houssaye, B. A., Wiitala, E., Florey, G., Jonscher, R. L., Potma, E. O., Fiehn, O. Friedman, J. E. Early PQQ supplementation has persistent long-term protective effects on developmental programming of hepatic lipotoxicity and inflammation in obese mice.
Collapse
Affiliation(s)
- Karen R Jonscher
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA;
| | - Michael S Stewart
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| | | | - Brian C DeFelice
- West Coast Metabolomics Center, University of California, Davis, Davis, CA USA
| | - Xiaoxin X Wang
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Yuhuan Luo
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Moshe Levi
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Margaret J R Heerwagen
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| | - Rachel C Janssen
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| | - Becky A de la Houssaye
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| | - Ellen Wiitala
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| | - Garrett Florey
- Department of Integrative Biology, University of Colorado, Denver, Denver, Colorado, USA; and
| | - Raleigh L Jonscher
- Department of Integrative Biology, University of Colorado, Denver, Denver, Colorado, USA; and
| | - Eric O Potma
- Beckman Laser Institute, and.,Department of Biomedical Engineering,University of California, Irvine, Irvine, California, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, Davis, CA USA.,Biochemistry Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jacob E Friedman
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| |
Collapse
|
159
|
Abstract
Type 2 diabetes is a typical multifactorial disease, but the causes can largely be divided into genetic and environmental factors. In recent years, focus has shifted to the interaction between these factors (i.e., gene-environment interactions). It has become widely known that changes in the intrauterine environment such as intrauterine growth restriction result in gene expression changes in various tissues, which ultimately lead to the onset of diabetes. Epigenetic modification is considered to be a particularly important mechanism in these effects, as it is easily affected by environmental changes that occur during the fetal and neonatal periods. Moreover, recent reports have revealed that epigenetic modifications are passed down through generations. Although genome-wide association studies have identified many type 2 diabetes susceptibility genes, these genes do not pose a significantly high risk when isolated as single factors. In particular, it has been suggested that the interaction of the FTO or KCNQ1 genes with environmental factors increases the incidence of diabetes. These findings suggest that detailed analyses of individual gene-environment interactions hold promise for gaining new insight into the mechanisms and risk factors contributing to type 2 diabetes, with application to personalized diagnoses and treatments. We look forward to future developments in this regard.
Collapse
Affiliation(s)
- Yoshiaki Kido
- 1Division of Metabolism and Disease, Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan.,2Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
160
|
Indicators of fetal growth and adult liver enzymes: the Bogalusa Heart Study and the Cardiovascular Risk in Young Finns Study. J Dev Orig Health Dis 2016; 8:226-235. [PMID: 27919310 DOI: 10.1017/s2040174416000635] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Despite the interest in the relationship of fetal exposures to adult cardiovascular disease, few studies have examined indicators of adult fatty liver disease as an outcome. Previous results are inconsistent, and indicate possible variation by sex. Adult liver enzymes [γ-glutamyl transferase (GGT), alanine transaminase (ALT) and aspartase transaminase (AST)] were measured in two cohort studies: the Bogalusa Heart Study (BHS; n=1803) and the Cardiovascular Risk in Young Finns (YF; n=3571) study, which also had ultrasound measures of liver fat (n=2546). Predictors of dichotomized (clinical cut-offs) and continuous (within the reference range) liver enzymes included low birthweight (4000 g), small-for-gestational-age (birthweight 90th percentile), and preterm birth. Multiple logistic and linear regression were conducted, adjusted for medical, behavioral and socioeconomic indicators. Interactions with sex were also examined. In BHS, birth measures were not strongly associated with clinically high levels of liver enzymes, and within the reference range measures of reduced growth were associated with increased AST in women. In the YF study, at least one marker of reduced growth was associated with higher GGT, higher ALT and higher AST (in women). Probable fatty liver on ultrasound was associated with low birthweight (2.41, 1.42-4.09) and preterm birth (2.84, 1.70-4.76). These results suggest a link between birth parameters and adult fatty liver, but encourage consideration of population variation in these relationships.
Collapse
|
161
|
Simpson J, Smith AD, Fraser A, Sattar N, Callaway M, Lindsay RS, Lawlor DA, Nelson SM. Cord Blood Adipokines and Lipids and Adolescent Nonalcoholic Fatty Liver Disease. J Clin Endocrinol Metab 2016; 101:4661-4668. [PMID: 27648968 PMCID: PMC5155695 DOI: 10.1210/jc.2016-2604] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/15/2016] [Indexed: 12/14/2022]
Abstract
CONTEXT Maternal adiposity in pregnancy is associated with offspring adiposity and metabolic dysfunction postnatally, including greater risk of nonalcoholic fatty liver disease (NAFLD). Recent genetic analyses suggest a causal effect of greater maternal body mass index on offspring birth weight and ponderal index, but the relative roles of the environment in utero or later in life remains unclear. OBJECTIVE We sought to determine whether markers of infant adiposity (birth weight, umbilical cord blood leptin, adiponectin, and lipids) were associated with markers of NAFLD in adolescence. DESIGN, SETTING, AND PARTICIPANTS This was a UK prospective birth cohort with 17 years of follow-up with liver function tests (aspartate aminotransferase, alanine aminotransferase, gamma-glutamyltransferase) (n = 1037 participants), and ultrasound scan assessed liver fat, volume, and sheer velocity at age 17 (n = 541 participants). Missing covariate data were imputed. MAIN OUTCOMES Ultrasound and biochemical measures of NAFLD were measured. RESULTS Birth weight, cord blood leptin, and adiponectin were not associated with a diagnosis of NAFLD. In adjusted analyses, 2 of 42 associations attained conventional 5% levels of significance. Birth weight was positively associated with liver volume (1.0% greater per 100 g [95% confidence interval 0.5%-2.0%]). Cord high-density lipoprotein-cholesterol was positively associated with alanine aminotransferase (11.6% higher per 1 mmol/L [95% confidence interval 0.3, 23.4]); however, this association was primarily mediated via offspring adiposity. CONCLUSIONS In this extensive analysis, we found little evidence measurements of infant fat mass and birth size were related to adolescent markers of NAFLD. The association between birth weight and adolescent liver volume may indicate the contribution of greater organ size to birth weight and tracking of organ size.
Collapse
Affiliation(s)
- Joy Simpson
- School of Medicine (J.S., S.M.N.), University of Glasgow, Glasgow G31 2ER, United Kingdom; Medical Research Council Integrative Epidemiology Unit (A.D.S., A.F., D.A.L.), School of Social and Community Medicine (A.D.S., A.F., D.A.L.), University of Bristol, Bristol BS13NY United Kingdom; Institute of Cardiovascular and Metabolic Medicine (N.S., R.S.L.), British Heart Foundation Glasgow Cardiovascular Research Centre, Glasgow G61 1QH, United Kingdom; and University Hospitals Bristol National Health Service Foundation Trust (M.C.), Bristol BS1 3NY, United Kingdom
| | - Andrew D Smith
- School of Medicine (J.S., S.M.N.), University of Glasgow, Glasgow G31 2ER, United Kingdom; Medical Research Council Integrative Epidemiology Unit (A.D.S., A.F., D.A.L.), School of Social and Community Medicine (A.D.S., A.F., D.A.L.), University of Bristol, Bristol BS13NY United Kingdom; Institute of Cardiovascular and Metabolic Medicine (N.S., R.S.L.), British Heart Foundation Glasgow Cardiovascular Research Centre, Glasgow G61 1QH, United Kingdom; and University Hospitals Bristol National Health Service Foundation Trust (M.C.), Bristol BS1 3NY, United Kingdom
| | - Abigail Fraser
- School of Medicine (J.S., S.M.N.), University of Glasgow, Glasgow G31 2ER, United Kingdom; Medical Research Council Integrative Epidemiology Unit (A.D.S., A.F., D.A.L.), School of Social and Community Medicine (A.D.S., A.F., D.A.L.), University of Bristol, Bristol BS13NY United Kingdom; Institute of Cardiovascular and Metabolic Medicine (N.S., R.S.L.), British Heart Foundation Glasgow Cardiovascular Research Centre, Glasgow G61 1QH, United Kingdom; and University Hospitals Bristol National Health Service Foundation Trust (M.C.), Bristol BS1 3NY, United Kingdom
| | - Naveed Sattar
- School of Medicine (J.S., S.M.N.), University of Glasgow, Glasgow G31 2ER, United Kingdom; Medical Research Council Integrative Epidemiology Unit (A.D.S., A.F., D.A.L.), School of Social and Community Medicine (A.D.S., A.F., D.A.L.), University of Bristol, Bristol BS13NY United Kingdom; Institute of Cardiovascular and Metabolic Medicine (N.S., R.S.L.), British Heart Foundation Glasgow Cardiovascular Research Centre, Glasgow G61 1QH, United Kingdom; and University Hospitals Bristol National Health Service Foundation Trust (M.C.), Bristol BS1 3NY, United Kingdom
| | - Mark Callaway
- School of Medicine (J.S., S.M.N.), University of Glasgow, Glasgow G31 2ER, United Kingdom; Medical Research Council Integrative Epidemiology Unit (A.D.S., A.F., D.A.L.), School of Social and Community Medicine (A.D.S., A.F., D.A.L.), University of Bristol, Bristol BS13NY United Kingdom; Institute of Cardiovascular and Metabolic Medicine (N.S., R.S.L.), British Heart Foundation Glasgow Cardiovascular Research Centre, Glasgow G61 1QH, United Kingdom; and University Hospitals Bristol National Health Service Foundation Trust (M.C.), Bristol BS1 3NY, United Kingdom
| | - Robert S Lindsay
- School of Medicine (J.S., S.M.N.), University of Glasgow, Glasgow G31 2ER, United Kingdom; Medical Research Council Integrative Epidemiology Unit (A.D.S., A.F., D.A.L.), School of Social and Community Medicine (A.D.S., A.F., D.A.L.), University of Bristol, Bristol BS13NY United Kingdom; Institute of Cardiovascular and Metabolic Medicine (N.S., R.S.L.), British Heart Foundation Glasgow Cardiovascular Research Centre, Glasgow G61 1QH, United Kingdom; and University Hospitals Bristol National Health Service Foundation Trust (M.C.), Bristol BS1 3NY, United Kingdom
| | - Debbie A Lawlor
- School of Medicine (J.S., S.M.N.), University of Glasgow, Glasgow G31 2ER, United Kingdom; Medical Research Council Integrative Epidemiology Unit (A.D.S., A.F., D.A.L.), School of Social and Community Medicine (A.D.S., A.F., D.A.L.), University of Bristol, Bristol BS13NY United Kingdom; Institute of Cardiovascular and Metabolic Medicine (N.S., R.S.L.), British Heart Foundation Glasgow Cardiovascular Research Centre, Glasgow G61 1QH, United Kingdom; and University Hospitals Bristol National Health Service Foundation Trust (M.C.), Bristol BS1 3NY, United Kingdom
| | - Scott M Nelson
- School of Medicine (J.S., S.M.N.), University of Glasgow, Glasgow G31 2ER, United Kingdom; Medical Research Council Integrative Epidemiology Unit (A.D.S., A.F., D.A.L.), School of Social and Community Medicine (A.D.S., A.F., D.A.L.), University of Bristol, Bristol BS13NY United Kingdom; Institute of Cardiovascular and Metabolic Medicine (N.S., R.S.L.), British Heart Foundation Glasgow Cardiovascular Research Centre, Glasgow G61 1QH, United Kingdom; and University Hospitals Bristol National Health Service Foundation Trust (M.C.), Bristol BS1 3NY, United Kingdom
| |
Collapse
|
162
|
Wesolowski SR, Hay WW. Role of placental insufficiency and intrauterine growth restriction on the activation of fetal hepatic glucose production. Mol Cell Endocrinol 2016; 435:61-68. [PMID: 26723529 PMCID: PMC4921201 DOI: 10.1016/j.mce.2015.12.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/16/2015] [Accepted: 12/18/2015] [Indexed: 02/07/2023]
Abstract
Glucose is the major fuel for fetal oxidative metabolism. A positive maternal-fetal glucose gradient drives glucose across the placenta and is sufficient to meet the demands of the fetus, eliminating the need for endogenous hepatic glucose production (HGP). However, fetuses with intrauterine growth restriction (IUGR) from pregnancies complicated by placental insufficiency have an early activation of HGP. Furthermore, this activated HGP is resistant to suppression by insulin. Here, we present the data demonstrating the activation of HGP in animal models, mostly fetal sheep, and human pregnancies with IUGR. We also discuss potential mechanisms and pathways that may produce and support HGP and hepatic insulin resistance in IUGR fetuses.
