1
|
Dayarathne LA, Ko SC, Yim MJ, Lee JM, Kim JY, Oh GW, Kim CH, Kim KW, Lee DS, Jung WK, Je JY. Purple Butter Clam ( Saxidomus Purpurata) as a Potential Functional Food Source for Obesity Treatment. J Med Food 2024; 27:1038-1049. [PMID: 39382491 DOI: 10.1089/jmf.2024.k.0169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024] Open
Abstract
Saxidomus purpurata extract (SPE) is a highly consumable seafood worldwide with known health-related benefits. However, there are no reports of its' anti-obesity effect. This study explores the potential of SPE for anti-obesity effects by modulating adipogenesis and lipolysis. SPE reduced intracellular lipid and triglyceride accumulation while increasing free glycerol release in adipocytes. SPE inhibited lipogenesis protein expressions and increased the phosphorylation of hormone-sensitive lipase and Adenosine monophosphate-activated protein kinase (AMPK) to promote lipolysis. In addition, SPE suppressed adipogenesis by downregulating protein expression of key adipogenic markers, peroxisome proliferator-activated receptor gamma (PPARγ), CCAAT/enhancer-binding protein α (C/EBPα), and sterol regulatory element-binding protein 1 (SREBP1) via Wnt/β-catenin signaling. SPE augmented the heme oxygenase-1 (HO-1) expression. Thus, pharmacological intervention with Zinc protoporphyrin (ZnPP-HO-1 antagonist) was employed to validate the HO-1 role. The presence of ZnPP increased the lipid accumulation and reduced the free glycerol release. At the molecular level, adipogenic transcription factors (PPARγ, C/EBPα, and SREBP1) expressions were restored in the presence of ZnPP. GC-MS analysis revealed that SPE was comprised of several fatty acids, contributing to its anti-obesity activity. SPE is an effective nutraceutical that can be used to reduce the progression of obesity. HO-1 expression during adipogenesis might be the mechanism of action for the anti-obesity effect of SPE.
Collapse
Affiliation(s)
- Lakshi A Dayarathne
- Department of Food and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Seok-Chun Ko
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Mi-Jin Yim
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Jeong Min Lee
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Ji-Yul Kim
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Gun-Woo Oh
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Chul Hwan Kim
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Kyung Woo Kim
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Dae-Sung Lee
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Won-Kyo Jung
- Major of Biomedical Engineering, Division of Smart Healthcare, Pukyong National University, Busan, Korea
| | - Jae-Young Je
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan, Korea
| |
Collapse
|
2
|
Lee YJ, Kim J, Kwon YH. Long-Term Effects of Maternal Fat Consumption on the Brain Transcriptome of Obesogenic Diet-Fed Young Adult Mice Offspring. J Nutr 2024; 154:1532-1539. [PMID: 38484978 DOI: 10.1016/j.tjnut.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/17/2024] [Accepted: 03/11/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Substantial evidence has demonstrated that maternal high-fat (HF) consumption during gestation and lactation plays as a risk factor for neurodevelopmental alterations and subsequent neurological disorders. OBJECTIVE We investigated the regulatory mechanisms of maternal fat consumption on brain development and function in offspring at different ages. METHODS Mouse dams were fed either a control diet [low-fat (LF)] or an HF diet for 3 wk before mating and throughout pregnancy and lactation. Offspring were killed at postnatal day (PD) 21 (LF21 and HF21), and the rest were fed an HF diet for 12 wk until the killing at PD 105 (LF105 and HF105). The expression levels of genes and proteins in the brains of offspring were analyzed by microarray and immunoblotting, respectively. RESULTS Maternal dietary fat content, offspring age, and their interaction affected the expression levels of 1215, 10,453, and 2105 genes, respectively. The 67 differentially expressed genes (DEGs) between the HF21 and LF21 groups were enriched in several Gene Ontology terms related to nervous system development. Among 45 DEGs of the HF105/LF105 comparison, several genes associated with neurotransmitter action are detected. In addition, we observed increased activation of the AMP-dependent protein kinase-cAMP response element binding protein signaling pathway in HF105/LF105 comparison. However, maternal fat content did not change the protein levels of amyloid-β and tau hyperphosphorylation, the markers of neuropathogenesis. CONCLUSIONS Maternal HF feeding altered the expression of genes involved in the development and neurotransmitter system in the brains of PD 21 and HF diet-fed PD 105 offspring, respectively. Especially, the absence of overlap between DEGs at each comparison highlights the dynamic nature of alterations in gene expression in offspring of dams fed an HF diet. Further investigation on older adult offspring is necessary to elucidate the effects of maternal fat intake on the brain pathophysiology of offspring.
Collapse
Affiliation(s)
- Youn Ji Lee
- Department of Food and Nutrition, Seoul National University, Seoul, Korea
| | - Juhae Kim
- Department of Food and Nutrition, Seoul National University, Seoul, Korea; Research Institute of Human Ecology, Seoul National University, Seoul, Korea
| | - Young Hye Kwon
- Department of Food and Nutrition, Seoul National University, Seoul, Korea; Research Institute of Human Ecology, Seoul National University, Seoul, Korea.
| |
Collapse
|
3
|
Arslan S, Yıldıran H, Seymen CM. The Effect of Maternal High-Fat Diet on Adipose Tissue Histology and Lipid Metabolism-Related Genes Expression in Offspring Rats. Nutrients 2024; 16:150. [PMID: 38201978 PMCID: PMC10780511 DOI: 10.3390/nu16010150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
The developing fetus is dependent on the maternal nutritional environment. This study was conducted to determine the effects of a maternal high-fat diet (HFD) applied during pregnancy and/or lactation on the expression levels of some lipid-related genes in rat models. Half of the pregnant rats (n: 6) were fed an HFD (energy from fat: 45%), while the other half (n: 6) were fed a control diet (CD) (energy from fat, 7.7%) during the pregnancy period. During lactation, dams in both groups were divided into two subgroups, with half fed the CD and the other half fed the HFD. Thus, four groups were obtained: CD-CD, CD-HFD, HFD-CD, and HFD-HFD. At the end of lactation, all mothers and half of the offspring were sacrificed. The remaining offspring were fed a CD for five weeks. The average birth weight of the CD group offspring was found to be lower than that of the HFD group (p < 0.05). The amount of adipose tissue was highest in CD-HFD (p < 0.05), while gene expression levels were similar between groups (p > 0.05), and the most degenerative histological changes were observed in the eight-week HFD-HFD (p < 0.05). This study suggests that maternal HFD during pregnancy and lactation may increase adiposity in offspring rats, especially during the weaning period.
Collapse
Affiliation(s)
- Sabriye Arslan
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara 06490, Turkey;
| | - Hilal Yıldıran
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara 06490, Turkey;
| | - Cemile Merve Seymen
- Department of Histology and Embryology, Faculty of Medicine, Gazi University, Ankara 06500, Turkey;
| |
Collapse
|
4
|
Cetin AK, Buyukdere Y, Gulec A, Akyol A. Taurine supplementation reduces adiposity and hepatic lipid metabolic activity in adult offspring following maternal cafeteria diet. Nutr Res 2023; 117:15-29. [PMID: 37423013 DOI: 10.1016/j.nutres.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 06/07/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023]
Abstract
Maternal taurine supplementation has been shown to exert protective effects following a maternal obesogenic diet on offspring growth and metabolism. However, the long-term effects of maternal cafeteria diet on adiposity, metabolic profile, and hepatic gene expression patterns following supplementation of taurine in adult offspring remains unclear. In this study, we hypothesized that exposure to maternal taurine supplementation would modulate the effects of maternal cafeteria diet by reducing adiposity and hepatic gene expression patterns involved in lipid metabolism in adult offspring. Female Wistar rats were fed a control diet, control diet supplemented with 1.5% taurine in drinking water, cafeteria diet (CAF) or CAF supplemented with taurine (CAFT) from weaning. After 8 weeks, all animals were mated and maintained on the same diets during pregnancy and lactation. After weaning, all offspring were fed with control chow diet until the age of 20 weeks. Despite similar body weights, CAFT offspring had significantly lower fat deposition and body fat when compared with CAF offspring. Microarray analysis revealed that genes (Akr1c3, Cyp7a1, Hsd17b6, Cd36, Acsm3, and Aldh1b1) related to steroid hormone biosynthesis, cholesterol metabolism, peroxisome proliferator-activated receptor signaling pathway, butanoate metabolism, and fatty acid degradation were down-regulated in CAFT offspring. The current study shows that exposure to maternal cafeteria diet promoted adiposity and taurine supplementation reduced lipid deposition and in both male and female offspring and led to alterations in hepatic gene expression patterns, reducing the detrimental effects of maternal cafeteria diet.
Collapse
Affiliation(s)
- Arzu Kabasakal Cetin
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, 06100 Sihhiye, Ankara, Turkey
| | - Yucel Buyukdere
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, 06100 Sihhiye, Ankara, Turkey
| | - Atila Gulec
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, 06100 Sihhiye, Ankara, Turkey
| | - Asli Akyol
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, 06100 Sihhiye, Ankara, Turkey.
| |
Collapse
|
5
|
Kabasakal Çetin A, Alkan Tuğ T, Güleç A, Akyol A. Effects of maternal taurine supplementation on maternal dietary intake, plasma metabolites and fetal growth and development in cafeteria diet fed rats. PeerJ 2021; 9:e11547. [PMID: 34141487 PMCID: PMC8180190 DOI: 10.7717/peerj.11547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/11/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Maternal obesity may disrupt the developmental process of the fetus during gestation in rats. Recent evidence suggests that taurine can exert protective role against detrimental influence of obesogenic diets. This study aimed to examine the effect of maternal cafeteria diet and/or taurine supplementation on maternal dietary intake, plasma metabolites, fetal growth and development. METHODS Female Wistar rats were fed a control diet (CON), CON supplemented with 1.5% taurine in drinking water (CONT), cafeteria diet (CAF) or CAF supplemented with taurine (CAFT) from weaning. After 8 weeks all animals were mated and maintained on the same diets during pregnancy and lactation. RESULTS Dietary intakes were significantly different between the groups. Both CAF and CAFT fed dams consumed less water in comparison to CON and CONT dams. Taurine supplementation only increased plasma taurine concentrations in CONT group. Maternal plasma adiponectin concentrations increased in CAF and CAFT fed dams compared to CON and CONT fed dams and there was no effect of taurine. Hyperleptinemia was observed in CAF fed dams but not in CAFT fed dams. Malondialdehyde was significantly increased only in CAF fed dams. Litter size, sex ratio and birth weight were similar between the groups. There was an increase in neonatal mortality in CONT group. DISCUSSION This study showed that maternal taurine supplementation exerted modest protective effects on cafeteria diet induced maternal obesity. The increased neonatal mortality in CONT neonates indicates possible detrimental effects of taurine supplementation in the setting of normal pregnancy. Therefore, future studies should investigate the optimal dose of taurine supplementation and long term potential effects on the offspring.