Collapse
Affiliation(s)
- Stephanie R Wesolowski
- Perinatal Research Center, Department of Pediatrics, University of Colorado School of Medicine, Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - William W Hay
- Perinatal Research Center, Department of Pediatrics, University of Colorado School of Medicine, Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
163
|
Harris RA, Alcott CE, Sullivan EL, Takahashi D, McCurdy CE, Comstock S, Baquero K, Blundell P, Frias AE, Kahr M, Suter M, Wesolowski S, Friedman JE, Grove KL, Aagaard KM. Genomic Variants Associated with Resistance to High Fat Diet Induced Obesity in a Primate Model. Sci Rep 2016; 6:36123. [PMID: 27811965 PMCID: PMC5095882 DOI: 10.1038/srep36123] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/07/2016] [Indexed: 12/28/2022] Open
Abstract
Maternal obesity contributes to an increased risk of lifelong morbidity and mortality for both the mother and her offspring. In order to better understand the molecular mechanisms underlying these risks, we previously established and extensively characterized a primate model in Macaca fuscata (Japanese macaque). In prior studies we have demonstrated that a high fat, caloric dense maternal diet structures the offspring’s epigenome, metabolome, and intestinal microbiome. During the course of this work we have consistently observed that a 36% fat diet leads to obesity in the majority, but not all, of exposed dams. In the current study, we sought to identify the genomic loci rendering resistance to obesity despite chronic consumption of a high fat diet in macaque dams. Through extensive phenotyping together with exon capture array and targeted resequencing, we identified three novel single nucleotide polymorphisms (SNPs), two in apolipoprotein B (APOB) and one in phospholipase A2 (PLA2G4A) that significantly associated with persistent weight stability and insulin sensitivity in lean macaques. By application of explicit orthogonal modeling (NOIA), we estimated the polygenic and interactive nature of these loci against multiple metabolic traits and their measures (i.e., serum LDL levels) which collectively render an obesity resistant phenotype in our adult female dams.
Collapse
Affiliation(s)
- R Alan Harris
- Department of Obstetrics &Gynecology, Division of Maternal-Fetal Medicine at Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA.,Department of Molecular and Human Genetics at Baylor College of Medicine, Houston, TX, USA
| | - Callison E Alcott
- Developmental Biology Interdisciplinary Program at Baylor College of Medicine, Houston, TX, USA
| | - Elinor L Sullivan
- Oregon National Primate Research Center, Oregon Health &Science University (OHSU), Beaverton, OR, USA.,Department of Biology, University of Portland, USA
| | - Diana Takahashi
- Oregon National Primate Research Center, Oregon Health &Science University (OHSU), Beaverton, OR, USA
| | - Carrie E McCurdy
- Department of Human Physiology, University of Oregon, Eugene, OR, USA
| | - Sarah Comstock
- Department of Biology, Corban University, Salem, OR, USA
| | - Karalee Baquero
- Oregon National Primate Research Center, Oregon Health &Science University (OHSU), Beaverton, OR, USA
| | - Peter Blundell
- Oregon National Primate Research Center, Oregon Health &Science University (OHSU), Beaverton, OR, USA
| | - Antonio E Frias
- Oregon National Primate Research Center, Oregon Health &Science University (OHSU), Beaverton, OR, USA.,Department of Obstetrics &Gynecology, Division of Maternal-Fetal Medicine, OHSU, Portland, OR, USA
| | - Maike Kahr
- Department of Obstetrics &Gynecology, Division of Maternal-Fetal Medicine at Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Melissa Suter
- Department of Obstetrics &Gynecology, Division of Maternal-Fetal Medicine at Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Stephanie Wesolowski
- Departments of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jacob E Friedman
- Departments of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kevin L Grove
- Oregon National Primate Research Center, Oregon Health &Science University (OHSU), Beaverton, OR, USA
| | - Kjersti M Aagaard
- Department of Obstetrics &Gynecology, Division of Maternal-Fetal Medicine at Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA.,Department of Molecular and Human Genetics at Baylor College of Medicine, Houston, TX, USA.,Developmental Biology Interdisciplinary Program at Baylor College of Medicine, Houston, TX, USA.,Oregon National Primate Research Center, Oregon Health &Science University (OHSU), Beaverton, OR, USA.,Department of Molecular and Cell Biology at Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
164
|
Comparison of maternal isocaloric high carbohydrate and high fat diets on osteogenic and adipogenic genes expression in adolescent mice offspring. Nutr Metab (Lond) 2016; 13:69. [PMID: 27777604 PMCID: PMC5069891 DOI: 10.1186/s12986-016-0130-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/06/2016] [Indexed: 01/12/2023] Open
Abstract
Background Maternal high fat/high calorie diet leads to adiposity and bone fracture in offspring. However, the effects of macronutrient distribution in maternal isocaloric diet have not been studied. The present study was designed to test the hypothesis that maternal isocaloric pair-fed high-carbohydrate diet will increase osteoblastic and decrease osteoclastic and adipogenic gene expression compared with high-fat diet in adolescent mice offspring. Methods Virgin female C57BL/6 mice were impregnated and fed either the AIN 93G isocaloric pair-fed high-carbohydrate (LF-HCD) or a high fat (HF-LCD) diet from the time of vaginal plug confirmation until the offspring was weaned. Results After adjusting for the sex of offspring, osteoprotegrin (OPG) and Ctnnb1 (beta-catenin) genes expression were significantly reduced by 98 % and 97 % in the bone of offspring born from the HF-LCD compared with the LF-HCD-fed mothers (p < 0.001 and p < 0.001, respectively). Peroxisome proliferator-activated receptor gamma-2 (PPAR γ2) gene expression in the bone of offspring born from the HF-LCD was 7.1-folds higher than the LF-HCD-fed mothers (p = 0.004). In the retroperitoneal fat mass of offspring born from HF-LCD, AdipoQ and LPL genes expression were respectively up-regulated 15.8 and 4.2-folds compared with the LF-HCD-fed mothers (p < 0.001 and p = 0.03, respectively). Conclusions Maternal isocaloric pair-fed high-carbohydrate diet enhances osteoblastogenesis and inhibits adipogenesis compared with high-fat diet in adolescent mice offspring.
Collapse
|
165
|
McCurdy CE, Schenk S, Hetrick B, Houck J, Drew BG, Kaye S, Lashbrook M, Bergman BC, Takahashi DL, Dean TA, Nemkov T, Gertsman I, Hansen KC, Philp A, Hevener AL, Chicco AJ, Aagaard KM, Grove KL, Friedman JE. Maternal obesity reduces oxidative capacity in fetal skeletal muscle of Japanese macaques. JCI Insight 2016; 1:e86612. [PMID: 27734025 DOI: 10.1172/jci.insight.86612] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Maternal obesity is proposed to alter the programming of metabolic systems in the offspring, increasing the risk for developing metabolic diseases; however, the cellular mechanisms remain poorly understood. Here, we used a nonhuman primate model to examine the impact of a maternal Western-style diet (WSD) alone, or in combination with obesity (Ob/WSD), on fetal skeletal muscle metabolism studied in the early third trimester. We find that fetal muscle responds to Ob/WSD by upregulating fatty acid metabolism, mitochondrial complex activity, and metabolic switches (CPT-1, PDK4) that promote lipid utilization over glucose oxidation. Ob/WSD fetuses also had reduced mitochondrial content, diminished oxidative capacity, and lower mitochondrial efficiency in muscle. The decrease in oxidative capacity and glucose metabolism was persistent in primary myotubes from Ob/WSD fetuses despite no additional lipid-induced stress. Switching obese mothers to a healthy diet prior to pregnancy did not improve fetal muscle mitochondrial function. Lastly, while maternal WSD alone led only to intermediary changes in fetal muscle metabolism, it was sufficient to increase oxidative damage and cellular stress. Our findings suggest that maternal obesity or WSD, alone or in combination, leads to programmed decreases in oxidative metabolism in offspring muscle. These alterations may have important implications for future health.
Collapse
Affiliation(s)
- Carrie E McCurdy
- Department of Human Physiology, University of Oregon, Eugene, Oregon, USA.,Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Simon Schenk
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, California, USA
| | - Byron Hetrick
- Department of Human Physiology, University of Oregon, Eugene, Oregon, USA
| | - Julie Houck
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Brian G Drew
- David Geffen School of Medicine, Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, University of California, Los Angeles, Los Angeles, California, USA.,Diabetes and Dyslipidaemia Laboratory, Baker IDI Heart and Diabetes Institute, Prahran, Victoria, Australia
| | - Spencer Kaye
- Departments of Health and Exercise Science and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Melanie Lashbrook
- Departments of Health and Exercise Science and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Bryan C Bergman
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Diana L Takahashi
- Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Tyler A Dean
- Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ilya Gertsman
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Andrew Philp
- School of Sport Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Andrea L Hevener
- David Geffen School of Medicine, Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, University of California, Los Angeles, Los Angeles, California, USA
| | - Adam J Chicco
- Departments of Health and Exercise Science and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Kjersti M Aagaard
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas, USA
| | - Kevin L Grove
- Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Beaverton, Oregon, USA.,Novo Nordisk Research Center, Seattle, Washington, USA
| | - Jacob E Friedman
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
166
|
Frihauf JB, Fekete ÉM, Nagy TR, Levin BE, Zorrilla EP. Maternal Western diet increases adiposity even in male offspring of obesity-resistant rat dams: early endocrine risk markers. Am J Physiol Regul Integr Comp Physiol 2016; 311:R1045-R1059. [PMID: 27654396 DOI: 10.1152/ajpregu.00023.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 08/30/2016] [Accepted: 09/12/2016] [Indexed: 01/08/2023]
Abstract
Maternal overnutrition or associated complications putatively mediate the obesogenic effects of perinatal high-fat diet on developing offspring. Here, we tested the hypothesis that a Western diet developmental environment increases adiposity not only in male offspring from obesity-prone (DIO) mothers, but also in those from obesity-resistant (DR) dams, implicating a deleterious role for the Western diet per se. Selectively bred DIO and DR female rats were fed chow (17% kcal fat) or Western diet (32%) for 54 days before mating and, thereafter, through weaning. As intended, despite chow-like caloric intake, Western diet increased prepregnancy weight gain and circulating leptin levels in DIO, but not DR, dams. Yet, in both genotypes, maternal Western diet increased the weight and adiposity of preweanlings, as early as in DR offspring, and increased plasma leptin, insulin, and adiponectin of weanlings. Although body weight normalized with chow feeding during adolescence, young adult Western diet offspring subsequently showed decreased energy expenditure and, in DR offspring, decreased lipid utilization as a fuel substrate. By mid-adulthood, maternal Western diet DR offspring ate more chow, weighed more, and were fatter than controls. Thus, maternal Western diet covertly programmed increased adiposity in childhood and adulthood, disrupted relations of energy regulatory hormones with body fat, and decreased energy expenditure in offspring of lean, genetically obesity-resistant mothers. Maternal Western diet exposure alone, without maternal obesity or overnutrition, can promote offspring weight gain.
Collapse
Affiliation(s)
- Jennifer B Frihauf
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California.,Neurosciences Graduate Program, University of California, San Diego, La Jolla, California
| | - Éva M Fekete
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California
| | - Tim R Nagy
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - Barry E Levin
- Neurology Service, VA Medical Center, East Orange, New Jersey; and.,Department of Neurology, Rutgers, New Jersey Medical School, Newark, New Jersey
| | - Eric P Zorrilla
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California; .,Neurosciences Graduate Program, University of California, San Diego, La Jolla, California
| |
Collapse
|
167
|
The effects of prenatal metformin on obesogenic diet-induced alterations in maternal and fetal fatty acid metabolism. Nutr Metab (Lond) 2016; 13:55. [PMID: 27555877 PMCID: PMC4994240 DOI: 10.1186/s12986-016-0115-9] [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: 06/29/2016] [Accepted: 08/13/2016] [Indexed: 12/21/2022] Open
Abstract
Background Maternal obesity may program the fetus and increase the susceptibility of the offspring to adult diseases. Metformin crosses the placenta and has been associated with decreased inflammation and reversal of fatty liver in obese leptin-deficient mice. We investigated the effects of metformin on maternal and fetal lipid metabolism and hepatic inflammation using a rat model of diet-induced obesity during pregnancy. Methods Female Wistar rats (6–7 weeks old) were fed normal or high calorie diets for 5 weeks. After mating with normal-diet fed males, half of the high calorie-fed dams received metformin (300 mg/kg, daily); dams (8 per group) continued diets through gestational day 19. Maternal and fetal livers and fetal brains were analyzed for fatty acids and for fatty acid metabolism-related gene expression. Data were analyzed by ANOVA followed by Dunnett’s post hoc testing. Results When compared to control-lean maternal livers, obesogenic-diet-exposed maternal livers showed significantly higher saturated fatty acids (14:0 and 16:0) and monounsaturated fatty acids (16:1n7 and 18:1n9) and lower polyunsaturated (18:2n6 and 20:4n6 [arachidonic acid]) and anti-inflammatory n3 polyunsaturated fatty acids (18:3n3 and 22:6n3 [docosahexaenoic acid]) (p < 0.05). Metformin did not affect diet-induced changes in maternal livers. Fetal livers exposed to the high calorie diet showed significantly increased saturated fatty acids (18:0) and monounsaturated fatty acids (18:1n9 and 18:1n7) and decreased polyunsaturated fatty acids (18:2n6, 20:4n6 and 22:6n3) and anti-inflammatory n3 polyunsaturated fatty acids, along with increased gene expression of fatty acid metabolism markers (Fasn, D5d, D6d, Scd1, Lxrα). Metformin significantly attenuated diet-induced inflammation and 18:1n9 and 22:6n3 in fetal livers, as well as n3 fatty acids (p < 0.05). Prenatal obesogenic diet exposure significantly increased fetal liver IFNγ levels (p < 0.05), which was reversed by maternal metformin treatment (p < 0.05). Conclusions Consumption of a high calorie diet significantly affected maternal and fetal fatty acid metabolism. It reduced anti-inflammatory polyunsaturated fatty acids in maternal and fetal livers, altered gene expression of fatty acid metabolism markers, and induced inflammation in the fetal livers. Prenatal metformin attenuated some diet-induced fatty acid changes and inflammation in the fetal livers without affecting maternal livers, suggesting that maternal metformin may impact fetal/neonatal fatty acid/lipid metabolism. Electronic supplementary material The online version of this article (doi:10.1186/s12986-016-0115-9) contains supplementary material, which is available to authorized users.
Collapse
|
168
|
Carr DJ, David AL, Aitken RP, Milne JS, Borowicz PP, Wallace JM, Redmer DA. Placental vascularity and markers of angiogenesis in relation to prenatal growth status in overnourished adolescent ewes. Placenta 2016; 46:79-86. [PMID: 27697225 PMCID: PMC5063007 DOI: 10.1016/j.placenta.2016.08.076] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 07/15/2016] [Accepted: 08/16/2016] [Indexed: 02/08/2023]
Abstract
Introduction Placental vascularity may be important in the development of fetal growth restriction (FGR). The overnourished adolescent ewe is a robust model of the condition, with ∼50% of offspring demonstrating FGR (birthweight >2 standard deviations below optimally-fed control mean). We studied whether placental vascularity, angiogenesis and glucose transport reflect FGR severity. Methods Singleton pregnancies were established in adolescent ewes either overnourished to putatively restrict fetoplacental growth (n = 27) or control-fed (n = 12). At 131d (term = 145d) pregnancies were interrupted and fetuses classified as FGR (n = 17, <4222 g, -2SD below control-fed mean) or non-FGR (n = 10). Placentome capillary area density (CAD), number density (CND), surface density (CSD), and area per capillary (APC) in the fetal cotyledon (COT) and maternal caruncle (CAR) were analysed using immunostaining. COT/CAR mRNA expression of angiogenic ligands/receptors and glucose transporters were measured by qRT-PCR. Results Fetal weight was reduced in FGR vs. Non-FGR/Control groups. Total placentome weight was Control > Non-FGR > FGR and fetal:placental weight ratios were higher in overnourished versus Control groups. COT vascular indices were Non-FGR > FGR > Control. COT-CAD, CSD and APC were significantly greater in Non-FGR overnourished versus Control and intermediate in FGR groups. CAR vascularity did not differ. CAR-VEGFA/FLT1/KDR/ANGPT1/ANGPT2/SLC2A1/SLC2A3 mRNA was lower and COT-ANGPT2 higher in overnourished versus Control groups. Discussion Relative to control-intake pregnancy, overnourished pregnancies are characterised by higher COT vascularity, potentially a compensatory response to reduced nutrient supply, reflected by higher fetal:placental weight ratios. Compared with overnourished pregnancies where fetal growth is relatively preserved, overnourished pregnancies culminating in marked FGR have less placental vascularity, suggesting incomplete adaptation to the prenatal insult. Overnourishment of adolescent sheep dams produces FGR in approximately 50% of cases. Cotyledonary vascularity is increased in overnourished vs. control-intake pregnancy. Cotyledonary vascularity is highest in non-FGR cases, suggesting greater adaptation. Changes in cotyledonary vascularity are mirrored by angiopoietin-2 mRNA expression. Caruncular angiogenic ligands are reduced in FGR without any changes in vascularity.
Collapse
Affiliation(s)
- David J Carr
- Rowett Institute of Nutrition and Health, University of Aberdeen, Greenburn Road, Bucksburn, Aberdeen, AB21 9SB, UK; UCL Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK.
| | - Anna L David
- UCL Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Raymond P Aitken
- Rowett Institute of Nutrition and Health, University of Aberdeen, Greenburn Road, Bucksburn, Aberdeen, AB21 9SB, UK
| | - John S Milne
- Rowett Institute of Nutrition and Health, University of Aberdeen, Greenburn Road, Bucksburn, Aberdeen, AB21 9SB, UK
| | - Pawel P Borowicz
- Department of Animal Sciences, North Dakota State University, Fargo, ND, 58108, USA
| | - Jacqueline M Wallace
- Rowett Institute of Nutrition and Health, University of Aberdeen, Greenburn Road, Bucksburn, Aberdeen, AB21 9SB, UK
| | - Dale A Redmer
- Department of Animal Sciences, North Dakota State University, Fargo, ND, 58108, USA
| |
Collapse
|
169
|
Enriori PJ, Chen W, Garcia-Rudaz MC, Grayson BE, Evans AE, Comstock SM, Gebhardt U, Müller HL, Reinehr T, Henry BA, Brown RD, Bruce CR, Simonds SE, Litwak SA, McGee SL, Luquet S, Martinez S, Jastroch M, Tschöp MH, Watt MJ, Clarke IJ, Roth CL, Grove KL, Cowley MA. α-Melanocyte stimulating hormone promotes muscle glucose uptake via melanocortin 5 receptors. Mol Metab 2016; 5:807-822. [PMID: 27688995 PMCID: PMC5034615 DOI: 10.1016/j.molmet.2016.07.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 07/26/2016] [Accepted: 07/28/2016] [Indexed: 01/21/2023] Open
Abstract
Objective Central melanocortin pathways are well-established regulators of energy balance. However, scant data exist about the role of systemic melanocortin peptides. We set out to determine if peripheral α-melanocyte stimulating hormone (α-MSH) plays a role in glucose homeostasis and tested the hypothesis that the pituitary is able to sense a physiological increase in circulating glucose and responds by secreting α-MSH. Methods We established glucose-stimulated α-MSH secretion using humans, non-human primates, and mouse models. Continuous α-MSH infusions were performed during glucose tolerance tests and hyperinsulinemic-euglycemic clamps to evaluate the systemic effect of α-MSH in glucose regulation. Complementary ex vivo and in vitro techniques were employed to delineate the direct action of α-MSH via the melanocortin 5 receptor (MC5R)–PKA axis in skeletal muscles. Combined treatment of non-selective/selective phosphodiesterase inhibitor and α-MSH was adopted to restore glucose tolerance in obese mice. Results Here we demonstrate that pituitary secretion of α-MSH is increased by glucose. Peripheral α-MSH increases temperature in skeletal muscles, acts directly on soleus and gastrocnemius muscles to significantly increase glucose uptake, and enhances whole-body glucose clearance via the activation of muscle MC5R and protein kinase A. These actions are absent in obese mice, accompanied by a blunting of α-MSH-induced cAMP levels in skeletal muscles of obese mice. Both selective and non-selective phosphodiesterase inhibition restores α-MSH induced skeletal muscle glucose uptake and improves glucose disposal in obese mice. Conclusion These data describe a novel endocrine circuit that modulates glucose homeostasis by pituitary α-MSH, which increases muscle glucose uptake and thermogenesis through the activation of a MC5R-PKA-pathway, which is disrupted in obesity. Glucose stimulates α-MSH release from the pituitary. Systemic α-MSH drives glucose disposal and thermogenesis in skeletal muscles. α-MSH acts on MC5R expressed on skeletal muscles and activate cAMP-PKA pathway. The combined treatment of nonselective or selective PDE 4 inhibitor and α-MSH ameliorates glucose intolerance in obese mice.
Collapse
Affiliation(s)
- Pablo J Enriori
- Biomedical Discovery Institute/Department of Physiology, Monash University, Vic, Australia
| | - Weiyi Chen
- Biomedical Discovery Institute/Department of Physiology, Monash University, Vic, Australia
| | - Maria C Garcia-Rudaz
- Biomedical Discovery Institute/Department of Physiology, Monash University, Vic, Australia
| | | | - Anne E Evans
- Division Neuroscience, Oregon Health and Science University, Oregon, USA
| | - Sarah M Comstock
- Division Neuroscience, Oregon Health and Science University, Oregon, USA
| | - Ursel Gebhardt
- Department of Pediatrics, Vestische Children Hospital Datteln, University of Witten/Herdecke, Germany
| | - Hermann L Müller
- Department of Pediatrics, Vestische Children Hospital Datteln, University of Witten/Herdecke, Germany
| | - Thomas Reinehr
- Department of Pediatrics, Klinikum Oldenburg GmbH, Germany
| | - Belinda A Henry
- Biomedical Discovery Institute/Department of Physiology, Monash University, Vic, Australia
| | - Russell D Brown
- Biomedical Discovery Institute/Department of Physiology, Monash University, Vic, Australia
| | - Clinton R Bruce
- Biomedical Discovery Institute/Department of Physiology, Monash University, Vic, Australia
| | - Stephanie E Simonds
- Biomedical Discovery Institute/Department of Physiology, Monash University, Vic, Australia
| | - Sara A Litwak
- Biomedical Discovery Institute/Department of Physiology, Monash University, Vic, Australia
| | - Sean L McGee
- Metabolic Research Unit, School of Medicine, Deakin University, Vic, Australia
| | - Serge Luquet
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, F-75205 Paris, France
| | - Sarah Martinez
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, F-75205 Paris, France
| | - Martin Jastroch
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg & Division of Metabolic Diseases, Technische Universität, München, Germany
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg & Division of Metabolic Diseases, Technische Universität, München, Germany
| | - Matthew J Watt
- Biomedical Discovery Institute/Department of Physiology, Monash University, Vic, Australia
| | - Iain J Clarke
- Biomedical Discovery Institute/Department of Physiology, Monash University, Vic, Australia
| | - Christian L Roth
- Division of Endocrinology, Seattle Children's Hospital Research Institute, WA, USA
| | - Kevin L Grove
- Division Neuroscience, Oregon Health and Science University, Oregon, USA
| | - Michael A Cowley
- Biomedical Discovery Institute/Department of Physiology, Monash University, Vic, Australia.
| |
Collapse
|
170
|
de Fante T, Simino LA, Reginato A, Payolla TB, Vitoréli DCG, de Souza M, Torsoni MA, Milanski M, Torsoni AS. Diet-Induced Maternal Obesity Alters Insulin Signalling in Male Mice Offspring Rechallenged with a High-Fat Diet in Adulthood. PLoS One 2016; 11:e0160184. [PMID: 27479001 PMCID: PMC4968809 DOI: 10.1371/journal.pone.0160184] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/14/2016] [Indexed: 12/27/2022] Open
Abstract
Modern lifestyle has resulted in an increase in the prevalence of obesity and its comorbidities in pregnant women and the young population. It has been well established that the consumption of a high-fat diet (HFD) has many direct effects on glucose metabolism. However, it is important to assess whether maternal consumption of a HFD during critical periods of development can lead to metabolic changes in the offspring metabolism. This study evaluated the potential effects of metabolic programming on the impairment of insulin signalling in recently weaned offspring from obese dams. Additionally, we investigated if early exposure to an obesogenic environment could exacerbate the impairment of glucose metabolism in adult life in response to a HFD. Swiss female mice were fed with Standard Chow (SC) or a HFD during gestation and lactation and tissues from male offspring were analysed at d28 and d82. Offspring from obese dams had greater weight gain and higher adiposity and food intake than offspring from control dams. Furthermore, they showed impairment in insulin signalling in central and peripheral tissues, which was associated with the activation of inflammatory pathways. Adipose tissue was ultimately the most affected in adult offspring after HFD rechallenge; this may have contributed to the metabolic deregulation observed. Overall, our results suggest that diet-induced maternal obesity leads to increased susceptibility to obesity and impairment of insulin signalling in offspring in early and late life that cannot be reversed by SC consumption, but can be aggravated by HFD re-exposure.
Collapse
Affiliation(s)
- Thaís de Fante
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas–UNICAMP, Limeira, São Paulo, Brazil
| | - Laís Angélica Simino
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas–UNICAMP, Limeira, São Paulo, Brazil
| | - Andressa Reginato
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas–UNICAMP, Limeira, São Paulo, Brazil
| | - Tanyara Baliani Payolla
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas–UNICAMP, Limeira, São Paulo, Brazil
| | | | - Monique de Souza
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas–UNICAMP, Limeira, São Paulo, Brazil
| | - Márcio Alberto Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas–UNICAMP, Limeira, São Paulo, Brazil
| | - Marciane Milanski
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas–UNICAMP, Limeira, São Paulo, Brazil
| | - Adriana Souza Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas–UNICAMP, Limeira, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
171
|
Kwon EJ, Lee HA, You YA, Park H, Cho SJ, Ha EH, Kim YJ. DNA methylations of MC4R and HNF4α are associated with increased triglyceride levels in cord blood of preterm infants. Medicine (Baltimore) 2016; 95:e4590. [PMID: 27583872 PMCID: PMC5008556 DOI: 10.1097/md.0000000000004590] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The association of preterm birth with obesity and metabolic syndrome later in life is well established. Although the biological mechanism for this association is poorly understood, epigenetic alterations of metabolic-related genes in early life may have important roles in metabolic dysfunction. Thus, we investigated the associations of DNA methylations of melanocortin 4 receptor (MC4R) and hepatocyte nuclear factor 4 alpha (HNF4α) with metabolic profiles in cord blood of term and preterm infants.We measured metabolic profiles in cord blood samples of 85 term and 85 preterm infants. DNA methylation and mRNA expression levels of MC4R and HNF4α in cord blood cells were quantified using pyrosequencing and real-time PCR. Triglyceride (TG) levels were grouped by percentile as low (<10th percentile), mid (11th-89th percentiles), and high (>90th percentile). A multiple linear regression model was used to assess the differential effects of DNA methylation on metabolic indices in cord blood between term and preterm infants.The beta-coefficients for associations between TG levels and methylation statuses of MC4R-CpG3 and HNF4α-CpG2 in the P1 promoter differed significantly between term and preterm infants (P = 0.04 and P = 0.003, respectively). DNA methylation statuses of MC4R-CpG3 and HNF4α-CpG2 in the P1 promoter were significantly lower in preterm infants in the high-TG group compared with those in the mid- and low-TG groups (P = 0.01). Notably, preterm infants in the high-TG group had higher TG levels in cord blood than term infants in the high-TG group (60.49 vs 54.57 mg/dL). In addition, MC4R and HNF4α expression levels were higher in preterm infants than in term infants (P < 0.05).Epigenetic alterations of the newly identified genes MC4R and HNF4α in early life might contribute to metabolic profile changes, especially increased TG levels, in the cord blood of preterm infants.