Collapse
Affiliation(s)
- Arzu Kabasakal Çetin
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara, Türkiye
| | - Tuǧba Alkan Tuğ
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara, Türkiye
| | - Atila Güleç
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara, Türkiye
| | - Aslı Akyol
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara, Türkiye
| |
Collapse
|
6
|
Benyoub N, Merzouk H, Merzouk AS, Ghorzi H. Changes in metabolic parameters in growing male rats exposed to 10% and 30% sucrose drinking. NUTR CLIN METAB 2021. [DOI: 10.1016/j.nupar.2020.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
7
|
Offspring of obese mice display enhanced intake and sensitivity for palatable stimuli, with altered expression of taste signaling elements. Sci Rep 2020; 10:12776. [PMID: 32728024 PMCID: PMC7391633 DOI: 10.1038/s41598-020-68216-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 06/11/2020] [Indexed: 01/20/2023] Open
Abstract
Maternal body mass index and gestational weight gain predict future obesity status of the offspring. In studies of both rodents and non-human primates, maternal obesity also predicts a preference for palatable foods in the offspring. In this study, we used C57BL/6J mice to investigate whether an underlying cause for an increase in palatable food consumption in the offspring of obese mice was a change in taste function. Adult female mice were fed a normal chow (NC) or a high fat diet (HFD) for 5 weeks before mating, then also during the gestation (3 weeks) and lactation (3 weeks) periods, with offspring always maintained on a normal chow diet; thus the only experience offspring had with high fat food was via maternal exposure. Offspring exhibited similar weight, blood glucose levels and baseline water and chow intake in adulthood. Taste response was assessed after reaching maturity, using brief-access taste testing, with female offspring of obese dams showing an enhanced response to sucrose, and both sexes consuming more sucrose, sucralose and high fat diet if from obese mothers. Offspring also exhibited increased taste bud expression of mRNA for sweet receptor subunits T1R (Taste receptor type) 2 and 3, as well as other markers associated with taste signaling. Taste morphology in both groups appeared similar. Results indicate that obesity in the mother may lead to unhealthy feeding behavior in the offspring, correlating with altered expression of taste signaling elements, which likely drive increased avidity for palatable foods.
Collapse
|
8
|
Loche E, Blackmore HL, Carpenter AA, Beeson JH, Pinnock A, Ashmore TJ, Aiken CE, de Almeida-Faria J, Schoonejans JM, Giussani DA, Fernandez-Twinn DS, Ozanne SE. Maternal diet-induced obesity programmes cardiac dysfunction in male mice independently of post-weaning diet. Cardiovasc Res 2019; 114:1372-1384. [PMID: 29635288 PMCID: PMC6054211 DOI: 10.1093/cvr/cvy082] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 04/03/2018] [Indexed: 11/14/2022] Open
Abstract
Aims Obesity during pregnancy increases risk of cardiovascular disease (CVD) in the offspring and individuals exposed to over-nutrition during fetal life are likely to be exposed to a calorie-rich environment postnatally. Here, we established the consequences of combined exposure to a maternal and post-weaning obesogenic diet on offspring cardiac structure and function using an established mouse model of maternal diet-induced obesity. Methods and results The impact of the maternal and postnatal environment on the offspring metabolic profile, arterial blood pressure, cardiac structure, and function was assessed in 8-week-old C57BL/6 male mice. Measurement of cardiomyocyte cell area, the transcriptional re-activation of cardiac fetal genes as well as genes involved in the regulation of contractile function and matrix remodelling in the adult heart were determined as potential mediators of effects on cardiac function. In the adult offspring: a post-weaning obesogenic diet coupled with exposure to maternal obesity increased serum insulin (P < 0.0001) and leptin levels (P < 0.0001); maternal obesity (P = 0.001) and a post-weaning obesogenic diet (P = 0.002) increased absolute heart weight; maternal obesity (P = 0.01) and offspring obesity (P = 0.01) caused cardiac dysfunction but effects were not additive; cardiac dysfunction resulting from maternal obesity was associated with re-expression of cardiac fetal genes (Myh7: Myh6 ratio; P = 0.0004), however, these genes were not affected by offspring diet; maternal obesity (P = 0.02); and offspring obesity (P = 0.05) caused hypertension and effects were additive. Conclusions Maternal diet-induced obesity and offspring obesity independently promote cardiac dysfunction and hypertension in adult male progeny. Exposure to maternal obesity alone programmed cardiac dysfunction, associated with hallmarks of pathological left ventricular hypertrophy, including increased cardiomyocyte area, upregulation of fetal genes, and remodelling of cardiac structure. These data highlight that the perinatal period is just as important as adult-onset obesity in predicting CVD risk. Therefore, early developmental periods are key intervention windows to reduce the prevalence of CVD.
Collapse
Affiliation(s)
- Elena Loche
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrookes' Treatment Centre, Addenbrookes' Hospital, Hills Road, Cambridge, CB2 OQQ, UK
| | - Heather L Blackmore
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrookes' Treatment Centre, Addenbrookes' Hospital, Hills Road, Cambridge, CB2 OQQ, UK
| | - Asha A Carpenter
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrookes' Treatment Centre, Addenbrookes' Hospital, Hills Road, Cambridge, CB2 OQQ, UK
| | - Jessica H Beeson
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrookes' Treatment Centre, Addenbrookes' Hospital, Hills Road, Cambridge, CB2 OQQ, UK
| | - Adele Pinnock
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrookes' Treatment Centre, Addenbrookes' Hospital, Hills Road, Cambridge, CB2 OQQ, UK
| | - Thomas J Ashmore
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrookes' Treatment Centre, Addenbrookes' Hospital, Hills Road, Cambridge, CB2 OQQ, UK
| | - Catherine E Aiken
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrookes' Treatment Centre, Addenbrookes' Hospital, Hills Road, Cambridge, CB2 OQQ, UK.,Department of Obstetrics and Gynaecology, The Rosie Hospital and NIHR Cambridge Comprehensive Biomedical Research Centre, University of Cambridge, Box 223, Cambridge, CB2 0SW, UK
| | - Juliana de Almeida-Faria
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrookes' Treatment Centre, Addenbrookes' Hospital, Hills Road, Cambridge, CB2 OQQ, UK.,University of Campinas, Faculty of Medical Sciences, Obesity and Comorbidities Research Center, Campinas, 13083-864, Brazil
| | - Josca M Schoonejans
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrookes' Treatment Centre, Addenbrookes' Hospital, Hills Road, Cambridge, CB2 OQQ, UK
| | - Dino A Giussani
- Department of Physiology, Development & Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Denise S Fernandez-Twinn
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrookes' Treatment Centre, Addenbrookes' Hospital, Hills Road, Cambridge, CB2 OQQ, UK
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrookes' Treatment Centre, Addenbrookes' Hospital, Hills Road, Cambridge, CB2 OQQ, UK
| |
Collapse
|
9
|
Santos LS, Cordeiro GS, Perez GS, Santo DAE, Macêdo APA, Lima MS, Carneiro IBC, Machado MEPC, Deiró TCJ, Barreto-Medeiros JM. Influence of mother nutrition during pregnancy and/or lactation on offspring food preference in experimental models. BRAZ J BIOL 2019; 79:220-232. [DOI: 10.1590/1519-6984.179134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 11/30/2017] [Indexed: 11/21/2022] Open
Abstract
Abstract Introduction Understanding associations between food preferences and maternal nutrition during pregnancy and lactation could inform efforts to understanding the obesity mechanisms and provide insight to prevent it. Objective: To identify studies that investigated the effects of nutritional interventions during the pregnancy and lactation on the food preferences of offspring. Method: The review was conducted with search for articles in the databases: Scopus, Pubmed, Medline, LILACS, Scielo and Science Direct. Exclusion criteria were used: reviews, human studies, studies with drugs or other substances not related to food. Results: At the end of the search in the databases, 176 references were found. After use the exclusion criteria, reading the titles, abstracts and full articles, were selected 11 articles to compose the review. Conclusion: The selected studies suggested that unbalanced nutrition in early life alters the food preference and neural components related to the consumption of fatty and sugary foods in offspring rodents.
Collapse
Affiliation(s)
- L. S. Santos
- Universidade Federal da Bahia, Brasil; Universidade Federal da Bahia, Brasil
| | - G. S. Cordeiro
- Universidade Federal da Bahia, Brasil; Universidade Federal da Bahia, Brasil
| | - G. S. Perez
- Universidade Federal da Bahia, Brasil; Universidade Federal da Bahia, Brasil
| | - D. A. E. Santo
- Universidade Federal da Bahia, Brasil; Universidade Federal da Bahia, Brasil
| | | | | | - I. B. C. Carneiro
- Universidade Federal da Bahia, Brasil; Universidade Federal da Bahia, Brasil
| | - M. E. P. C. Machado
- Universidade Federal da Bahia, Brasil; Universidade Federal da Bahia, Brasil
| | - T. C.B. J. Deiró
- Universidade Federal da Bahia, Brasil; Universidade Federal da Bahia, Brasil
| | | |
Collapse
|
10
|
Lee YQ, Collins CE, Gordon A, Rae KM, Pringle KG. The Relationship between Maternal Nutrition during Pregnancy and Offspring Kidney Structure and Function in Humans: A Systematic Review. Nutrients 2018; 10:nu10020241. [PMID: 29466283 PMCID: PMC5852817 DOI: 10.3390/nu10020241] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 02/14/2018] [Accepted: 02/15/2018] [Indexed: 12/30/2022] Open
Abstract
The intrauterine environment is critical for fetal growth and organ development. Evidence from animal models indicates that the developing kidney is vulnerable to suboptimal maternal nutrition and changes in health status. However, evidence from human studies are yet to be synthesised. Therefore, the aim of the current study was to systematically review current research on the relationship between maternal nutrition during pregnancy and offspring kidney structure and function in humans. A search of five databases identified 9501 articles, of which three experimental and seven observational studies met the inclusion criteria. Nutrients reviewed to date included vitamin A (n = 3), folate and vitamin B12 (n = 2), iron (n = 1), vitamin D (n = 1), total energy (n = 2) and protein (n = 1). Seven studies were assessed as being of "positive" and three of "neutral" quality. A variety of populations were studied, with limited studies investigating maternal nutrition during pregnancy, while measurements of offspring kidney outcomes were diverse across studies. There was a lack of consistency in the timing of follow-up for offspring kidney structure and/or function assessments, thus limiting comparability between studies. Deficiencies in maternal folate, vitamin A, and total energy during pregnancy were associated with detrimental impacts on kidney structure and function, measured by kidney volume, proteinuria, eGFRcystC and mean creatinine clearance in the offspring. Additional experimental and longitudinal prospective studies are warranted to confirm this relationship, especially in Indigenous populations where the risk of renal disease is greater.