Collapse
Affiliation(s)
- Eun Jin Kwon
- Department of Obstetrics and Gynecology
- Department of Occupational and Environmental Medicine
| | | | | | | | - Su Jin Cho
- Department of Pediatrics, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Eun Hee Ha
- Department of Occupational and Environmental Medicine
| | - Young Ju Kim
- Department of Obstetrics and Gynecology
- Correspondence: Young Ju Kim, Department of Obstetrics and Gynecology, School of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-ku, Seoul 158-710, Republic of Korea (e-mail: )
| |
Collapse
|
172
|
Ambrosetti V, Guerra M, Ramírez LA, Reyes A, Álvarez D, Olguín S, González-Mañan D, Fernandois D, Sotomayor-Zárate R, Cruz G. Increase in endogenous estradiol in the progeny of obese rats is associated with precocious puberty and altered follicular development in adulthood. Endocrine 2016; 53:258-70. [PMID: 26767652 DOI: 10.1007/s12020-016-0858-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 01/04/2016] [Indexed: 01/07/2023]
Abstract
Maternal obesity during pregnancy has been related with several pathological states in offspring. However, the impact of maternal obesity on reproductive system on the progeny is beginning to be elucidated. In this work, we characterize the effect of maternal obesity on puberty onset and follicular development in adult offspring in rats. We also propose that alterations in ovarian physiology observed in offspring of obese mothers are due to increased levels of estradiol during early development. Offspring of control dams and offspring of dams exposed to a high-fat diet (HF) were studied at postnatal days (PND) 1, 7, 14, 30, 60, and 120. Body weight and onset of puberty were measured. Counting of ovarian follicles was performed at PND 60 and 120. Serum estradiol, estriol, androstenedione, FSH, LH, and insulin levels were measured by ELISA. Hepatic CYP3A2 expression was determined by Western blot. HF rats had a higher weight than controls at all ages and they also had a precocious puberty. Estradiol levels were increased while CYP3A2 expression was reduced from PND 1 until PND 60 in HF rats compared to controls. Estriol was decreased at PND60 in HF rats. Ovaries from HF rats had a decrease in antral follicles at PND60 and PND120 and an increase in follicular cysts at PND60 and PND120. In this work, we demonstrated that maternal obesity in rats alters follicular development and induces follicular cysts generation in the adult offspring. We observed that maternal obesity produces an endocrine disruption through increasing endogenous estradiol in early life. A programmed failure in hepatic metabolism of estradiol is probably the cause of its increase.
Collapse
Affiliation(s)
- Valery Ambrosetti
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Facultad de Ciencias, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, 2360102, Valparaiso, Chile
| | - Marcelo Guerra
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Facultad de Ciencias, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, 2360102, Valparaiso, Chile
| | - Luisa A Ramírez
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Facultad de Ciencias, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, 2360102, Valparaiso, Chile
| | - Aldo Reyes
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Facultad de Ciencias, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, 2360102, Valparaiso, Chile
| | - Daniela Álvarez
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Facultad de Ciencias, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, 2360102, Valparaiso, Chile
| | - Sofía Olguín
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Facultad de Ciencias, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, 2360102, Valparaiso, Chile
| | - Daniel González-Mañan
- Molecular and Clinical Pharmacology Program, Faculty of Medicine, Institute of Biomedical Sciences, University of Chile, 8380492, Santiago, Chile
| | - Daniela Fernandois
- Programa de Doctorado en Farmacología, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, 8380492, Santiago, Chile
| | - Ramón Sotomayor-Zárate
- Laboratorio de Neuroquímica y Neurofarmacología, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Facultad de Ciencias, Instituto de Fisiología, Universidad de Valparaíso, 2360102, Valparaiso, Chile
| | - Gonzalo Cruz
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Facultad de Ciencias, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, 2360102, Valparaiso, Chile.
| |
Collapse
|
173
|
Chon YE, Kim KJ, Jung KS, Kim SU, Park JY, Kim DY, Ahn SH, Chon CY, Chung JB, Park KH, Bae JC, Han KH. The Relationship between Type 2 Diabetes Mellitus and Non-Alcoholic Fatty Liver Disease Measured by Controlled Attenuation Parameter. Yonsei Med J 2016; 57:885-92. [PMID: 27189281 PMCID: PMC4951464 DOI: 10.3349/ymj.2016.57.4.885] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 09/21/2015] [Accepted: 10/17/2015] [Indexed: 01/02/2023] Open
Abstract
PURPOSE The severity of non-alcoholic fatty liver disease (NAFLD) in type 2 diabetes mellitus (T2DM) population compared with that in normal glucose tolerance (NGT) individuals has not yet been quantitatively assessed. We investigated the prevalence and the severity of NAFLD in a T2DM population using controlled attenuation parameter (CAP). MATERIALS AND METHODS Subjects who underwent testing for biomarkers related to T2DM and CAP using Fibroscan® during a regular health check-up were enrolled. CAP values of 250 dB/m and 300 dB/m were selected as the cutoffs for the presence of NAFLD and for moderate to severe NAFLD, respectively. Biomarkers related to T2DM included fasting glucose/insulin, fasting C-peptide, hemoglobin A1c (HbA1c), glycoalbumin, and homeostasis model assessment of insulin resistance of insulin resistance (HOMA-IR). RESULTS Among 340 study participants (T2DM, n=66; pre-diabetes, n=202; NGT, n=72), the proportion of subjects with NAFLD increased according to the glucose tolerance status (31.9% in NGT; 47.0% in pre-diabetes; 57.6% in T2DM). The median CAP value was significantly higher in subjects with T2DM (265 dB/m) than in those with pre-diabetes (245 dB/m) or NGT (231 dB/m) (all p<0.05). Logistic regression analysis showed that subjects with moderate to severe NAFLD had a 2.8-fold (odds ratio) higher risk of having T2DM than those without NAFLD (p=0.02; 95% confidence interval, 1.21-6.64), and positive correlations between the CAP value and HOMA-IR (ρ0.407) or fasting C-peptide (ρ0.402) were demonstrated. CONCLUSION Subjects with T2DM had a higher prevalence of severe NAFLD than those with NGT. Increased hepatic steatosis was significantly associated with the presence of T2DM, and insulin resistance induced by hepatic fat may be an important mechanistic connection.
Collapse
Affiliation(s)
- Young Eun Chon
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Liver Cirrhosis Clinical Research Center, Seoul, Korea
| | - Kwang Joon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Executive Healthcare Clinic, Severance Hospital, Yonsei Health System, Seoul, Korea
| | - Kyu Sik Jung
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Liver Cirrhosis Clinical Research Center, Seoul, Korea
| | - Seung Up Kim
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Liver Cirrhosis Clinical Research Center, Seoul, Korea
| | - Jun Yong Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Liver Cirrhosis Clinical Research Center, Seoul, Korea
| | - Do Young Kim
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Liver Cirrhosis Clinical Research Center, Seoul, Korea
| | - Sang Hoon Ahn
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Liver Cirrhosis Clinical Research Center, Seoul, Korea
| | - Chae Yoon Chon
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Liver Cirrhosis Clinical Research Center, Seoul, Korea
| | - Jae Bock Chung
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Liver Cirrhosis Clinical Research Center, Seoul, Korea
- Executive Healthcare Clinic, Severance Hospital, Yonsei Health System, Seoul, Korea
| | - Kyeong Hye Park
- Executive Healthcare Clinic, Severance Hospital, Yonsei Health System, Seoul, Korea
| | - Ji Cheol Bae
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kwang Hyub Han
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Liver Cirrhosis Clinical Research Center, Seoul, Korea.
| |
Collapse
|
174
|
Edlow AG, Hui L, Wick HC, Fried I, Bianchi DW. Assessing the fetal effects of maternal obesity via transcriptomic analysis of cord blood: a prospective case-control study. BJOG 2016; 123:180-9. [PMID: 26840378 DOI: 10.1111/1471-0528.13795] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2015] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To analyse fetal gene expression at term using umbilical cord blood, in order to provide insights into the effects of maternal obesity on human development. DESIGN Prospective case-control study. SETTING Academic tertiary care centre. POPULATION Eight obese (body mass index ≥30 kg/m(2)) and eight lean (body mass index <25 kg/m(2)) pregnant women undergoing prelabour caesarean delivery at term. METHODS Women were matched for gestational age and fetal sex. Cord blood RNA was extracted and hybridised to gene expression arrays. Differentially regulated genes were identified using paired t-tests and the Benjamini-Hochberg correction. Functional analyses were performed using Ingenuity Pathway Analysis, BioGPS and Gene Set Enrichment Analysis with a fetal-specific annotation. Z-scores ≥2.0 or P-values <0.01 were considered significant. MAIN OUTCOME MEASURE Functions of differentially regulated genes in fetuses of obese women. RESULTS A total of 701 differentially regulated genes were identified, producing an expression profile implicating neurodegeneration, decreased survival of sensory neurons, and decreased neurogenesis in the fetuses of obese women. Upstream regulators related to inflammatory signalling were significantly activated; those related to insulin receptor signalling, lipid homeostasis, regulation of axonal guidance, and cellular response to oxidative stress were significantly inhibited. Of 26 tissue-specific genes that were differentially regulated in fetuses of obese women, six mapped to the fetal brain. CONCLUSION Maternal obesity affects fetal gene expression at term, implicating dysregulated brain development, inflammatory and immune signalling, glucose and lipid homeostasis, and oxidative stress. This may have implications for postnatal neurodevelopment and metabolism.
Collapse
Affiliation(s)
- A G Edlow
- Mother Infant Research Institute and Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA, USA
| | - L Hui
- Mother Infant Research Institute and Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA, USA.,Department of Perinatal Medicine, Mercy Hospital for Women, Heidelberg, Vic., Australia
| | - H C Wick
- Department of Computer Science, Tufts University, Medford, MA, USA
| | - I Fried
- Department of Computer Science, Tufts University, Medford, MA, USA
| | - D W Bianchi
- Mother Infant Research Institute and Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
175
|
Trans-Fatty Acids Aggravate Obesity, Insulin Resistance and Hepatic Steatosis in C57BL/6 Mice, Possibly by Suppressing the IRS1 Dependent Pathway. Molecules 2016; 21:molecules21060705. [PMID: 27248994 PMCID: PMC6273562 DOI: 10.3390/molecules21060705] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/12/2016] [Accepted: 05/19/2016] [Indexed: 12/14/2022] Open
Abstract
Trans-fatty acid consumption has been reported as a risk factor for metabolic disorders and targeted organ damages. Nonetheless, little is known about the roles and mechanisms of trans-fatty acids in obesity, insulin resistance (IR) and hepatic steatosis. Adult C57BL/6 male mice were fed with four different diets for 20 weeks: normal diet (ND), high fat diet (HFD), low trans-fatty acids diet (LTD) and high trans-fatty acid diet (HTD). The diet-induced metabolic disorders were assessed by evaluating body weight, glucose tolerance test, hepatic steatosis and plasma lipid profiles post 20-week diet. Histological (H&E, Oil-Red-O) staining and western blot analysis were employed to assess liver steatosis and potential signaling pathways. After 20-weeks of diet, the body weights of the four groups were 29.61 ± 1.89 g (ND), 39.04 ± 4.27 g (HFD), 34.09 ± 2.62 g (LTD) and 43.78 ± 4.27 g (HTD) (p < 0.05), respectively. HFD intake significantly impaired glucose tolerance, which was impaired further in the mice consuming the HTD diet. The effect was further exacerbated by HTD diet. Moreover, the HTD group exhibited significantly more severe liver steatosis compared with HFD group possibly through regulating adipose triglyceride lipase. The group consuming the HTD also exhibited significantly reduced levels of IRS1, phosphor-PKC and phosphor-AKT. These results support our hypothesis that consumption of a diet high in trans-fatty acids induces higher rates of obesity, IR and hepatic steatosis in male C57BL/6 mice, possibly by suppressing the IRS1dependent pathway.
Collapse
|
176
|
Alam S, Mustafa G, Alam M, Ahmad N. Insulin resistance in development and progression of nonalcoholic fatty liver disease. World J Gastrointest Pathophysiol 2016; 7:211-217. [PMID: 27190693 PMCID: PMC4867400 DOI: 10.4291/wjgp.v7.i2.211] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 01/03/2016] [Accepted: 03/09/2016] [Indexed: 02/06/2023] Open
Abstract
Although insulin resistance (IR) is strongly associated with nonalcoholic fatty liver disease (NAFLD), the association of IR and NAFLD is not universal and correlation between IR and severity of NAFLD is still controversial. In this review, we summarize recent evidence that partially dissociates insulin resistance from NAFLD. It has also been reported that single-nucleotide polymorphisms in the diacylglycerol acyltransferase gene, rather than IR, account for the variability in liver fat content. Polymorphisms of the patatin-like phospholipase 3 gene have also been reported to be associated with NAFLD without metabolic syndrome, which suggests that genetic conditions that promote the development of fatty changes in the liver may occur independently of IR. Moreover, environmental factors such as nutrition and physical activity as well as small intestinal bacterial overgrowth have been linked to the pathogenesis of NAFLD, although some of the data are conflicting. Therefore, findings from both genetically engineered animal models and humans with genetic conditions, as well as recent studies that have explored the role of environmental factors, have confirmed the view that NAFLD is a polygenic disease process caused by both genetic and environmental factors. Therefore, IR is not the sole predictor of the pathogenesis of NAFLD.
Collapse
|
177
|
Hernandez TL. Carbohydrate Content in the GDM Diet: Two Views: View 1: Nutrition Therapy in Gestational Diabetes: The Case for Complex Carbohydrates. Diabetes Spectr 2016; 29:82-8. [PMID: 27182176 PMCID: PMC4865387 DOI: 10.2337/diaspect.29.2.82] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
IN BRIEF Restriction of dietary carbohydrate has been the cornerstone for treatment of gestational diabetes mellitus (GDM). However, there is evidence that a balanced liberalization of complex carbohydrate as part of an overall eating plan in GDM meets treatment goals and may mitigate maternal adipose tissue insulin resistance, both of which may promote optimal metabolic outcomes for mother and offspring.
Collapse
Affiliation(s)
- Teri L Hernandez
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes and College of Nursing, University of Colorado, Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
178
|
Soderborg TK, Borengasser SJ, Barbour LA, Friedman JE. Microbial transmission from mothers with obesity or diabetes to infants: an innovative opportunity to interrupt a vicious cycle. Diabetologia 2016; 59:895-906. [PMID: 26843076 PMCID: PMC4829383 DOI: 10.1007/s00125-016-3880-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/06/2016] [Indexed: 12/28/2022]
Abstract
Maternal obesity and diabetes dramatically increase the long-term risk for obesity in the next generation, and pregnancy and lactation may be critical periods at which to aim primary prevention to break the obesity cycle. It is becoming increasingly clear that the gut microbiome in newborns and infants plays a significant role in gut health and therefore child development. Alteration of the early infant gut microbiome has been correlated with the development of childhood obesity and autoimmune conditions, including asthma, allergies and, more recently, type 1 diabetes. This is likely to be due to complex interactions between mode of delivery, antibiotic use, maternal diet, components of breastfeeding and a network of regulatory events involving both the innate and adaptive immune systems within the infant host. Each of these factors are critical for informing microbiome development and can affect immune signalling, toxin release and metabolic signals, including short-chain fatty acids and bile acids, that regulate appetite, metabolism and inflammation. In several randomised controlled trials, probiotics have been administered with the aim of targeting the microbiome during pregnancy to improve maternal and infant health but the findings have often been confounded by mode of delivery, antibiotic use, ethnicity, infant sex, maternal health and length of exposure. Understanding how nutritional exposure, including breast milk, affects the assembly and development of both maternal and infant microbial communities may help to identify targeted interventions during pregnancy and in infants born to mothers with obesity or diabetes to slow the transmission of obesity risk to the next generation. The aim of this review is to discuss influences on infant microbiota colonisation and the mechanism(s) underlying how alterations due to maternal obesity and diabetes may lead to increased risk of childhood obesity.
Collapse
Affiliation(s)
- Taylor K Soderborg
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Anschutz Medical Campus, Mail Stop 8106, 12801 East 17th Avenue, Aurora, CO, 80045, USA
| | - Sarah J Borengasser
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Linda A Barbour
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Jacob E Friedman
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Anschutz Medical Campus, Mail Stop 8106, 12801 East 17th Avenue, Aurora, CO, 80045, USA.
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA.
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
179
|
Capobianco V, Caterino M, Iaffaldano L, Nardelli C, Sirico A, Del Vecchio L, Martinelli P, Pastore L, Pucci P, Sacchetti L. Proteome analysis of human amniotic mesenchymal stem cells (hA-MSCs) reveals impaired antioxidant ability, cytoskeleton and metabolic functionality in maternal obesity. Sci Rep 2016; 6:25270. [PMID: 27125468 PMCID: PMC4850482 DOI: 10.1038/srep25270] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/13/2016] [Indexed: 12/20/2022] Open
Abstract
Maternal obesity increases the risk of obesity and/or obesity-related diseases in the offspring of animal models. The aim of this study was to identify metabolic dysfunctions that could represent an enhanced risk for human obesity or obesity-related diseases in newborn or in adult life, similar to what occurs in animal models. To this aim, we studied the proteome of 12 obese (Ob-) and 6 non-obese (Co-) human amniotic mesenchymal stem cells (hA-MSCs) obtained from women at delivery by cesarean section (pre-pregnancy body mass index [mean ± SD]: 42.7 ± 7.7 and 21.3 ± 3.3 kg/m2, respectively). The proteome, investigated by two-dimensional fluorescence difference gel electrophoresis/mass spectrometry, revealed 62 differently expressed proteins in Ob- vs Co-hA-MSCs (P < 0.05), nine of which were confirmed by western blotting. Bioinformatics analysis showed that these 62 proteins are involved in several statistically significant pathways (P < 0.05), including the stress response, cytoskeleton and metabolic pathways. Oxidative stress was shown to be an early triggering factor of tissue fat accumulation and obesity-related disorders in the offspring of obese animal models. Our finding of a reduced stress response in Ob-hA-MSCs suggests that a similar mechanism could occur also in humans. Long-term follow-up studies of newborns of obese mothers are required to verify this hypothesis.
Collapse
Affiliation(s)
- Valentina Capobianco
- CEINGE-Biotecnologie Avanzate S.C.a R.L., Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Marianna Caterino
- CEINGE-Biotecnologie Avanzate S.C.a R.L., Via Gaetano Salvatore 486, 80145 Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Laura Iaffaldano
- CEINGE-Biotecnologie Avanzate S.C.a R.L., Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Carmela Nardelli
- CEINGE-Biotecnologie Avanzate S.C.a R.L., Via Gaetano Salvatore 486, 80145 Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Angelo Sirico
- Dipartimento di Neuroscienze e Scienze Riproduttive ed Odontostomatologiche, Via S. Pansini 5, 80131 Naples, Italy
| | - Luigi Del Vecchio
- CEINGE-Biotecnologie Avanzate S.C.a R.L., Via Gaetano Salvatore 486, 80145 Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Pasquale Martinelli
- Dipartimento di Neuroscienze e Scienze Riproduttive ed Odontostomatologiche, Via S. Pansini 5, 80131 Naples, Italy
| | - Lucio Pastore
- CEINGE-Biotecnologie Avanzate S.C.a R.L., Via Gaetano Salvatore 486, 80145 Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Pietro Pucci
- CEINGE-Biotecnologie Avanzate S.C.a R.L., Via Gaetano Salvatore 486, 80145 Naples, Italy.,Dipartimento di Scienze chimiche, Via Cintia, Complesso Monte Sant'Angelo 21, 80126 Naples, Italy
| | - Lucia Sacchetti
- CEINGE-Biotecnologie Avanzate S.C.a R.L., Via Gaetano Salvatore 486, 80145 Naples, Italy
| |
Collapse
|
180
|
Bruce KD, Szczepankiewicz D, Sihota KK, Ravindraanandan M, Thomas H, Lillycrop KA, Burdge GC, Hanson MA, Byrne CD, Cagampang FR. Altered cellular redox status, sirtuin abundance and clock gene expression in a mouse model of developmentally primed NASH. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:584-93. [PMID: 27040510 PMCID: PMC4874946 DOI: 10.1016/j.bbalip.2016.03.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 03/07/2016] [Accepted: 03/25/2016] [Indexed: 02/06/2023]
Abstract
Background We have previously shown that high fat (HF) feeding during pregnancy primes the development of non-alcoholic steatohepatits (NASH) in the adult offspring. However, the underlying mechanisms are unclear. Aims Since the endogenous molecular clock can regulate hepatic lipid metabolism, we investigated whether exposure to a HF diet during development could alter hepatic clock gene expression and contribute to NASH onset in later life. Methods Female mice were fed either a control (C, 7% kcal fat) or HF (45% kcal fat) diet. Offspring were fed either a C or HF diet resulting in four offspring groups: C/C, C/HF, HF/C and HF/HF. NAFLD progression, cellular redox status, sirtuin expression (Sirt1, Sirt3), and the expression of core clock genes (Clock, Bmal1, Per2, Cry2) and clock-controlled genes involved in lipid metabolism (Rev-Erbα, Rev-Erbβ, RORα, and Srebp1c) were measured in offspring livers. Results Offspring fed a HF diet developed NAFLD. However HF fed offspring of mothers fed a HF diet developed NASH, coupled with significantly reduced NAD+/NADH (p < 0.05, HF/HF vs C/C), Sirt1 (p < 0.001, HF/HF vs C/C), Sirt3 (p < 0.01, HF/HF vs C/C), perturbed clock gene expression, and elevated expression of genes involved lipid metabolism, such as Srebp1c (p < 0.05, C/HF and HF/HF vs C/C). Conclusion Our results suggest that exposure to excess dietary fat during early and post-natal life increases the susceptibility to develop NASH in adulthood, involving altered cellular redox status, reduced sirtuin abundance, and desynchronized clock gene expression. Offspring of mothers fed a high fat diet show severe fatty liver in later life. HF feeding is associated with altered cellular redox status and reduced sirtuin gene expression. HF feeding desynchronises the expression of core clock genes and lipogenic transcription factors. Exposure to a HF diet during development causes changes in liver metabolism that precede severe fatty liver disease.
Collapse
Affiliation(s)
- Kimberley D Bruce
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK; University of Colorado Anschutz Medical Campus, Endocrinology, Metabolism and Diabetes, Aurora, USA.
| | - Dawid Szczepankiewicz
- Poznań University of Life Sciences, Department of Animal Physiology and Biochemistry, Poznań, Poland
| | - Kiran K Sihota
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Manoj Ravindraanandan
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Hugh Thomas
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Karen A Lillycrop
- Centre for Biological Sciences, Institute of Developmental Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | - Graham C Burdge
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Mark A Hanson
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Christopher D Byrne
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Felino R Cagampang
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| |
Collapse
|
181
|
Padmanabhan V, Cardoso RC, Puttabyatappa M. Developmental Programming, a Pathway to Disease. Endocrinology 2016; 157:1328-40. [PMID: 26859334 PMCID: PMC4816734 DOI: 10.1210/en.2016-1003] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 01/30/2016] [Indexed: 02/07/2023]
Abstract
Accumulating evidence suggests that insults occurring during the perinatal period alter the developmental trajectory of the fetus/offspring leading to long-term detrimental outcomes that often culminate in adult pathologies. These perinatal insults include maternal/fetal disease states, nutritional deficits/excess, stress, lifestyle choices, exposure to environmental chemicals, and medical interventions. In addition to reviewing the various insults that contribute to developmental programming and the benefits of animal models in addressing underlying mechanisms, this review focuses on the commonalities in disease outcomes stemming from various insults, the convergence of mechanistic pathways via which various insults can lead to common outcomes, and identifies the knowledge gaps in the field and future directions.
Collapse
Affiliation(s)
- Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan 48109-5718
| | - Rodolfo C Cardoso
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan 48109-5718
| | - Muraly Puttabyatappa
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan 48109-5718
| |
Collapse
|
182
|
Roberts VH, Lo JO, Salati JA, Lewandowski KS, Lindner JR, Morgan TK, Frias AE. Quantitative assessment of placental perfusion by contrast-enhanced ultrasound in macaques and human subjects. Am J Obstet Gynecol 2016; 214:369.e1-8. [PMID: 26928151 DOI: 10.1016/j.ajog.2016.01.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/18/2015] [Accepted: 01/02/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND The uteroplacental vascular supply is a critical determinant of placental function and fetal growth. Current methods for the in vivo assessment of placental blood flow are limited. OBJECTIVE We demonstrate the feasibility of the use of contrast-enhanced ultrasound imaging to visualize and quantify perfusion kinetics in the intervillous space of the primate placenta. STUDY DESIGN Pregnant Japanese macaques were studied at mid second trimester and in the early third trimester. Markers of injury were assessed in placenta samples from animals with or without contrast-enhanced ultrasound exposure (n = 6/group). Human subjects were recruited immediately before scheduled first-trimester pregnancy termination. All studies were performed with maternal intravenous infusion of lipid-shelled octofluoropropane microbubbles with image acquisition with a multipulse contrast-specific algorithm with destruction-replenishment analysis of signal intensity for assessment of perfusion. RESULTS In macaques, the rate of perfusion in the intervillous space was increased with advancing gestation. No evidence of microvascular hemorrhage or acute inflammation was found in placental villous tissue and expression levels of caspase-3, nitrotyrosine and heat shock protein 70 as markers of apoptosis, nitrative, and oxidative stress, respectively, were unchanged by contrast-enhanced ultrasound exposure. In humans, placental perfusion was visualized at 11 weeks gestation, and preliminary data reveal regional differences in intervillous space perfusion within an individual placenta. By electron microscopy, we demonstrate no evidence of ultrastructure damage to the microvilli on the syncytiotrophoblast after first-trimester ultrasound studies. CONCLUSIONS Use of contrast-enhanced ultrasound did not result in placental structural damage and was able to identify intervillous space perfusion rate differences within a placenta. Contrast-enhanced ultrasound imaging may offer a safe clinical tool for the identification of pregnancies that are at risk for vascular insufficiency; early recognition may facilitate intervention and improved pregnancy outcomes.