Collapse
Affiliation(s)
- Yu Qi Lee
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, NSW 2308, Australia.
- Priority Research Centre for Reproductive Sciences, University of Newcastle, Newcastle, NSW 2308, Australia.
| | - Clare E Collins
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Newcastle, NSW 2308, Australia.
- Priority Research Centre in Physical Activity and Nutrition, University of Newcastle, Newcastle, NSW 2308, Australia.
| | - Adrienne Gordon
- Charles Perkins Centre, University of Sydney, Camperdown, NSW 2006, Australia.
| | - Kym M Rae
- Priority Research Centre for Reproductive Sciences, University of Newcastle, Newcastle, NSW 2308, Australia.
- Gomeroi Gaaynggal Centre, Faculty of Health and Medicine, University of Newcastle, Tamworth, NSW 2340, Australia.
- Department of Rural Health, School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Tamworth, NSW 2340, Australia.
- Priority Research Centre for Generational Health and Ageing, University of Newcastle, Newcastle, NSW 2308, Australia.
| | - Kirsty G Pringle
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, NSW 2308, Australia.
- Priority Research Centre for Reproductive Sciences, University of Newcastle, Newcastle, NSW 2308, Australia.
- Gomeroi Gaaynggal Centre, Faculty of Health and Medicine, University of Newcastle, Tamworth, NSW 2340, Australia.
| |
Collapse
|
11
|
Perinatal maternal high-fat diet induces early obesity and sex-specific alterations of the endocannabinoid system in white and brown adipose tissue of weanling rat offspring. Br J Nutr 2017; 118:788-803. [PMID: 29110748 DOI: 10.1017/s0007114517002884] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Perinatal maternal high-fat (HF) diet programmes offspring obesity. Obesity is associated with overactivation of the endocannabinoid system (ECS) in adult subjects, but the role of the ECS in the developmental origins of obesity is mostly unknown. The ECS consists of endocannabinoids, cannabinoid receptors (cannabinoid type-1 receptor (CB1) and cannabinoid type-2 receptor (CB2)) and metabolising enzymes. We hypothesised that perinatal maternal HF diet would alter the ECS in a sex-dependent manner in white and brown adipose tissue of rat offspring at weaning in parallel to obesity development. Female rats received standard diet (9 % energy content from fat) or HF diet (29 % energy content from fat) before mating, during pregnancy and lactation. At weaning, male and female offspring were killed for tissue harvest. Maternal HF diet induced early obesity, white adipocyte hypertrophy and increased lipid accumulation in brown adipose tissue associated with sex-specific changes of the ECS's components in weanling rats. In male pups, maternal HF diet decreased CB1 and CB2 protein in subcutaneous adipose tissue. In female pups, maternal HF diet increased visceral and decreased subcutaneous CB1. In brown adipose tissue, maternal HF diet increased CB1 regardless of pup sex. In addition, maternal HF diet differentially changed oestrogen receptor across the adipose depots in male and female pups. The ECS and oestrogen signalling play an important role in lipogenesis, adipogenesis and thermogenesis, and we observed early changes in their targets in adipose depots of the offspring. The present findings provide insights into the involvement of the ECS in the developmental origins of metabolic disease induced by inadequate maternal nutrition in early life.
Collapse
|
12
|
Dhasarathy A, Roemmich JN, Claycombe KJ. Influence of maternal obesity, diet and exercise on epigenetic regulation of adipocytes. Mol Aspects Med 2016; 54:37-49. [PMID: 27825817 DOI: 10.1016/j.mam.2016.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 10/25/2016] [Accepted: 10/25/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Archana Dhasarathy
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - James N Roemmich
- USDA-ARS-PA, Grand Forks Human Nutrition Research Center, 2420 2nd Avenue North, Grand Forks, ND 58203, USA
| | - Kate J Claycombe
- USDA-ARS-PA, Grand Forks Human Nutrition Research Center, 2420 2nd Avenue North, Grand Forks, ND 58203, USA.
| |
Collapse
|
13
|
Lecoutre S, Deracinois B, Laborie C, Eberlé D, Guinez C, Panchenko PE, Lesage J, Vieau D, Junien C, Gabory A, Breton C. Depot- and sex-specific effects of maternal obesity in offspring's adipose tissue. J Endocrinol 2016; 230:39-53. [PMID: 27122310 DOI: 10.1530/joe-16-0037] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/27/2016] [Indexed: 12/19/2022]
Abstract
According to the Developmental Origin of Health and Disease (DOHaD) concept, alterations of nutrient supply in the fetus or neonate result in long-term programming of individual body weight (BW) setpoint. In particular, maternal obesity, excessive nutrition, and accelerated growth in neonates have been shown to sensitize offspring to obesity. The white adipose tissue may represent a prime target of metabolic programming induced by maternal obesity. In order to unravel the underlying mechanisms, we have developed a rat model of maternal obesity using a high-fat (HF) diet (containing 60% lipids) before and during gestation and lactation. At birth, newborns from obese dams (called HF) were normotrophs. However, HF neonates exhibited a rapid weight gain during lactation, a key period of adipose tissue development in rodents. In males, increased BW at weaning (+30%) persists until 3months of age. Nine-month-old HF male offspring was normoglycemic but showed mild glucose intolerance, hyperinsulinemia, and hypercorticosteronemia. Despite no difference in BW and energy intake, HF adult male offspring was predisposed to fat accumulation showing increased visceral (gonadal and perirenal) depots weights and hyperleptinemia. However, only perirenal adipose tissue depot exhibited marked adipocyte hypertrophy and hyperplasia with elevated lipogenic (i.e. sterol-regulated element binding protein 1 (Srebp1), fatty acid synthase (Fas), and leptin) and diminished adipogenic (i.e. peroxisome proliferator-activated receptor gamma (Pparγ), 11β-hydroxysteroid dehydrogenase type 1 (11β-Hds1)) mRNA levels. By contrast, very few metabolic variations were observed in HF female offspring. Thus, maternal obesity and accelerated growth during lactation program offspring for higher adiposity via transcriptional alterations of visceral adipose tissue in a depot- and sex-specific manner.
Collapse
Affiliation(s)
- Simon Lecoutre
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| | - Barbara Deracinois
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| | - Christine Laborie
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| | - Delphine Eberlé
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| | - Céline Guinez
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| | - Polina E Panchenko
- INRAUMR1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France
| | - Jean Lesage
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| | - Didier Vieau
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| | - Claudine Junien
- INRAUMR1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France UVSQUniversité Versailles-Saint-Quentin-en-Yvelines, Versailles, France
| | - Anne Gabory
- INRAUMR1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France
| | - Christophe Breton
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| |
Collapse
|
14
|
Maternal Obesity Promotes Diabetic Nephropathy in Rodent Offspring. Sci Rep 2016; 6:27769. [PMID: 27277011 PMCID: PMC4899795 DOI: 10.1038/srep27769] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 05/25/2016] [Indexed: 02/07/2023] Open
Abstract
Maternal obesity is known to increase the risk of obesity and diabetes in offspring. Though diabetes is a key risk factor for the development of chronic kidney disease (CKD), the relationship between maternal obesity and CKD has not been clearly defined. In this study, a mouse model of maternal obesity was employed to determine the impact of maternal obesity on development of diabetic nephropathy in offspring. Female C57BL/6 mice were fed high-fat diet (HFD) for six weeks prior to mating, during gestation and lactation. Male offspring were weaned to normal chow diet. At postnatal Week 8, offspring were randomly administered low dose streptozotocin (STZ, 55 mg/kg/day for five days) to induce diabetes. Assessment of renal damage took place at postnatal Week 32. We found that offspring of obese mothers had increased renal fibrosis, inflammation and oxidative stress. Importantly, offspring exposed to maternal obesity had increased susceptibility to renal damage when an additional insult, such as STZ-induced diabetes, was imposed. Specifically, renal inflammation and oxidative stress induced by diabetes was augmented by maternal obesity. Our findings suggest that developmental programming induced by maternal obesity has implications for renal health in offspring. Maternal obesity should be considered a risk factor for CKD.
Collapse
|
15
|
Lim WC, Ho JN, Lee HS, Cho HY. Germinated Waxy Black Rice Suppresses Weight Gain in High-Fat Diet-Induced Obese Mice. J Med Food 2016; 19:410-7. [DOI: 10.1089/jmf.2015.3590] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Won-Chul Lim
- Department of Food and Biotechnology, Korea University, Sejong, Korea
| | - Jin-Nyoung Ho
- Biomedical Research Institute, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hee-Seop Lee
- Department of Food and Biotechnology, Korea University, Sejong, Korea
| | - Hong-Yon Cho
- Department of Food and Biotechnology, Korea University, Sejong, Korea
| |
Collapse
|
16
|
Imessaoudene A, Merzouk H, Berroukeche F, Mokhtari N, Bensenane B, Cherrak S, Merzouk SA, Elhabiri M. Beneficial effects of quercetin–iron complexes on serum and tissue lipids and redox status in obese rats. J Nutr Biochem 2016; 29:107-15. [DOI: 10.1016/j.jnutbio.2015.11.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 11/02/2015] [Accepted: 11/20/2015] [Indexed: 01/08/2023]
|
17
|
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: 0.8] [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
|
18
|
Penfold NC, Ozanne SE. Developmental programming by maternal obesity in 2015: Outcomes, mechanisms, and potential interventions. Horm Behav 2015; 76:143-52. [PMID: 26145566 DOI: 10.1016/j.yhbeh.2015.06.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 06/23/2015] [Accepted: 06/24/2015] [Indexed: 02/06/2023]
Abstract
This article is part of a Special Issue "SBN 2014". Obesity in women of child-bearing age is a growing problem in developed and developing countries. Evidence from human studies indicates that maternal BMI correlates with offspring adiposity from an early age and predisposes to metabolic disease in later life. Thus the early life environment is an attractive target for intervention to improve public health. Animal models have been used to investigate the specific physiological outcomes and mechanisms of developmental programming that result from exposure to maternal obesity in utero. From this research, targeted intervention strategies can be designed. In this review we summarise recent progress in this field, with a focus on cardiometabolic disease and central control of appetite and behaviour. We highlight key factors that may mediate programming by maternal obesity, including leptin, insulin, and ghrelin. Finally, we explore potential lifestyle and pharmacological interventions in humans and the current state of evidence from animal models.
Collapse
Affiliation(s)
- Naomi C Penfold
- University of Cambridge, Metabolic Research Laboratories MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom.
| | - Susan E Ozanne
- University of Cambridge, Metabolic Research Laboratories MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
19
|
Pereira TJ, Moyce BL, Kereliuk SM, Dolinsky VW. Influence of maternal overnutrition and gestational diabetes on the programming of metabolic health outcomes in the offspring: experimental evidence. Biochem Cell Biol 2015; 93:438-451. [PMID: 25673017 DOI: 10.1139/bcb-2014-0141] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2023] Open
Abstract
The incidence of obesity and type 2 diabetes mellitus have risen across the world during the past few decades and has also reached an alarming level among children. In addition, women are currently more likely than ever to enter pregnancy obese. As a result, the incidence of gestational diabetes mellitus is also on the rise. While diet and lifestyle contribute to these trends, population health data show that maternal obesity and diabetes during pregnancy during critical stages of development are major factors that contribute to the development of chronic disease in adolescent and adult offspring. Fetal programming of metabolic function, through physiological and (or) epigenetic mechanisms, may also have an intergenerational effect, and as a result may perpetuate metabolic disorders in the next generation. In this review, we summarize the existing literature that characterizes how maternal obesity and gestational diabetes mellitus contribute to metabolic and cardiovascular disorders in the offspring. In particular, we focus on animal studies that investigate the molecular mechanisms that are programmed by the gestational environment and lead to disease phenotypes in the offspring. We also review interventional studies that prevent disease with a developmental origin in the offspring.
Collapse
Affiliation(s)
- Troy J Pereira
- University of Manitoba, Department of Pharmacology and Therapeutics, Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada
- University of Manitoba, Department of Pharmacology and Therapeutics, Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada
| | - Brittany L Moyce
- University of Manitoba, Department of Pharmacology and Therapeutics, Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada
- University of Manitoba, Department of Pharmacology and Therapeutics, Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada
| | - Stephanie M Kereliuk
- University of Manitoba, Department of Pharmacology and Therapeutics, Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada
- University of Manitoba, Department of Pharmacology and Therapeutics, Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada
| | - Vernon W Dolinsky
- University of Manitoba, Department of Pharmacology and Therapeutics, Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada
- University of Manitoba, Department of Pharmacology and Therapeutics, Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
20
|
Pereira TJ, Fonseca MA, Campbell KE, Moyce BL, Cole LK, Hatch GM, Doucette CA, Klein J, Aliani M, Dolinsky VW. Maternal obesity characterized by gestational diabetes increases the susceptibility of rat offspring to hepatic steatosis via a disrupted liver metabolome. J Physiol 2015; 593:3181-97. [PMID: 25922055 DOI: 10.1113/jp270429] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/17/2015] [Indexed: 12/16/2022] Open
Abstract
Maternal obesity is associated with a high risk for gestational diabetes mellitus (GDM), which is a common complication of pregnancy. The influence of maternal obesity and GDM on the metabolic health of the offspring is poorly understood. We hypothesize that GDM associated with maternal obesity will cause obesity, insulin resistance and hepatic steatosis in the offspring. Female Sprague-Dawley rats were fed a high-fat (45%) and sucrose (HFS) diet to cause maternal obesity and GDM. Lean control pregnant rats received low-fat (LF; 10%) diets. To investigate the interaction between the prenatal environment and postnatal diets, rat offspring were assigned to LF or HFS diets for 12 weeks, and insulin sensitivity and hepatic steatosis were evaluated. Pregnant GDM dams exhibited excessive gestational weight gain, hyperinsulinaemia and hyperglycaemia. Offspring of GDM dams gained more weight than the offspring of lean dams due to excess adiposity. The offspring of GDM dams also developed hepatic steatosis and insulin resistance. The postnatal consumption of a LF diet did not protect offspring of GDM dams against these metabolic disorders. Analysis of the hepatic metabolome revealed increased diacylglycerol and reduced phosphatidylethanolamine in the offspring of GDM dams compared to offspring of lean dams. Consistent with altered lipid metabolism, the expression of CTP:phosphoethanolamine cytidylyltransferase, and peroxisomal proliferator activated receptor-α mRNA was reduced in the livers of GDM offspring. GDM exposure programs gene expression and hepatic metabolite levels and drives the development of hepatic steatosis and insulin resistance in young adult rat offspring.
Collapse
Affiliation(s)
- Troy J Pereira
- Department of Pharmacology & Therapeutics.,Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme.,Children's Hospital Research Institute of Manitoba
| | - Mario A Fonseca
- Department of Pharmacology & Therapeutics.,Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme.,Children's Hospital Research Institute of Manitoba
| | - Kristyn E Campbell
- Department of Pharmacology & Therapeutics.,Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme.,Children's Hospital Research Institute of Manitoba
| | - Brittany L Moyce
- Department of Pharmacology & Therapeutics.,Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme.,Children's Hospital Research Institute of Manitoba
| | - Laura K Cole
- Department of Pharmacology & Therapeutics.,Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme.,Children's Hospital Research Institute of Manitoba
| | - Grant M Hatch
- Department of Pharmacology & Therapeutics.,Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme.,Children's Hospital Research Institute of Manitoba
| | - Christine A Doucette
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme.,Children's Hospital Research Institute of Manitoba.,Department of Physiology and Pathophysiology
| | | | - Michel Aliani
- Department of Human Nutrition, University of Manitoba, Winnipeg, MB, Canada
| | - Vernon W Dolinsky
- Department of Pharmacology & Therapeutics.,Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme.,Children's Hospital Research Institute of Manitoba
| |
Collapse
|
21
|
Lecoutre S, Breton C. Maternal nutritional manipulations program adipose tissue dysfunction in offspring. Front Physiol 2015; 6:158. [PMID: 26029119 PMCID: PMC4429565 DOI: 10.3389/fphys.2015.00158] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/30/2015] [Indexed: 12/11/2022] Open
Abstract
Based on the concept of Developmental Origin of Health and Disease, both human and animal studies have demonstrated a close link between nutrient supply perturbations in the fetus or neonate (i.e., maternal undernutrition, obesity, gestational diabetes and/or rapid catch-up growth) and increased risk of adult-onset obesity. Indeed, the adipose tissue has been recognized as a key target of developmental programming in a sex-and depot-specific manner. Despite different developmental time windows, similar mechanisms of adipose tissue programming have been described in rodents and in bigger mammals (sheep, primates). Maternal nutritional manipulations reprogram offspring's adipose tissue resulting in series of alterations: enhanced adipogenesis and lipogenesis, impaired sympathetic activity with reduced noradrenergic innervations and thermogenesis as well as low-grade inflammation. These changes affect adipose tissue development, distribution and composition predisposing offspring to fat accumulation. Modifications of hormonal tissue sensitivity (i.e., leptin, insulin, glucocorticoids) and/or epigenetic mechanisms leading to persistent changes in gene expression may account for long-lasting programming across generations.
Collapse
Affiliation(s)
- Simon Lecoutre
- Unité Environnement Périnatal et Santé, UPRES EA 4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Université de Lille Villeneuve d'Ascq, France
| | - Christophe Breton
- Unité Environnement Périnatal et Santé, UPRES EA 4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Université de Lille Villeneuve d'Ascq, France
| |
Collapse
|
22
|
Maternal high-fat-diet programs rat offspring liver fatty acid metabolism. Lipids 2015; 50:565-73. [PMID: 25899040 DOI: 10.1007/s11745-015-4018-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 04/02/2015] [Indexed: 12/28/2022]
Abstract
In offspring exposed in utero to a maternal diet high in fat (HF), we have previously demonstrated that despite similar birth weights, HF adult offspring at 6 months of age had significantly higher body weights, greater adiposity, and increased triacylglycerol (TAG) levels as compared to controls. We hypothesized that a maternal HF diet predisposes to offspring adiposity via a programmed increase in the synthesis of monounsaturated fatty acids in the liver and hence increased substrate availability for liver TAG synthesis. We further hypothesized that programmed changes in offspring liver fatty acid metabolism are associated with increased liver expression of the lipogenic enzyme stearoyl-CoA desaturase-1 (SCD-1). Female rats were maintained on a HF diet rich in monounsaturated fatty acids (MUFA) prior to and throughout pregnancy and lactation. After birth, newborns were nursed by the same dam, and all offspring were weaned to control diet. Plasma and liver fatty acid compositions were determined using gas chromatography/mass spectrometry. Fatty acid C16 desaturation indices of palmitoleic/palmitic and (vaccenic + palmitoleic)/palmitic and the C18 desaturation index of oleic/stearic were calculated. Liver protein abundance of SCD-1 was analyzed in newborns and adult offspring. Plasma and liver C16 desaturation indices were decreased in HF newborns, but increased in the adult offspring. Liver SCD-1 expression was increased in the HF adult offspring. These data show that the maternal HF diet during pregnancy and lactation increases offspring liver SCD-1 protein abundance and alters the liver C16 desaturase pathway.