Collapse
|
183
|
Expression of epigenetic machinery genes is sensitive to maternal obesity and weight loss in relation to fetal growth in mice. Clin Epigenetics 2016; 8:22. [PMID: 26925174 PMCID: PMC4769534 DOI: 10.1186/s13148-016-0188-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/12/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Maternal obesity impacts fetal growth and pregnancy outcomes. To counteract the deleterious effects of obesity on fertility and pregnancy issue, preconceptional weight loss is recommended to obese women. Whether this weight loss is beneficial/detrimental for offspring remains poorly explored. Epigenetic mechanisms could be affected by maternal weight changes, perturbing expression of key developmental genes in the placenta or fetus. Our aim was to investigate the effects of chronic maternal obesity on feto-placental growth along with the underlying epigenetic mechanisms. We also tested whether preconceptional weight loss could alleviate these effects. RESULTS Female mice were fed either a control diet (CTRL group), a high-fat diet (obese (OB) group), or a high-fat diet switched to a control diet 2 months before conception (weight loss (WL) group). At mating, OB females presented an obese phenotype while WL females normalized metabolic parameters. At embryonic day 18.5 (E18.5), fetuses from OB females presented fetal growth restriction (FGR; -13 %) and 28 % of the fetuses were small for gestational age (SGA). Fetuses from WL females normalized this phenotype. The expression of 60 epigenetic machinery genes and 32 metabolic genes was measured in the fetal liver, placental labyrinth, and junctional zone. We revealed 23 genes altered by maternal weight trajectories in at least one of three tissues. The fetal liver and placental labyrinth were more responsive to maternal obesity than junctional zone. One third (18/60) of the epigenetic machinery genes were differentially expressed between at least two maternal groups. Interestingly, genes involved in the histone acetylation pathway were particularly altered (13/18). In OB group, lysine acetyltransferases and Bromodomain-containing protein 2 were upregulated, while most histone deacetylases were downregulated. In WL group, the expression of only a subset of these genes was normalized. CONCLUSIONS This study highlights the high sensitivity of the epigenetic machinery gene expression, and particularly the histone acetylation pathway, to maternal obesity. These obesity-induced transcriptional changes could alter the placental and the hepatic epigenome, leading to FGR. Preconceptional weight loss appears beneficial to fetal growth, but some effects of previous obesity were retained in offspring phenotype.
Collapse
|
184
|
Tobita T, Guzman-Lepe J, Takeishi K, Nakao T, Wang Y, Meng F, Deng CX, Collin de l’Hortet A, Soto-Gutierrez A. SIRT1 Disruption in Human Fetal Hepatocytes Leads to Increased Accumulation of Glucose and Lipids. PLoS One 2016; 11:e0149344. [PMID: 26890260 PMCID: PMC4758736 DOI: 10.1371/journal.pone.0149344] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 01/29/2016] [Indexed: 01/23/2023] Open
Abstract
There are unprecedented epidemics of obesity, such as type II diabetes and non-alcoholic fatty liver diseases (NAFLD) in developed countries. A concerning percentage of American children are being affected by obesity and NAFLD. Studies have suggested that the maternal environment in utero might play a role in the development of these diseases later in life. In this study, we documented that inhibiting SIRT1 signaling in human fetal hepatocytes rapidly led to an increase in intracellular glucose and lipids levels. More importantly, both de novo lipogenesis and gluconeogenesis related genes were upregulated upon SIRT1 inhibition. The AKT/FOXO1 pathway, a major negative regulator of gluconeogenesis, was decreased in the human fetal hepatocytes inhibited for SIRT1, consistent with the higher level of gluconeogenesis. These results indicate that SIRT1 is an important regulator of lipid and carbohydrate metabolisms within human fetal hepatocytes, acting as an adaptive transcriptional response to environmental changes.
Collapse
Affiliation(s)
- Takamasa Tobita
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jorge Guzman-Lepe
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Kazuki Takeishi
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Toshimasa Nakao
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Yang Wang
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Hepatobiliary Surgery, Peking University People’s Hospital, Beijing, China
| | - Fanying Meng
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Chu-Xia Deng
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, China
| | - Alexandra Collin de l’Hortet
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (ACH); (ASG)
| | - Alejandro Soto-Gutierrez
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Thomas E Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- McGowan Institute for Regenerative Medicine; Pittsburgh, Pennsylvania, United States of America
- * E-mail: (ACH); (ASG)
| |
Collapse
|
185
|
Griffiths PS, Walton C, Samsell L, Perez MK, Piedimonte G. Maternal high-fat hypercaloric diet during pregnancy results in persistent metabolic and respiratory abnormalities in offspring. Pediatr Res 2016; 79:278-86. [PMID: 26539661 PMCID: PMC4829879 DOI: 10.1038/pr.2015.226] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 08/08/2015] [Indexed: 01/21/2023]
Abstract
BACKGROUND We have shown in a previous population-based study significant correlation between childhood asthma and early abnormalities of lipid and glucose metabolism. This study's specific aim was to determine whether maternal nutrition in pregnancy affects postnatal metabolic and respiratory outcomes in the offspring. METHODS On gestation day 1, dams were switched from standard chow to either high-fat hypercaloric diet or control diet. Terminal experiments were performed on newborn and weanling offspring of dams fed the study diet during gestation and lactation, and on adult offspring maintained on the same diet as their mother. RESULTS Pups born from high-fat hypercaloric diet (HFD) dams developed metabolic abnormalities persistent throughout development. Cytokine expression analysis of lung tissues from newborns born to HFD dams revealed a strong proinflammatory pattern. Gene expression of neurotrophic factors and receptors was upregulated in lungs of weanlings born to HFD dams, and this was associated to higher respiratory system resistance and lower compliance at baseline, as well as hyperreactivity to aerosolized methacholine. Furthermore, HFD dams delivered pups prone to develop more severe disease after respiratory syncytial virus (RSV) infection. CONCLUSION Maternal nutrition in pregnancy is a critical determinant of airway inflammation and hyperreactivity in offspring and also increases risk for bronchiolitis independent from prepregnancy nutrition.
Collapse
Affiliation(s)
- Pamela S Griffiths
- Department of Pediatrics, West Virginia University, Morgantown, West Virginia
| | - Cheryl Walton
- Department of Pediatrics, West Virginia University, Morgantown, West Virginia
| | - Lennie Samsell
- Department of Pediatrics, West Virginia University, Morgantown, West Virginia
| | - Miriam K Perez
- Department of Community Pediatrics, The Cleveland Clinic, Cleveland, Ohio
| | | |
Collapse
|
186
|
Hernandez TL, Van Pelt RE, Anderson MA, Reece MS, Reynolds RM, de la Houssaye BA, Heerwagen M, Donahoo WT, Daniels LJ, Chartier-Logan C, Janssen RC, Friedman JE, Barbour LA. Women With Gestational Diabetes Mellitus Randomized to a Higher-Complex Carbohydrate/Low-Fat Diet Manifest Lower Adipose Tissue Insulin Resistance, Inflammation, Glucose, and Free Fatty Acids: A Pilot Study. Diabetes Care 2016; 39. [PMID: 26223240 PMCID: PMC4686845 DOI: 10.2337/dc15-0515] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Diet therapy in gestational diabetes mellitus (GDM) has focused on carbohydrate restriction but is poorly substantiated. In this pilot randomized clinical trial, we challenged the conventional low-carbohydrate/higher-fat (LC/CONV) diet, hypothesizing that a higher-complex carbohydrate/lower-fat (CHOICE) diet would improve maternal insulin resistance (IR), adipose tissue (AT) lipolysis, and infant adiposity. RESEARCH DESIGN AND METHODS At 31 weeks, 12 diet-controlled overweight/obese women with GDM were randomized to an isocaloric LC/CONV (40% carbohydrate/45% fat/15% protein; n = 6) or CHOICE (60%/25%/15%; n = 6) diet. All meals were provided. AT was biopsied at 37 weeks. RESULTS After ∼7 weeks, fasting glucose (P = 0.03) and free fatty acids (P = 0.06) decreased on CHOICE, whereas fasting glucose increased on LC/CONV (P = 0.03). Insulin suppression of AT lipolysis was improved on CHOICE versus LC/CONV (56 vs. 31%, P = 0.005), consistent with improved IR. AT expression of multiple proinflammatory genes was lower on CHOICE (P < 0.01). Infant adiposity trended lower with CHOICE (10.1 ± 1.4 vs. 12.6 ± 2%, respectively). CONCLUSIONS A CHOICE diet may improve maternal IR and infant adiposity, challenging recommendations for a LC/CONV diet.
Collapse
Affiliation(s)
- Teri L Hernandez
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO College of Nursing, University of Colorado, Anschutz Medical Campus, Aurora, CO Center for Women's Health Research, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
| | - Rachael E Van Pelt
- Division of Geriatric Medicine, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
| | - Molly A Anderson
- Division of Geriatric Medicine, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
| | - Melanie S Reece
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
| | - Regina M Reynolds
- Division of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
| | - Becky A de la Houssaye
- Division of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
| | - Margaret Heerwagen
- Division of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
| | - William T Donahoo
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO Kaiser Permanente Colorado, Denver, CO
| | | | - Catherine Chartier-Logan
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
| | - Rachel C Janssen
- Division of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
| | - Jacob E Friedman
- Division of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
| | - Linda A Barbour
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
187
|
Boone-Heinonen J, Messer LC, Fortmann SP, Wallack L, Thornburg KL. From fatalism to mitigation: A conceptual framework for mitigating fetal programming of chronic disease by maternal obesity. Prev Med 2015; 81:451-9. [PMID: 26522092 PMCID: PMC4679670 DOI: 10.1016/j.ypmed.2015.10.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 10/21/2015] [Accepted: 10/23/2015] [Indexed: 02/07/2023]
Abstract
Prenatal development is recognized as a critical period in the etiology of obesity and cardiometabolic disease. Potential strategies to reduce maternal obesity-induced risk later in life have been largely overlooked. In this paper, we first propose a conceptual framework for the role of public health and preventive medicine in mitigating the effects of fetal programming. Second, we review a small but growing body of research (through August 2015) that examines interactive effects of maternal obesity and two public health foci - diet and physical activity - in the offspring. Results of the review support the hypothesis that diet and physical activity after early life can attenuate disease susceptibility induced by maternal obesity, but human evidence is scant. Based on the review, we identify major gaps relevant for prevention research, such as characterizing the type and dose response of dietary and physical activity exposures that modify the adverse effects of maternal obesity in the offspring. Third, we discuss potential implications of interactions between maternal obesity and postnatal dietary and physical activity exposures for interventions to mitigate maternal obesity-induced risk among children. Our conceptual framework, evidence review, and future research directions offer a platform to develop, test, and implement fetal programming mitigation strategies for the current and future generations of children.
Collapse
Affiliation(s)
| | - Lynne C Messer
- School of Community Health, College of Urban and Public Affairs, Portland State University, Portland, OR, USA
| | | | - Lawrence Wallack
- School of Community Health, College of Urban and Public Affairs, Portland State University, Portland, OR, USA
| | - Kent L Thornburg
- Bob and Charlee Moore Institute for Nutrition and Wellness, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
188
|
Fu Q, Olson P, Rasmussen D, Keith B, Williamson M, Zhang KK, Xie L. A short-term transition from a high-fat diet to a normal-fat diet before pregnancy exacerbates female mouse offspring obesity. Int J Obes (Lond) 2015; 40:564-72. [PMID: 26607040 DOI: 10.1038/ijo.2015.236] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 10/19/2015] [Accepted: 11/01/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND/OBJECTIVES Recent findings have highlighted the detrimental influence of maternal overnutrition and obesity on fetal development and early life development. However, there are no evidence-based guidelines regarding the optimal strategy for dietary intervention before pregnancy. SUBJECTS/METHODS We used a murine model to study whether switching from a high-fat (HF) diet to a normal-fat (NF) diet (H1N group) 1 week before pregnancy could lead to in utero reprogramming of female offspring obesity; comparator groups were offspring given a consistent maternal HF group or NF group until weaning. After weaning, all female offspring were given the HF diet for either 9 or 12 weeks before being killed humanely. RESULTS H1N treatment did not result in maternal weight loss before pregnancy. NF offsprings were neither obese nor glucose intolerant during the entire experimental period. H1N offsprings were most obese after the 12-week postweaning HF diet and displayed glucose intolerance earlier than HF offsprings. Our mechanistic study showed reduced adipocyte insulin receptor substrate 1 (IRS1) and hepatic IRS2 expression and increased adipocyte p-Ser(636/639) and p-Ser(612) of H1N or HF offspring compared with that in the NF offspring. Among all groups, the H1N offspring had lowest level of IRS1 and the highest levels of p-Ser(636/639) and p-Ser(612) in gonadal adipocyte. In addition, the H1N offspring further reduced the expression of Glut4 and Glut2, vs those of the HF offspring, which was lower compared with the NF offspring. There were also enhanced expression of genes inhibiting glycogenesis and decreased hepatic glycogen in H1N vs HF or NF offspring. Furthermore, we showed extremely higher expression of lipogenesis and adipogenesis genes in gonadal adipocytes of H1N offspring compared with all other groups. CONCLUSIONS Our results suggest that a transition from an HF diet to an NF diet shortly before pregnancy, without resulting in maternal weight loss, is not necessarily beneficial and may have deleterious effects on offspring.