Collapse
|
23
|
Seo YS, Kang OH, Kim SB, Mun SH, Kang DH, Yang DW, Choi JG, Lee YM, Kang DK, Lee HS, Kwon DY. Quercetin prevents adipogenesis by regulation of transcriptional factors and lipases in OP9 cells. Int J Mol Med 2015; 35:1779-85. [PMID: 25891365 DOI: 10.3892/ijmm.2015.2185] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 04/08/2015] [Indexed: 11/06/2022] Open
Abstract
With the industrialization of society, the increase in the prevalence of obesity and metabolic disorders has become an important health concern in a number of countries. Quercetin (3,30,40,5,7-pentahydroxyflavone) is well known as a bioactive flavonoid in a variety of biological resources. The aim of the present study was to explore the machanisms responsible for the anti-adipogenic activity of quercetin and its effects on the lipolysis in OP9 mouse stromal cells which rapidly differentiate into adipocytes. The differentiation of OP9 cells into adipocytes was evaluated by the measurement of lipid accumulation by Oil Red O (ORO) staining; lipid accumulation was significantly impaired by treatment with quercetin. Reverse transcription-polymerase chain reaction (RT-PCR) and western blot analysis were used to measure the expression levels of CCAAT/enhancer binding protein α (C/EBPα), proliferator-activated receptor γ (PPARγ), sterol regulatory element-binding protein-1 (SREBP-1) and fatty acid synthase (FAS). The mRNA expression levels of lipases, such as adipose triglyceride lipase (ATGL), hormone sensitive lipase (HSL) and lipoprotein lipase (LPL) were also measured by RT-PCR. Quercetin significantly decreased the expression of transcription factors, including C/EBPα, PPARγ and SREBP-1c both at the protein and mRNA level. The results from the present study demonstrate that quercetin prevents adipogenesis by upregulating ATGL and HSL expression and downregulating FAS, LPL and adipocyte fatty acid-binding protein (aP2) expression, as well as the expression of transcription factors. Our data suggest that quercetin has therapeutic potential by regulating the expression of transcriptional factors and enzymes associated with adipogenesis.
Collapse
Affiliation(s)
- Yun-Soo Seo
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang-Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | - Ok-Hwa Kang
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang-Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | - Sung-Bae Kim
- BK21 Plus Team, Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | - Su-Hyun Mun
- BK21 Plus Team, Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | - Da-Hye Kang
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang-Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | - Da-Wun Yang
- BK21 Plus Team, Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | - Jang-Gi Choi
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang-Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | - Young-Mi Lee
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang-Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | - Dae-Kil Kang
- BK21 Plus Team, Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | - Ho-Seog Lee
- BK21 Plus Team, Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | - Dong-Yeul Kwon
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang-Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| |
Collapse
|
24
|
Raipuria M, Bahari H, Morris MJ. Effects of maternal diet and exercise during pregnancy on glucose metabolism in skeletal muscle and fat of weanling rats. PLoS One 2015; 10:e0120980. [PMID: 25853572 PMCID: PMC4390148 DOI: 10.1371/journal.pone.0120980] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 02/09/2015] [Indexed: 11/19/2022] Open
Abstract
Obesity during pregnancy contributes to the development of metabolic disorders in offspring. Maternal exercise may limit gestational weight gain and ameliorate these programming effects. We previously showed benefits of post-weaning voluntary exercise in offspring from obese dams. Here we examined whether voluntary exercise during pregnancy influences lipid and glucose homeostasis in muscle and fat in offspring of both lean and obese dams. Female Sprague-Dawley rats were fed chow (C) or high fat (F) diet for 6 weeks before mating. Half underwent voluntary exercise (CE/FE) with a running wheel introduced 10 days prior to mating and available until the dams delivered; others remained sedentary (CS/FS). Male and female pups were killed at postnatal day (PND)19 and retroperitoneal fat and gastrocnemius muscle were collected for gene expression. Lean and obese dams achieved similar modest levels of exercise. At PND1, both male and female pups from exercised lean dams were significantly lighter (CE versus CS), with no effect in those from obese dams. At PND19, maternal obesity significantly increased offspring body weight and adiposity, with no effect of maternal exercise. Exercise significantly reduced insulin concentrations in males (CE/FE versus CS/FS), with reduced glucose in male FE pups. In males, maternal obesity significantly decreased muscle myogenic differentiation 1 (MYOD1) and glucose transporter type 4 (GLUT4) mRNA expressions (FS vs CS); these were normalized by exercise. Maternal exercise upregulated adipose GLUT4, interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and peroxisome proliferator activated receptor gamma coactivator 1 alpha (PGC1α) mRNA expression in offspring of dams consuming chow. Modest voluntary exercise during pregnancy was associated with lower birth weight in pups from lean dams. Maternal exercise appeared to decrease the metabolic risk induced by maternal obesity, improving insulin/glucose metabolism, with greater effects in male than female offspring.
Collapse
Affiliation(s)
- Mukesh Raipuria
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia
| | - Hasnah Bahari
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Margaret J. Morris
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia
- * E-mail:
| |
Collapse
|
25
|
Giblin L, Darimont C, Leone P, McNamara LB, Blancher F, Berry D, Castañeda-Gutiérrez E, Lawlor PG. Offspring subcutaneous adipose markers are sensitive to the timing of maternal gestational weight gain. Reprod Biol Endocrinol 2015; 13:16. [PMID: 25879645 PMCID: PMC4363193 DOI: 10.1186/s12958-015-0009-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/12/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Excessive maternal weight gain during pregnancy impacts on offspring health. This study focused on the timing of maternal gestational weight gain, using a porcine model with mothers of normal pre-pregnancy weight. METHODS Trial design ensured the trajectory of maternal gestational weight gain differed across treatments in early, mid and late gestation. Diet composition did not differ. On day 25 gestation, sows were assigned to one of five treatments: Control sows received a standard gestation diet of 2.3 kg/day (30 MJ DE/day) from early to late gestation (day 25-110 gestation). E sows received 4.6 kg food/day in early gestation (day 25-50 gestation). M sows doubled their food intake in mid gestation (day 50-80 gestation). EM sows doubled their food intake during both early and mid gestation (day 25-80 gestation). L sows consumed 3.5 kg food/day in late gestation (day 80-110 gestation). Offspring body weight and food intake levels were measured from birth to adolescence. Markers of lipid metabolism, hypertrophy and inflammation were investigated in subcutaneous adipose tissue of adolescent offspring. RESULTS The trajectory of gestational weight gain differed across treatments. However total gestational weight gain did not differ except for EM sows who were the heaviest and fattest mothers at parturition. Offspring birth weight did not differ across treatments. Subcutaneous adipose tissue from EM offspring differed significantly from controls, with elevated mRNA levels of lipogenic (CD36, ACACB and LPL), nutrient transporters (FABP4 and GLUT4), lipolysis (HSL and ATGL), adipocyte size (MEST) and inflammation (PAI-1) indicators. The subcutaneous adipose depot from L offspring exhibited elevated levels of CD36, ACACB, LPL, GLUT4 and FABP4 mRNA transcripts compared to control offspring. CONCLUSIONS Increasing gestational weight gain in early gestation had the greatest impact on offspring postnatal growth rate. Increasing maternal food allowance in late gestation appeared to shift the offspring adipocyte focus towards accumulation of fat. Mothers who gained the most weight during gestation (EM mothers) gave birth to offspring whose subcutaneous adipose tissue, at adolescence, appeared hyperactive compared to controls. This study concluded that mothers, who gained more than the recommended weight gain in mid and late gestation, put their offspring adipose tissue at risk of dysfunction.
Collapse
Affiliation(s)
- Linda Giblin
- Teagasc Food Research Centre, Moorepark, Fermoy, Co.Cork, Ireland.
| | - Christian Darimont
- Nestlé Research Centre, Nutrition & Health Research Department, Vers-Chez-les-Blanc, Lausanne, Switzerland.
| | - Patricia Leone
- Nestlé Research Centre, Nutrition & Health Research Department, Vers-Chez-les-Blanc, Lausanne, Switzerland.
| | - Louise B McNamara
- Teagasc Food Research Centre, Moorepark, Fermoy, Co.Cork, Ireland.
- Animal and Grassland Research and Innovation Centre, Teagasc, Moorepark, Fermoy, Co. Cork, Ireland.
| | - Florence Blancher
- Nestlé Research Centre, Nutrition & Health Research Department, Vers-Chez-les-Blanc, Lausanne, Switzerland.
| | - Donagh Berry
- Animal and Grassland Research and Innovation Centre, Teagasc, Moorepark, Fermoy, Co. Cork, Ireland.
| | | | - Peadar G Lawlor
- Animal and Grassland Research and Innovation Centre, Teagasc, Moorepark, Fermoy, Co. Cork, Ireland.
| |
Collapse
|
26
|
Impact of cafeteria feeding during lactation in the rat on novel object discrimination in the offspring. Br J Nutr 2014; 112:1933-7. [DOI: 10.1017/s0007114514003134] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
There is increasing evidence that hyperenergetic diets have an impact on memory in rodents. However, it is largely unknown how diets, such as a cafeteria diet (CD), that mimic a Western-type diet act on learning and memory, in particular when fed during early stages of development. Here, we fed lactating dams a CD and exposed both male and female offspring to a novel object discrimination (NOD) task, a two-trial test of recognition memory in which rats exposed to two identical objects during a training/familiarisation trial can discriminate a novel from a familiar object during the subsequent choice trial. The choice trial was performed following inter-trial interval (ITI) delays of up to 4 h. Maternal diet did not have an impact on exploration of the objects by either sex during the familiarisation trial. Control males discriminated the novel from the familiar object, indicating intact memory with an ITI of 1 h, but not 2 or 4 h. The CD delayed this natural forgetting in male rats such that discrimination was also evident after a 2 h ITI. In contrast, control females exhibited discrimination following both 1 and 2 h ITI, but the CD impaired performance. In summary, the present study shows that maternal exposure to the CD programmes NOD in the adult. In better-performing females, dietary programming interferes with NOD, whereas NOD was improved in males after lactational CD feeding.
Collapse
|
27
|
Lecoutre S, Breton C. The cellularity of offspring's adipose tissue is programmed by maternal nutritional manipulations. Adipocyte 2014; 3:256-62. [PMID: 26317049 DOI: 10.4161/adip.29806] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/23/2014] [Accepted: 07/01/2014] [Indexed: 12/17/2022] Open
Abstract
Epidemiological studies initially demonstrated that maternal undernutrition leads to low birth weight with increased risk of adult-onset obesity. Maternal obesity and diabetes associated with high birth weight, excessive nutrition in neonates, and rapid catch-up growth also predispose offspring to fat accumulation. As stated by the Developmental Origin of Health and Disease concept, nutrient supply perturbations in the fetus or neonate result in long-term programming of individual body weight set-point. Adipose tissue is a key fuel storage unit mainly involved in the maintenance of energy homeostasis. Studies in numerous animal models have demonstrated that the adipose tissue is the focus of developmental programming events in a gender- and depot-specific manner. This review summarizes the impact of maternal nutritional manipulations on cellularity (i.e., cell number, size, and type) of adipose tissue in programmed offspring. In rodents, adipose tissue development is particularly active during the perinatal period, especially during the last week of gestation and during early postnatal life. In contrast to rodents, this process essentially takes place before birth in bigger mammals. Despite these different developmental time windows, altricial and precocial species share several mechanisms of adipose tissue programming. Maternal nutritional manipulations result in increased adipogenesis and modified fat distribution and composition. Inflammation changes such as infiltration of macrophages and increased inflammatory markers are also observed. Overall, it may predispose offspring to fat accumulation and obesity. Inappropriate hormone levels, modified tissue sensitivity, and epigenetic mechanisms are key factors involved in the programming of adipose tissue's cellularity during the perinatal period.
Collapse
|
28
|
Park JE, Oh SH, Cha YS. Lactobacillus brevis OPK-3 isolated from kimchi inhibits adipogenesis and exerts anti-inflammation in 3T3-L1 adipocyte. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2014; 94:2514-2520. [PMID: 24453065 DOI: 10.1002/jsfa.6588] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 12/26/2013] [Accepted: 01/22/2014] [Indexed: 06/03/2023]
Abstract
BACKGROUND Kimchi is a traditional fermented food in Korea that contains various unique microorganisms. Diverse bacteria are involved in the process of Kimchi fermentation and the healthful advantages; one of the major species is Lactobacillus. We investigated whether lactic acid bacteria isolated from Kimchi (KLAB) are capable of reducing intracellular lipid accumulation by downregulating the expression of adipogenesis and lipogenesis promoting genes in differentiating 3T3-L1 cells. RESULTS KLAB (Lactobacillus brevis OPK-3) mediated dose-dependent inhibition of adipocyte differentiation, intracellular triglyceride accumulation and glycerol-3-phosphate dehydrogenase (GPDH) activity. The expression of transcription factors such as peroxisome proliferator-activated receptor γ and CCAAT/enhancer-binding protein α involved in adipogenesis was markedly decreased by the KLAB treatment. Terminal adipogenic marker, e.g. adipocyte fatty acid binding protein (aP2), lipoprotein lipase, liver X receptor α, leptin and GPDH were significantly downregulated by KLAB treatment compared to untreated control. Moreover, cytokine genes, such as tumor necrosis factor-α and interleukin-6 mRNA expressions level were also decreased, whereas adiponectin mRNA level was upregulated by KLAB. CONCLUSION These results suggest that the KLAB inhibits lipid accumulation in the differentiating adipocyte through downregulating the expression of adipogenic transcription factors and other specific genes involved in lipid metabolism.
Collapse
Affiliation(s)
- Jeong-Eun Park
- Department of Food Science and Human Nutrition, Chonbuk National University, Jeonju, Republic of Korea; Jeonju Makgeolli Research Center, Chonbuk National University, Jeonju, Republic of Korea
| | | | | |
Collapse
|
29
|
Mark PJ, Wyrwoll CS, Zulkafli IS, Mori TA, Waddell BJ. Rescue of glucocorticoid-programmed adipocyte inflammation by omega-3 fatty acid supplementation in the rat. Reprod Biol Endocrinol 2014; 12:39. [PMID: 24886466 PMCID: PMC4022445 DOI: 10.1186/1477-7827-12-39] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 04/26/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Adverse fetal environments predispose offspring to pathologies associated with the metabolic syndrome. Previously we demonstrated that adult offspring of dexamethasone-treated mothers had elevated plasma insulin and pro-inflammatory cytokines, effects prevented by a postnatal diet enriched with omega (n)-3 fatty acids. Here we tested whether prenatal glucocorticoid excess also programmed the adipose tissue phenotype, and whether this outcome is rescued by dietary n-3 fatty acids. METHODS Offspring of control and dexamethasone-treated mothers (0.75 μg/ml in drinking water, day 13 to term) were cross-fostered to mothers on a standard (Std) or high n-3 (Hn3) diet at birth. Offspring remained on these diets post-weaning, and serum and retroperitoneal fat were obtained at 6 months of age (n = 5-8 per group). Serum was analysed for blood lipids and fatty acid profiles, adipocyte cross sectional area was measured by unbiased stereological analysis and adipose expression of markers of inflammation, glucocorticoid sensitivity and lipid metabolism were determined by RT-qPCR analysis. RESULTS Serum total fatty acid levels were elevated (P < 0.01) in male offspring of dexamethasone-treated mothers, an effect prevented by Hn3 consumption. Prenatal dexamethasone also programmed increased adipose expression of Il6, Il1b (both P < 0.05) and Tnfa (P < 0.001) mRNAs regardless of fetal sex, but again this effect was prevented (for Il6 and Il1b) by Hn3 consumption. Offspring of dexamethasone-treated mothers had increased adipose expression of Gr (P = 0.008) and Ppara (P < 0.05) regardless of sex or postnatal diet, while 11bHsd1 was upregulated in males only. The Hn3 diet increased Ppard expression and reduced adipocyte size in all offspring (both P < 0.05) irrespective of prenatal treatment. CONCLUSIONS Prenatal glucocorticoid exposure programmed increased expression of inflammatory markers and enhanced glucocorticoid sensitivity of adipose tissue. Partial prevention of this phenotype by high n-3 consumption indicates that postnatal dietary manipulations can limit adverse fetal programming effects on adipose tissue.
Collapse
Affiliation(s)
- Peter J Mark
- School of Anatomy, Physiology & Human Biology, The University of Western Australia, Perth, Australia
| | - Caitlin S Wyrwoll
- School of Anatomy, Physiology & Human Biology, The University of Western Australia, Perth, Australia
| | - Intan S Zulkafli
- School of Anatomy, Physiology & Human Biology, The University of Western Australia, Perth, Australia
| | - Trevor A Mori
- School of Medicine and Pharmacology, The University of Western Australia, Perth, Australia
| | - Brendan J Waddell
- School of Anatomy, Physiology & Human Biology, The University of Western Australia, Perth, Australia
| |
Collapse
|
30
|
HO JN, KIM OK, NAM DE, JUN W, LEE J. Pycnogenol Supplementation Promotes Lipolysis via Activation of cAMP-Dependent PKA in ob/ob Mice and Primary-Cultured Adipocytes. J Nutr Sci Vitaminol (Tokyo) 2014; 60:429-35. [DOI: 10.3177/jnsv.60.429] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Jin-Nyoung HO
- Department of Medical Nutrition, Kyung Hee University
| | - Ok-Kyung KIM
- Department of Medical Nutrition, Kyung Hee University
| | - Da-Eun NAM
- Department of Medical Nutrition, Kyung Hee University
| | - Woojin JUN
- Department of Food and Nutrition, Chonnam National University
| | - Jeongmin LEE
- Research Institute of Medical Nutrition, Kyung Hee University
- Department of Medical Nutrition, Kyung Hee University
| |
Collapse
|
31
|
Effects of exposure to a cafeteria diet during gestation and after weaning on the metabolism and body weight of adult male offspring in rats. Br J Nutr 2013; 111:1499-506. [DOI: 10.1017/s0007114513003838] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the present study, we investigated whether maternal exposure to a cafeteria diet affects the metabolism and body composition of offspring and whether such an exposure has a cumulative effect during the lifetime of the offspring. Female rats were fed a control (CON) or a cafeteria (CAF) diet from their own weaning to the weaning of their offspring. At 21 d of age, male offspring were divided into four groups by diet during gestation and after weaning (CON-CON, CON-CAF, CAF-CON and CAF-CAF). Blood was collected from dams (after weaning) and pups (at 30 and 120 d of age) by decapitation. CAF dams had significantly greater body weight and adipose tissue weight and higher concentrations of total cholesterol, insulin and leptin than CON dams (Student's t test). The energy intake of CAF rats was higher than that of CON rats regardless of the maternal diet (two-way ANOVA). Litters had similar body weights at weaning and at 30 d of age, but at 120 d, CON-CAF rats were heavier. At both ages, CAF rats had greater adipose tissue weight than CON rats regardless of the maternal diet, and the concentrations of TAG and cholesterol were similar between the two groups, as were blood glucose concentrations at 30 d of age. However, at 120 d of age, CAF rats were hyperglycaemic, hyperinsulinaemic and hyperleptinaemic regardless of the maternal diet. These findings suggest that maternal obesity does not modulate the metabolism of male offspring independently, modifying body weight only when associated with the intake of a cafeteria diet by the offspring.