Collapse
Affiliation(s)
- Q Fu
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA.,Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| | - P Olson
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - D Rasmussen
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - B Keith
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - M Williamson
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - K K Zhang
- Department of Pathology, University of North Dakota, Grand Forks, ND, USA
| | - L Xie
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA.,Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| |
Collapse
|
189
|
Nathanielsz PW, Yan J, Green R, Nijland M, Miller JW, Wu G, McDonald TJ, Caudill MA. Maternal obesity disrupts the methionine cycle in baboon pregnancy. Physiol Rep 2015; 3:e12564. [PMID: 26537341 PMCID: PMC4673623 DOI: 10.14814/phy2.12564] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 09/04/2015] [Accepted: 09/11/2015] [Indexed: 02/06/2023] Open
Abstract
Maternal intake of dietary methyl-micronutrients (e.g. folate, choline, betaine and vitamin B-12) during pregnancy is essential for normal maternal and fetal methionine metabolism, and is critical for important metabolic processes including those involved in developmental programming. Maternal obesity and nutrient excess during pregnancy influence developmental programming potentially predisposing adult offspring to a variety of chronic health problems. In the present study, we hypothesized that maternal obesity would dysregulate the maternal and fetal methionine cycle. To test this hypothesis, we developed a nulliparous baboon obesity model fed a high fat, high energy diet (HF-HED) prior to and during gestation, and examined methionine cycle biomarkers (e.g., circulating concentrations of homocysteine, methionine, choline, betaine, key amino acids, folate, and vitamin B-12). Animals were group housed allowing full physical activity and social interaction. Maternal prepregnancy percent body fat was 5% in controls and 19% in HF-HED mothers, while fetal weight was 16% lower in offspring of HF-HED mothers at term. Maternal and fetal homocysteine were higher, while maternal and fetal vitamin B-12 and betaine were lower in the HF-HED group. Elevations in circulating maternal folate were evident in the HF-HED group indicating impaired folate metabolism (methyl-trap) as a consequence of maternal vitamin B-12 depletion. Finally, fetal methionine, glycine, serine, and taurine were lower in the HF-HED fetuses. These data show that maternal obesity disturbs the methionine cycle in primate pregnancy, providing a mechanism for the epigenetic changes observed among obese pregnant women and suggesting diagnostic and therapeutic opportunities in human pregnancies complicated by obesity.
Collapse
Affiliation(s)
- Peter W Nathanielsz
- Center for Pregnancy and Newborn Research, Department OB/GYN, University of Texas Health Science Center San Antonio, San Antonio, Texas Texas Center for Pregnancy and Life Course Health, Southwest National Primate Research Institute Texas Biomedical Research Institute, San Antonio, Texas
| | - Jian Yan
- Division of Nutritional Sciences, Savage Hall, Ithaca, New York
| | - Ralph Green
- Department of Medical Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, California
| | - Mark Nijland
- Center for Pregnancy and Newborn Research, Department OB/GYN, University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - Joshua W Miller
- Department of Medical Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, California Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey
| | - Guoyao Wu
- Department of Animal Science and Center for Animal Biotechnology and Genomics, and Faculty of Nutrition, Texas A&M University, College Station, Texas
| | - Thomas J McDonald
- Center for Pregnancy and Newborn Research, Department OB/GYN, University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - Marie A Caudill
- Division of Nutritional Sciences, Savage Hall, Ithaca, New York
| |
Collapse
|
190
|
Rivera HM, Kievit P, Kirigiti MA, Bauman LA, Baquero K, Blundell P, Dean TA, Valleau JC, Takahashi DL, Frazee T, Douville L, Majer J, Smith MS, Grove KL, Sullivan EL. Maternal high-fat diet and obesity impact palatable food intake and dopamine signaling in nonhuman primate offspring. Obesity (Silver Spring) 2015; 23:2157-64. [PMID: 26530932 PMCID: PMC4636015 DOI: 10.1002/oby.21306] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/15/2015] [Accepted: 07/31/2015] [Indexed: 01/31/2023]
Abstract
OBJECTIVE To utilize a nonhuman primate model to examine the impact of maternal high-fat diet (HFD) consumption and pre-pregnancy obesity on offspring intake of palatable food and to examine whether maternal HFD consumption impaired development of the dopamine system, critical for the regulation of hedonic feeding. METHODS The impact of exposure to maternal HFD and obesity on offspring consumption of diets of varying composition was assessed after weaning. The influence of maternal HFD consumption on the development of the prefrontal cortex-dopaminergic system at 13 months of age was also examined. RESULTS During a preference test, offspring exposed to maternal HFD consumption and obesity displayed increased intake of food high in fat and sugar content relative to offspring from lean control mothers. Maternal HFD consumption suppressed offspring dopamine signaling (as assessed by immunohistochemistry) relative to control offspring. Specifically, there was decreased abundance of dopamine fibers and of dopamine receptor 1 and 2 proteins. CONCLUSIONS This study reveals that offspring exposed to both maternal HFD consumption and maternal obesity during early development are at increased risk for obesity due to overconsumption of palatable energy-dense food, a behavior that may be related to reduced central dopamine signaling.
Collapse
Affiliation(s)
- Heidi M. Rivera
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR 97006, United States
| | - Paul Kievit
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR 97006, United States
| | - Melissa A. Kirigiti
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR 97006, United States
| | - Leigh Ann Bauman
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR 97006, United States
| | - Karalee Baquero
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR 97006, United States
| | - Peter Blundell
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR 97006, United States
| | - Tyler A. Dean
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR 97006, United States
| | - Jeanette C. Valleau
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR 97006, United States
| | - Diana L. Takahashi
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR 97006, United States
| | - Tim Frazee
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR 97006, United States
| | - Luke Douville
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR 97006, United States
- Department of Biology, University of Portland, Portland, OR, 97203, United States
| | - Jordan Majer
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR 97006, United States
| | - M. Susan Smith
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR 97006, United States
| | - Kevin L. Grove
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR 97006, United States
| | - Elinor L. Sullivan
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR 97006, United States
- Department of Biology, University of Portland, Portland, OR, 97203, United States
| |
Collapse
|
191
|
Astbury S, Mostyn A, Symonds ME, Bell RC. Nutrient availability, the microbiome, and intestinal transport during pregnancy. Appl Physiol Nutr Metab 2015; 40:1100-6. [DOI: 10.1139/apnm-2015-0117] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adequate adaptation of the gastrointestinal tract is important during pregnancy to ensure that the increased metabolic demands by the developing fetus are met. These include changes in surface area mediated by villus hypertrophy and enhanced functional capacity of individual nutrient receptors, including those transporting glucose, fructose, leucine, and calcium. These processes are regulated either by the enhanced nutrient demand or are facilitated by changes in the secretion of pregnancy hormones. Our review also covers recent research into the microbiome, and how pregnancy could lead to microbial adaptations, which are beneficial to the mother, yet are also similar to those seen in the metabolic syndrome. The potential role of diet in modulating the microbiome during pregnancy, as well as the potential for the intestinal microbiota to induce pregnancy complications, are examined. Gaps in the current literature are highlighted, including those where only historical evidence is available, and we suggest areas that should be a priority for further research. In summary, although a significant degree of adaptation has been described, there are both well-established processes and more recent discoveries, such as changes within the maternal microbiome, that pose new questions as to how the gastrointestinal tract effectively adapts to pregnancy, especially in conjunction with maternal obesity.
Collapse
Affiliation(s)
- Stuart Astbury
- Department of Agricultural, Food and Nutritional Science, Human Nutrition, Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Child Health, Obstetrics and Gynaecology, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| | - Alison Mostyn
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Leicestershire LE12 5RD, UK
| | - Michael E. Symonds
- Child Health, Obstetrics and Gynaecology, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| | - Rhonda C. Bell
- Department of Agricultural, Food and Nutritional Science, Human Nutrition, Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
| |
Collapse
|
192
|
Díaz P, Harris J, Rosario FJ, Powell TL, Jansson T. Increased placental fatty acid transporter 6 and binding protein 3 expression and fetal liver lipid accumulation in a mouse model of obesity in pregnancy. Am J Physiol Regul Integr Comp Physiol 2015; 309:R1569-77. [PMID: 26491104 DOI: 10.1152/ajpregu.00385.2015] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 10/19/2015] [Indexed: 02/07/2023]
Abstract
Obesity in pregnancy is associated with increased fetal growth and adiposity, which, in part, is determined by transplacental nutrient supply. Trophoblast uptake and intracellular trafficking of lipids are dependent on placental fatty acid transport proteins (FATP), translocase (FAT/CD36), and fatty acid binding proteins (FABP). We hypothesized that maternal obesity in mice leads to increased placental expression of FAT/CD36, FATPs, and FABPs, and lipid accumulation in the fetal liver. C57/BL6J female mice were fed either a control (C; n = 10) or an obesogenic (OB; n = 10) high-fat, high-sugar diet before mating and throughout pregnancy. At E18.5, placentas and fetal livers were collected. Trophoblast plasma membranes (TPM) were isolated from placental homogenates. Expression of FAT/CD36 and FATP (TPM) and FABP (homogenates) was determined by immunoblotting. Gene expression was assessed by RT-quantitative PCR. Sections of fetal livers were stained for Oil Red O, and lipid droplets were quantified. TPM protein expression of FAT/CD36, FATP 2, and FATP 4 was comparable between C and OB groups. Conversely, TPM FATP 6 expression was increased by 35% in OB compared with C placentas without changes in mRNA expression. FABPs 1, 3-5 and PPARγ were expressed in homogenates, and FABP 3 expression increased 27% in OB compared with C placentas; however, no changes were observed in mRNA expression. Lipid droplet accumulation was 10-fold higher in the livers of fetuses from OB compared with C group. We propose that increased lipid transport capacity in obese mice promotes transplacental fatty acid transport and contributes to excess lipid accumulation in the fetal liver.
Collapse
Affiliation(s)
- Paula Díaz
- Department of Pediatrics, Section of Neonatology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado; Department of Obstetrics and Gynecology, University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - Jessica Harris
- Department of Obstetrics and Gynecology, University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - Fredrick J Rosario
- Department of Obstetrics and Gynecology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado; and
| | - Theresa L Powell
- Department of Pediatrics, Section of Neonatology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado; Department of Obstetrics and Gynecology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado; and
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado; and
| |
Collapse
|
193
|
Kabaran S, Besler HT. Do fatty acids affect fetal programming? JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2015; 33:14. [PMID: 26825664 PMCID: PMC5025983 DOI: 10.1186/s41043-015-0018-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 06/26/2015] [Indexed: 05/29/2023]
Abstract
BACKGROUND In this study discussed the primary and regulatory roles of fatty acids, and investigated the affects of fatty acids on metabolic programming. METHODS Review of the literature was carried out on three electronic databases to assess the roles of fatty acids in metabolic programming. All abstracts and full-text articles were examined, and the most relevant articles were selected for screening and inclusion in this review. RESULTS The mother's nutritional environment during fetal period has important effects on long term health. Fatty acids play a primary role in growth and development. Alterations in fatty acid intake in the fetal period may increase the risk of obesity and metabolic disorders in later life. Maternal fatty acid intakes during pregnancy and lactation are passed to the fetus and the newborn via the placenta and breast milk, respectively. Imbalances in fatty acid intake during the fetal period change the fatty acid composition of membrane phospholipids, which can cause structural and functional problems in cells. Additionally, the metabolic and neuroendocrine environments of the fetus and the newborn play key roles in the regulation of energy balance. CONCLUSIONS Imbalances in fatty acid intake during pregnancy and lactation may result in permanent changes in appetite control, neuroendocrine function and energy metabolism in the fetus, leading to metabolic programming. Further studies are needed to determine the role of fatty acid intake in metabolic programming.
Collapse
Affiliation(s)
- Seray Kabaran
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Eastern Mediterranean University, Famagusta, T.R. North Cyprus via Mersin 10, Turkey.
| | - H Tanju Besler
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Samanpazarı/Ankara, Turkey
| |
Collapse
|
194
|
Friedman JE. Obesity and Gestational Diabetes Mellitus Pathways for Programming in Mouse, Monkey, and Man—Where Do We Go Next? The 2014 Norbert Freinkel Award Lecture. Diabetes Care 2015; 38. [PMID: 26207051 PMCID: PMC4512131 DOI: 10.2337/dc15-0628] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity and gestational diabetes mellitus continue to increase worldwide and span the spectrum of age, race, ethnicity, and socioeconomic status. Alarmingly, 1 in 10 infants and toddlers is obese, and 1 in 5 youths is both obese and at risk for metabolic syndrome prior to puberty. The mechanisms underlying how poor maternal health imparts risk for future metabolic disease in the offspring are beginning to emerge in deeply phenotyped human and nonhuman primate models. Maternal diet and obesity impact fuels, hormones, and inflammation with powerful effects on fetal metabolic systems. These are accompanied by persistent changes in the infant microbiome and epigenome and in offspring behavior. These results suggest that gestational and lactational dietary exposures are driving health risks in the next generation. Whether maternal diet can prevent changes in the womb to alter infant life-course disease risk is still unknown. Controlled, mechanistic studies to identify interventions are sorely needed for a healthier next generation.