Collapse
|
32
|
Lukaszewski MA, Eberlé D, Vieau D, Breton C. Nutritional manipulations in the perinatal period program adipose tissue in offspring. Am J Physiol Endocrinol Metab 2013; 305:E1195-207. [PMID: 24045869 DOI: 10.1152/ajpendo.00231.2013] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Epidemiological studies demonstrated initially that maternal undernutrition results in low birth weight with increased risk for long-lasting energy balance disorders. Maternal obesity and diabetes associated with high birth weight, excessive nutrition in neonates, and rapid catchup growth also increase the risk of adult-onset obesity. As stated by the Developmental Origin of Health and Disease concept, nutrient supply perturbations in the fetus or neonate result in long-term programming of individual body weight set point. Adipose tissue is a key fuel storage unit involved mainly in the maintenance of energy homeostasis. Studies in numerous animal models have demonstrated that the adipose tissue is the focus of developmental programming events in a sex- and depot-specific manner. In rodents, adipose tissue development is particularly active during the perinatal period, especially during the last week of gestation and during early postnatal life. In contrast to rodents, this process essentially takes place before birth in bigger mammals. Despite these different developmental time windows, altricial and precocial species share several mechanisms of adipose tissue programming. Offspring from malnourished dams present adipose tissue with a series of alterations: impaired glucose uptake, insulin and leptin resistance, low-grade inflammation, modified sympathetic activity with reduced noradrenergic innervations, and thermogenesis. These modifications reprogram adipose tissue metabolism by changing fat distribution and composition and by enhancing adipogenesis, predisposing the offspring to fat accumulation. Subtle adipose tissue circadian rhythm changes are also observed. Inappropriate hormone levels, modified tissue sensitivity (especially glucocorticoid system), and epigenetic mechanisms are key factors for adipose tissue programming during the perinatal period.
Collapse
Affiliation(s)
- Marie-Amélie Lukaszewski
- Unité Environnement Périnatal et Croissance, UPRES EA 4489, Equipe Dénutritions Maternelles Périnatales, Université Lille-Nord de France, Villeneuve d'Ascq, France
| | | | | | | |
Collapse
|
33
|
Christante CM, Taboga SR, Pinto-Fochi ME, Góes RM. Maternal obesity disturbs the postnatal development of gonocytes in the rat without impairment of testis structure at prepubertal age. Reproduction 2013; 146:549-58. [PMID: 24043845 DOI: 10.1530/rep-13-0037] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In this study, we evaluated whether maternal obesity (MO) affects testis development and gonocyte differentiation in the rat from 0.5 to 14.5 postnatal days. Male Wistar rats were used at 0.5, 4.5, 7.5, and 14.5 days post partum (dpp). These rats were born from obese mothers, previously fed with a high-fat diet (20% saturated fat), for 15 weeks, or normal mothers that had received a balanced murine diet (4% lipids). MO did not affect testis weight or histology at birth but changed the migratory behavior of gonocytes. The density of relocated cells was higher in MO pups at 0.5 dpp, decreased at 4.5 dpp, and differed from those of control pups, where density increased exponentially from 0.5 to 7.5 dpp. The numerical density of gonocytes within seminiferous cords did not vary in MO, in relation to control neonates, for any age considered, but the testis weight was 50% lower at 4.5 dpp. A wide variation in plasmatic testosterone and estrogen levels was observed among the groups during the first week of age and MO pups exhibited higher steroid concentrations at 4.5 dpp, in comparison with controls. At this age, higher estrogen levels of MO pups impaired the gonocyte proliferation. At 7.5 dpp, the testicular size and other parameters of gonocyte development are retrieved. In conclusion, MO and saturated lipid diets disturb gonocyte development and sexual steroid levels during the first days of life, with recovery at prepubertal age.
Collapse
Affiliation(s)
- Caroline Maria Christante
- Department of Biology, Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, IBILCE/UNESP, Rua Cristóvão Colombo, 2265, CEP 15054-000 São José do Rio Preto, São Paulo, Brazil
| | | | | | | |
Collapse
|
34
|
MacKay H, Khazall R, Patterson ZR, Wellman M, Abizaid A. Rats perinatally exposed to food restriction and high-fat diet show differences in adipose tissue gene expression under chronic caloric restriction. Adipocyte 2013; 2:237-45. [PMID: 24052899 PMCID: PMC3774699 DOI: 10.4161/adip.24752] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 04/12/2013] [Accepted: 04/19/2013] [Indexed: 11/24/2022] Open
Abstract
The aim of this study is to analyze how maternal diet during the lactational period influences the adipose tissue response to chronic caloric restriction in offspring. Lactating dams were subjected to one of three treatments: 50% food restriction (FR), ad lib standard chow (AL), or ad lib high-fat diet (HF). Juveniles were first weaned onto standard chow, then in adulthood 50% calorically restricted and maintained at 90% of normal body weight for 60 d. HF animals showed increased percent body fat compared with AL and FR animals despite equivalent body weights. HF animals showed alterations in the balance of adipose tissue lipogenic (FAS, LPL) and lipolytic (HSL) gene expression that may underlie their propensity to maintain fat stores under caloric restriction.
Collapse
|
35
|
Ho JN, Son ME, Lim WC, Lim ST, Cho HY. Germinated brown rice extract inhibits adipogenesis through the down-regulation of adipogenic genes in 3T3-L1 adipocytes. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2013; 68:274-278. [PMID: 23907589 DOI: 10.1007/s11130-013-0366-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The aim of this study was to examine the anti-adipogenic effect of germinated brown rice methanol extract (GBR) in 3T3-L1 adipocytes. The GBR inhibited adipocyte differentiation was measured by Oil Red O staining and glycerol-3-phosphate dehydrogenase (GPDH) activity in a dose-dependent manner without initiating any cytotoxicity. The mRNA levels of adipogenic transcription factors such as CCAAT/enhancer binding protein (C/EBPα), proliferator-activated receptorγ (PPARγ), and sterol regulatory element-binding protein-1c (SREBP-1c), and adipogenic genes, such as fatty acid synthase (FAS), adipocyte fatty acid-binding protein (aP2), and lipoprotein lipase (LPL), were significantly down-regulated by treatment with GBR when compared to that of untreated control cells. Moreover, tumor necrosis factor-α (TNF-α) and interlukin-6 (IL-6) mRNA expressions were attenuated by GBR in mature adipocytes. These data suggest that GBR exhibits an anti-adipogenic effect through the suppression of adipogenesis in 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Jin-Nyoung Ho
- Department of Medical Nutrition, Kyung Hee University, Yongin, Korea
| | | | | | | | | |
Collapse
|
36
|
Kalanderian A, Abate N, Patrikeev I, Wei J, Vincent KL, Motamedi M, Saade GR, Bytautiene E. Pioglitazone therapy in mouse offspring exposed to maternal obesity. Am J Obstet Gynecol 2013; 208:308.e1-7. [PMID: 23313309 DOI: 10.1016/j.ajog.2013.01.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 12/07/2012] [Accepted: 01/09/2013] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Pioglitazone (PIO), an antidiabetic drug of the thiazolidinedione family, improves glucose and lipid metabolism in muscle, adipose, and liver tissues via peroxisome proliferator-activated receptor gamma activation. We hypothesize that PIO therapy will improve the metabolic status of offspring exposed to maternal obesity in a mouse model developmentally programmed for metabolic syndrome. STUDY DESIGN CD-1 female mice were fed a high-fat diet for 3 months prior to breeding and throughout pregnancy and lactation. The pups were weaned to a standard-fat diet. Offspring were randomly assigned to receive 40 mg/kg of PIO in 0.5% of methyl cellulose or 0.5% methyl cellulose by daily oral gavage for 2 weeks. The pre- and posttreatment total body weights of the pups were recorded. Visceral and subcutaneous adipose tissue were evaluated using microcomputed tomography. Serum analytes were measured. After treatment, minimally invasive microendoscopic fluorescence confocal imaging and intraperitoneal glucose tolerance tests were performed. The data were analyzed using appropriate statistical tests (significance, P < .05). RESULTS PIO therapy resulted in lower total body weight and lower visceral adipose tissue gain and increased subcutaneous adipose tissue. PIO significantly lowered triglycerides, insulin levels, and homeostasis model assessment of insulin resistance in males and fasting glucose in females. There was a trend toward larger adipocyte size. CONCLUSION Short-term PIO therapy in the offspring of obese mothers attenuates metabolic changes associated with the developmental programming of metabolic syndrome. These novel data suggest a potential role for drugs that activate peroxisome proliferator-activated receptor gamma receptors to prevent metabolic syndrome in the adult offspring at risk to develop metabolic alterations.
Collapse
|
37
|
Muhlhausler BS, Gugusheff JR, Ong ZY, Vithayathil MA. Nutritional approaches to breaking the intergenerational cycle of obesity. Can J Physiol Pharmacol 2013; 91:421-8. [PMID: 23745987 DOI: 10.1139/cjpp-2012-0353] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The link between poor maternal nutrition and an increased burden of disease in subsequent generations has been widely demonstrated in both human and animal studies. Historically, the nutritional challenges experienced by pregnant and lactating women were largely those of insufficient calories and severe micronutrient deficiencies. More recently, however, Western societies have been confronted with a new nutritional challenge; that of maternal obesity and excessive maternal intake of calories, fat, and sugar. Exposure of the developing fetus and infant to this obesogenic environment results in an increased risk of obesity and metabolic disease later in life. Furthermore, increased caloric, fat, and sugar intake can occur in conjunction with micronutrient deficiency, which may further exacerbate these programming effects. In light of the current epidemic of obesity and metabolic disease, attention has now turned to identifying nutritional interventions for breaking this intergenerational obesity cycle. In this review, we discuss the approaches that have been explored to date and highlight the need for further research.
Collapse
Affiliation(s)
- Beverly S Muhlhausler
- FOODplus Research Centre, School of Agriculture Food and Wine, Waite Main Building, The University of Adelaide, SA 5064, Australia.
| | | | | | | |
Collapse
|
38
|
Yan J, Gan L, Yang H, Sun C. The proliferation and differentiation characteristics of co-cultured porcine preadipocytes and muscle satellite cells in vitro. Mol Biol Rep 2012; 40:3197-202. [PMID: 23271122 DOI: 10.1007/s11033-012-2395-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 12/17/2012] [Indexed: 11/26/2022]
Abstract
To explore the proliferation and differentiation characteristics of co-cultured porcine preadipocytes and muscle satellite cells, preadipocytes and muscle satellite cells were isolated from the healthy nascent landrace. Oil Red O stain and desmin immunohistochemistry were used to identify the two solo-cultured cells. Methyl thiazolyl tetrazolium (MTT) colorimetric assay was used to detect the proliferation characteristic of co-cultured cells, and the expression level of differentiation marker genes lipoprotein lipase (LPL), peroxisome proliferator-activated receptors (PPARγ), myogenic factor 5 (Myf5), myogenin (MyoG) were analyzed with reverse transcription PCR (RT-PCR) and western blot. The success of co-culture system was proved. In the co-cultured cells, slight lipid droplets were observed and appeared more slowly. The polykaryocytes fused into myotubes in co-cultured cells were less and relatively slow than that in solo myocytes. After fusion, the proliferation rate of co-cultured cells was higher than that in the solo-cultured muscle satellite cells (P < 0.01), and the duration were also longer. On day 5 and 10, the expression of the marker genes in earlier stage of cell differentiation (LPL and Myf5) were lower than those in the solo-cultured cells (P < 0.01) (except LPL gene at day 5). Moreover, the expression of intermediate and advanced stages' maker genes (PPARγ2 and MyoG) were hardly detectable at day 5, but increased significantly on day 10 (P < 0.01). These results confirm that the co-culture system could facilitate the cells' growth and proliferation, meanwhile, inhibited the cell differentiation.
Collapse
Affiliation(s)
- Jun Yan
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | | | | | | |
Collapse
|
39
|
Flynn ER, Alexander BT, Lee J, Hutchens ZM, Maric-Bilkan C. High-fat/fructose feeding during prenatal and postnatal development in female rats increases susceptibility to renal and metabolic injury later in life. Am J Physiol Regul Integr Comp Physiol 2012; 304:R278-85. [PMID: 23255587 DOI: 10.1152/ajpregu.00433.2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Accumulating evidence suggests that both an adverse prenatal and early postnatal environment increase susceptibility to renal and metabolic dysfunction later in life; however, whether exposure to adverse conditions during both prenatal and postnatal development act synergistically to potentiate the severity of renal and metabolic injury remains unknown. Sprague-Dawley rats were fed either a standard diet or a diet high in fat/fructose throughout pregnancy and lactation. After being weaned, female offspring were randomized to either standard diet or the high-fat/high-fructose diet, resulting in the following treatment groups: NF-NF, offspring of mothers fed a standard diet and fed a standard diet postnatally; NF-HF, offspring of mothers fed a standard diet and fed a high-fat/fructose diet postnatally; HF-NF, offspring of mothers fed a high-fat/fructose diet and fed a standard diet postnatally; HF-HF, offspring of mothers fed a high-fat/fructose diet and fed a high-fat/fructose diet postnatally. At the time of euthanasia (17 wk of age), HF-HF offspring weighed 30% more and had 110% more visceral fat than NF-NF offspring. The HF-HF offspring also had elevated blood glucose levels, glucose intolerance, 286% increase in urine albumin excretion, and 60% increase in glomerulosclerosis compared with NF-NF. In addition, HF-HF offspring exhibited a 100% increase in transforming growth factor-β protein expression and 116% increase in the abundance of infiltrated macrophages compared with the NF-NF offspring. These observations suggest that high-fat/fructose feeding during prenatal and throughout postnatal life increases the susceptibility to renal and metabolic injury later in life.
Collapse
Affiliation(s)
- Elizabeth R Flynn
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | | | | | | | | |
Collapse
|
40
|
PPARγ2 Pro12Ala polymorphism is associated with improved lipoprotein lipase functioning in adipose tissue of insulin resistant obese women. Gene 2012; 511:404-10. [PMID: 23036713 DOI: 10.1016/j.gene.2012.09.057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 07/31/2012] [Accepted: 09/12/2012] [Indexed: 11/20/2022]
Abstract
Lipoprotein lipase (LPL) plays a pivotal role in lipid metabolism, contributes to metabolic disorders related to insulin action and body weight regulation, and is influenced by inflammation. The Pro12Ala polymorphism of the peroxisome proliferator-activated receptor (PPAR)γ2 gene seems to influence LPL functioning, but its role in obesity and insulin resistance status, which usually coexist in the clinical setting, has not been explored. Our aim was to analyze the association of obesity and insulin resistance with adipose LPL activity and expression, and the influence of the PPARγ2 Pro12Ala polymorphism. A cross-sectional study was conducted in 58 reproductive-age women who underwent elective abdominal surgery. Free-fatty acids, glucose, insulin, and selected adipokines were measured in fasting blood samples. DNA was isolated and the polymorphism genotyped. Biopsies of abdominal subcutaneous adipose tissue obtained during surgery were used to determine enzymatic LPL activity and expression; and expression of selected cytokines. Overweight/obese women presented lower LPL activity (P=0.022) and higher circulating TNF-α (P=0.020) than controls. Insulin resistant women also showed borderline lower LPL activity than non-resistant (P=0.052), but adiposity and inflammatory molecules were comparable. Nevertheless, LPL activity was higher in Pro12Ala carriers than in non-carriers after adjusting for obesity, insulin resistance and inflammation. Likewise, adipose LPL expression was increased in carriers while expression of cytokines was decreased. Our data suggest that insulin resistance is associated with low adipose LPL activity independently of obesity, but the PPARγ2 Pro12Ala polymorphism seems to protect the LPL functioning of obese insulin resistant women, likely through regulating inflammation in adipose tissue.
Collapse
|
41
|
Poston L. Maternal obesity, gestational weight gain and diet as determinants of offspring long term health. Best Pract Res Clin Endocrinol Metab 2012; 26:627-39. [PMID: 22980045 DOI: 10.1016/j.beem.2012.03.010] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This review addresses the increasingly prolific literature from studies in man and animals suggesting that maternal obesity, a diet rich in calories or excess gestational weight gain may, through perturbation of the intrauterine environment, lead to lifelong risk of obesity and related disorders in the child. In addressing maternal- child obesity relationships it remains a challenge to distinguish the influence of the intrauterine environment from the contribution of shared genetic traits, and to adequately adjust for postnatal determinants of childhood obesity. Studies in genetically identical rodents convincingly show that maternal obesity, as well as elements of a hypercalorific diet can permanently influence offspring risk of obesity, and are these are supported by studies in larger mammals. Importantly, dissection of the mechanism in animals has led to description of novel interactive pathways between maternal environment and fetus which are amenable to investigation in humans.
Collapse
Affiliation(s)
- Lucilla Poston
- Division of Women's Health, Women's Health Academic Centre, King's College London, St. Thomas' Hospital, London, United Kingdom.
| |
Collapse
|
42
|
Current world literature. Curr Opin Lipidol 2012; 23:156-63. [PMID: 22418573 DOI: 10.1097/mol.0b013e3283521229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
43
|
Finalism in Darwinian and Lamarckian Evolution: Lessons from Epigenetics and Developmental Biology. Evol Biol 2012. [DOI: 10.1007/s11692-012-9163-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
44
|
Jackson CM, Alexander BT, Roach L, Haggerty D, Marbury DC, Hutchens ZM, Flynn ER, Maric-Bilkan C. Exposure to maternal overnutrition and a high-fat diet during early postnatal development increases susceptibility to renal and metabolic injury later in life. Am J Physiol Renal Physiol 2011; 302:F774-83. [PMID: 22160775 DOI: 10.1152/ajprenal.00491.2011] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Overnutrition during pre- and postnatal development both confer increased susceptibility to renal and metabolic risks later in life; however, whether they have an additive effect on the severity of renal and metabolic injury remains unknown. The present study tested the hypothesis that a combination of a pre- and postnatal diet high in fat/fructose would exacerbate renal and metabolic injury in male offspring later in life. Male offspring born to high fat/high-fructose-fed mothers and fed a high-fat/high-fructose diet postnatally (HF-HF) had increased urine albumin excretion (450%), glomerulosclerosis (190%), and tubulointerstitial fibrosis (101%) compared with offspring born to mothers fed a standard diet and fed a standard diet postnatally (NF-NF). No changes in blood pressure or glomerular filtration were observed between any of the treatment groups. The HF-HF offspring weighed ∼23% more than offspring born to mothers fed a high-fat/high-fructose diet and fed a normal diet postnatally (HF-NF), as well as offspring born to mothers fed a standard diet regardless of their postnatal diet. The HF-HF rats also had increased (and more variable) blood glucose levels over 12 wk of being fed a high-fat/high-fructose diet. A combination of exposure to a high-fat/high-fructose diet in utero and postnatally increased plasma insulin levels by 140% compared with NF-NF offspring. Our data suggest that the combined exposure to overnutrition during fetal development and early postnatal development potentiate the susceptibility to renal and metabolic disturbances later in life.
Collapse
Affiliation(s)
- Colette M Jackson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Battista MC, Hivert MF, Duval K, Baillargeon JP. Intergenerational cycle of obesity and diabetes: how can we reduce the burdens of these conditions on the health of future generations? EXPERIMENTAL DIABETES RESEARCH 2011; 2011:596060. [PMID: 22110473 PMCID: PMC3205776 DOI: 10.1155/2011/596060] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 09/05/2011] [Accepted: 09/06/2011] [Indexed: 12/22/2022]
Abstract
Prepregnancy overweight or obesity and excessive gestational weight gain have been associated with increased risk of maternal and neonatal complications. Moreover, offspring from obese women are more likely to develop obesity, diabetes mellitus, and cardiovascular diseases in their lifetime. Gestational diabetes mellitus (GDM) is one of the most common complications associated with obesity and appears to have a direct impact on the future metabolic health of the child. Fetal programming of metabolic function induced by obesity and GDM may have intergenerational effect and thus perpetuate the epidemic of cardiometabolic conditions. The present paper thus aims at discussing the impact of maternal obesity and GDM on the developmental programming of obesity and metabolic disorders in the offspring. The main interventions designed to reduce maternal obesity and GDM and their ability to break the vicious circle that perpetuates the transmission of obesity and metabolic conditions to the next generations are also addressed.
Collapse
Affiliation(s)
- Marie-Claude Battista
- Division of Endocrinology, Department of Medicine, University of Sherbrooke, Sherbrooke, QC, Canada J1H 5N4
| | - Marie-France Hivert
- Division of Endocrinology, Department of Medicine, University of Sherbrooke, Sherbrooke, QC, Canada J1H 5N4
| | - Karine Duval
- Division of Endocrinology, Department of Medicine, University of Sherbrooke, Sherbrooke, QC, Canada J1H 5N4
| | - Jean-Patrice Baillargeon
- Division of Endocrinology, Department of Medicine, University of Sherbrooke, Sherbrooke, QC, Canada J1H 5N4
| |
Collapse
|