Collapse
Affiliation(s)
- Jacob E Friedman
- Departments of Pediatrics, Biochemistry and Molecular Genetics, Endocrinology, Metabolism & Diabetes, and Basic Reproductive Sciences, University of Colorado School of Medicine; Colorado Program for Nutrition and Healthy Development, Children's Hospital Colorado Research Institute; University of Colorado Nutrition and Obesity Research Center, Aurora, CO
| |
Collapse
|
195
|
O'Tierney-Ginn P, Roberts V, Gillingham M, Walker J, Glazebrook PA, Thornburg KL, Grove K, Frias AE. Influence of high fat diet and resveratrol supplementation on placental fatty acid uptake in the Japanese macaque. Placenta 2015; 36:903-10. [PMID: 26145226 PMCID: PMC4529757 DOI: 10.1016/j.placenta.2015.06.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 05/27/2015] [Accepted: 06/05/2015] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Adequate maternal supply and placental delivery of long chain polyunsaturated fatty acids (LCPUFA) is essential for normal fetal development. In humans, maternal obesity alters placental FA uptake, though the impact of diet remains uncertain. The fatty fetal liver observed in offspring of Japanese macaques fed a high fat diet (HFD) was prevented with resveratrol supplementation during pregnancy. We sought to determine the effect of HFD and resveratrol, a supplement with insulin-sensitizing properties, on placental LCPUFA uptake in this model. METHODS J. macaques were fed control chow (15% fat, n = 5), HFD (35% fat, n = 10) or HFD containing 0.37% resveratrol (n = 5) prior to- and throughout pregnancy. At ∼ 130 d gestation (term = 173 d), placentas were collected by caesarean section. Fatty acid uptake studies using (14)C-labeled oleic acid, arachidonic acid (AA) and docosahexanoic acid (DHA) were performed in placental explants. RESULTS Resveratrol supplementation increased placental uptake of DHA (P < 0.05), while HFD alone had no measurable effect. Resveratrol increased AMP-activated protein kinase activity and mRNA expression of the fatty acid transporters FATP-4, CD36 and FABPpm (P < 0.05). Placental DHA content was decreased in HFD dams; resveratrol had no effect on tissue fatty acid profiles. DISCUSSION Maternal HFD did not significantly affect placental LCPUFA uptake. Furthermore, resveratrol stimulated placental DHA uptake capacity, AMPK activation and transporter expression. Placental handling of DHA is particularly sensitive to the dramatic alterations in the maternal metabolic phenotype and placental AMPK activity associated with resveratrol supplementation.
Collapse
Affiliation(s)
- P O'Tierney-Ginn
- Center for Reproductive Health, Case Western Reserve University at MetroHealth Medical Center, Cleveland, OH, USA.
| | - V Roberts
- Div of Diabetes, Obesity & Metabolism, Oregon National Primate Research Center, Beaverton, OR, USA
| | - M Gillingham
- Department of Nutrition, Oregon Health & Science University, Portland, OR, USA
| | - J Walker
- Div of Diabetes, Obesity & Metabolism, Oregon National Primate Research Center, Beaverton, OR, USA
| | - P A Glazebrook
- Center for Reproductive Health, Case Western Reserve University at MetroHealth Medical Center, Cleveland, OH, USA
| | - K L Thornburg
- Center for Developmental Health, Oregon Health & Science University, Portland, OR, USA
| | - K Grove
- Div of Diabetes, Obesity & Metabolism, Oregon National Primate Research Center, Beaverton, OR, USA
| | - A E Frias
- Div of Diabetes, Obesity & Metabolism, Oregon National Primate Research Center, Beaverton, OR, USA; Center for Developmental Health, Oregon Health & Science University, Portland, OR, USA; Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
196
|
Oh KY, Roberts VHJ, Schabel MC, Grove KL, Woods M, Frias AE. Gadolinium Chelate Contrast Material in Pregnancy: Fetal Biodistribution in the Nonhuman Primate. Radiology 2015; 276:110-8. [PMID: 25763829 PMCID: PMC4485748 DOI: 10.1148/radiol.15141488] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE To determine the extent to which gadolinium chelate is found in nonhuman primate fetal tissues and amniotic fluid at 19-45 hours after intravenous injection of a weight-appropriate maternal dose of the contrast agent gadoteridol. MATERIALS AND METHODS Gravid Japanese macaques (n = 14) were maintained as approved by the institutional animal care and utilization committee. In the 3rd trimester of pregnancy, the macaques were injected with gadoteridol (0.1 mmol per kilogram of maternal weight). Fetuses were delivered by means of cesarean section within 24 hours of maternal injection (range, 19-21 hours; n = 11) or 45 hours after injection (n = 3). Gadolinium chelate levels in the placenta, fetal tissues, and amniotic fluid were obtained by using inductively coupled plasma mass spectrometry. The Wilcoxon rank sum test was used for quantitative comparisons. RESULTS Gadoteridol was present in the fetoplacental circulation at much lower quantities than in the mother. At both time points, the distribution of gadolinium chelate in the fetus was comparable to that expected in an adult. The highest concentration of the injected dose (ID) was found in the fetal kidney (0.0161% ID per gram in the 19-21-hour group). The majority of the in utero gadolinium chelate was found in the amniotic fluid and the placenta (mean, 0.1361% ID per organ ± 0.076 [standard deviation] and 0.0939% ID per organ ± 0.0494, respectively). Data acquired 45 hours after injection showed a significant decrease in the gadolinium chelate concentration in amniotic fluid compared with that in the 19-21-hour group (from 0.0017% to 0.0007% ID per gram; P = .01). CONCLUSION Amounts of gadolinium chelate in the fetal tissues and amniotic fluid were minimal compared with the maternal ID. This may impact future clinical studies on the safety of gadolinium contrast agent use in pregnancy.
Collapse
Affiliation(s)
- Karen Y. Oh
- From the Department of Radiology (K.Y.O.), Advanced Imaging Research Center (M.C.S., M.W.), and Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine (A.E.F.), Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, MC L340, Portland, OR 97239; Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Ore (V.H.J.R., K.L.G., A.E.F.); and Department of Chemistry, Portland State University, Portland, Ore (M.W.)
| | - Victoria H. J. Roberts
- From the Department of Radiology (K.Y.O.), Advanced Imaging Research Center (M.C.S., M.W.), and Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine (A.E.F.), Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, MC L340, Portland, OR 97239; Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Ore (V.H.J.R., K.L.G., A.E.F.); and Department of Chemistry, Portland State University, Portland, Ore (M.W.)
| | - Matthias C. Schabel
- From the Department of Radiology (K.Y.O.), Advanced Imaging Research Center (M.C.S., M.W.), and Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine (A.E.F.), Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, MC L340, Portland, OR 97239; Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Ore (V.H.J.R., K.L.G., A.E.F.); and Department of Chemistry, Portland State University, Portland, Ore (M.W.)
| | - Kevin L. Grove
- From the Department of Radiology (K.Y.O.), Advanced Imaging Research Center (M.C.S., M.W.), and Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine (A.E.F.), Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, MC L340, Portland, OR 97239; Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Ore (V.H.J.R., K.L.G., A.E.F.); and Department of Chemistry, Portland State University, Portland, Ore (M.W.)
| | - Mark Woods
- From the Department of Radiology (K.Y.O.), Advanced Imaging Research Center (M.C.S., M.W.), and Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine (A.E.F.), Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, MC L340, Portland, OR 97239; Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Ore (V.H.J.R., K.L.G., A.E.F.); and Department of Chemistry, Portland State University, Portland, Ore (M.W.)
| | - Antonio E. Frias
- From the Department of Radiology (K.Y.O.), Advanced Imaging Research Center (M.C.S., M.W.), and Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine (A.E.F.), Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, MC L340, Portland, OR 97239; Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Ore (V.H.J.R., K.L.G., A.E.F.); and Department of Chemistry, Portland State University, Portland, Ore (M.W.)
| |
Collapse
|
197
|
Rivera HM, Christiansen KJ, Sullivan EL. The role of maternal obesity in the risk of neuropsychiatric disorders. Front Neurosci 2015; 9:194. [PMID: 26150767 PMCID: PMC4471351 DOI: 10.3389/fnins.2015.00194] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 05/16/2015] [Indexed: 12/22/2022] Open
Abstract
Recent evidence indicates that perinatal exposure to maternal obesity, metabolic disease, including diabetes and hypertension, and unhealthy maternal diet has a long-term impact on offspring behavior and physiology. During the past three decades, the prevalence of both obesity and neuropsychiatric disorders has rapidly increased. Epidemiologic studies provide evidence that maternal obesity and metabolic complications increase the risk of attention deficit hyperactivity disorder (ADHD), autism spectrum disorders, anxiety, depression, schizophrenia, eating disorders (food addiction, anorexia nervosa, and bulimia nervosa), and impairments in cognition in offspring. Animal models of maternal high-fat diet (HFD) induced obesity also document persistent changes in offspring behavior and impairments in critical neural circuitry. Animals exposed to maternal obesity and HFD consumption display hyperactivity, impairments in social behavior, increased anxiety-like and depressive-like behaviors, substance addiction, food addiction, and diminished cognition. During development, these offspring are exposed to elevated levels of nutrients (fatty acids, glucose), hormones (leptin, insulin), and inflammatory factors (C-reactive protein, interleukin, and tumor necrosis factor). Such factors appear to permanently change neuroendocrine regulation and brain development in offspring. In addition, inflammation of the offspring brain during gestation impairs the development of neural pathways critical in the regulation of behavior, such as serotoninergic, dopaminergic, and melanocortinergic systems. Dysregulation of these circuits increases the risk of mental health disorders. Given the high rates of obesity in most developed nations, it is critical that the mechanisms by which maternal obesity programs offspring behavior are thoroughly characterized. Such knowledge will be critical in the development of preventative strategies and therapeutic interventions.
Collapse
Affiliation(s)
- Heidi M Rivera
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR, USA
| | | | - Elinor L Sullivan
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center Beaverton, OR, USA ; Department of Biology, University of Portland Portland, OR, USA
| |
Collapse
|
198
|
Developmental Programming of Nonalcoholic Fatty Liver Disease: The Effect of Early Life Nutrition on Susceptibility and Disease Severity in Later Life. BIOMED RESEARCH INTERNATIONAL 2015; 2015:437107. [PMID: 26090409 PMCID: PMC4450221 DOI: 10.1155/2015/437107] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/15/2015] [Indexed: 12/20/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is fast becoming the most common liver disease globally and parallels rising obesity rates. The developmental origins of health and disease hypothesis have linked alterations in the early life environment to an increased risk of metabolic disorders in later life. Altered early life nutrition, in addition to increasing risk for the development of obesity, type 2 diabetes, and cardiovascular disease in offspring, is now associated with an increased risk for the development of NAFLD. This review summarizes emerging research on the developmental programming of NAFLD by both maternal obesity and undernutrition with a particular focus on the possible mechanisms underlying the development of hepatic dysfunction and potential strategies for intervention.
Collapse
|
199
|
Epigenetic and transgenerational reprogramming of brain development. Nat Rev Neurosci 2015; 16:332-44. [PMID: 25921815 DOI: 10.1038/nrn3818] [Citation(s) in RCA: 330] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurodevelopmental programming - the implementation of the genetic and epigenetic blueprints that guide and coordinate normal brain development - requires tight regulation of transcriptional processes. During prenatal and postnatal time periods, epigenetic processes fine-tune neurodevelopment towards an end product that determines how an organism interacts with and responds to exposures and experiences throughout life. Epigenetic processes also have the ability to reprogramme the epigenome in response to environmental challenges, such as maternal stress, making the organism more or less adaptive depending on the future challenges presented. Epigenetic marks generated within germ cells as a result of environmental influences throughout life can also shape future generations long before conception occurs.
Collapse
|
200
|
Strakovsky RS, Wang H, Engeseth NJ, Flaws JA, Helferich WG, Pan YX, Lezmi S. Developmental bisphenol A (BPA) exposure leads to sex-specific modification of hepatic gene expression and epigenome at birth that may exacerbate high-fat diet-induced hepatic steatosis. Toxicol Appl Pharmacol 2015; 284:101-12. [PMID: 25748669 DOI: 10.1016/j.taap.2015.02.021] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 02/18/2015] [Accepted: 02/23/2015] [Indexed: 12/25/2022]
Abstract
Developmental bisphenol A (BPA) exposure increases adulthood hepatic steatosis with reduced mitochondrial function. To investigate the potential epigenetic mechanisms behind developmental BPA-induced hepatic steatosis, pregnant Sprague-Dawley rats were dosed with vehicle (oil) or BPA (100μg/kg/day) from gestational day 6 until postnatal day (PND) 21. After weaning, offspring were either challenged with a high-fat (HF; 45% fat) or remained on a control (C) diet until PND110. From PND60 to 90, both BPA and HF diet increased the fat/lean ratio in males only, and the combination of BPA and HF diet appeared to cause the highest ratio. On PND110, Oil-HF, BPA-C, and BPA-HF males had higher hepatic lipid accumulation than Oil-C, with microvesicular steatosis being marked in the BPA-HF group. Furthermore, on PND1, BPA increased and modified hepatic triglyceride (TG) and free fatty acid (FFA) compositions in males only. In PND1 males, BPA increased hepatic expression of FFA uptake gene Fat/Cd36, and decreased the expression of TG synthesis- and β-oxidation-related genes (Dgat, Agpat6, Cebpα, Cebpβ, Pck1, Acox1, Cpt1a, Cybb). BPA altered DNA methylation and histone marks (H3Ac, H4Ac, H3Me2K4, H3Me3K36), and decreased the binding of several transcription factors (Pol II, C/EBPβ, SREBP1) within the male Cpt1a gene, the key β-oxidation enzyme. In PND1 females, BPA only increased the expression of genes involved in FFA uptake and TG synthesis (Lpl, Fasn, and Dgat). These data suggest that developmental BPA exposure alters and reprograms hepatic β-oxidation capacity in males, potentially through the epigenetic regulation of genes, and further alters the response to a HF diet.
Collapse
Affiliation(s)
- Rita S Strakovsky
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, USA
| | - Huan Wang
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, USA
| | - Nicki J Engeseth
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, USA
| | - Jodi A Flaws
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, USA
| | - William G Helferich
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, USA
| | - Yuan-Xiang Pan
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, USA.
| | - Stéphane Lezmi
- Department of Pathobiology, University of Illinois Urbana-Champaign, USA.
| |
Collapse
|