1
|
Rigal E, Josse M, Greco C, Rosenblatt N, Rochette L, Guenancia C, Vergely C. Short-Term Postnatal Overfeeding Induces Long-Lasting Cardiometabolic Syndrome in Mature and Old Mice Associated with Increased Sensitivity to Myocardial Infarction. Mol Nutr Food Res 2024; 68:e2400136. [PMID: 38937861 DOI: 10.1002/mnfr.202400136] [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: 02/21/2024] [Revised: 05/28/2024] [Indexed: 06/29/2024]
Abstract
SCOPE Perinatal nutritional disturbances may "program" an increased cardio-metabolic risk in adulthood; however, few experimental studies have explored their effects on mature and/or old animal. This study aims to investigate the influence of postnatal overfeeding (PNOF) on cardiac function, sensitivity to ischemia-reperfusion (I-R) injury in vivo, glucose metabolism, and metabolic profile of pericardial adipose tissue (PAT) in young (4 months), adult (6 months), old (12 months), and very old (18 months) male mice. METHODS AND RESULTS Two days after birth, PNOF is induced by adjusting the litter size of C57BL/6 male mice to three pups/mother, while the normally fed (NF) control group is normalized to nine pups/mother. After weaning, all mice have free access to standard diet. Glucose/insulin tests and in vivo myocardial I-R injury are conducted on mice aged from 2 to 12 months, while echocardiography is performed at all ages up to 18 months. PNOF mice exhibit an early and persistent 10-20% increase in body weight and a 10% decrease in left ventricular ejection fraction throughout their lifespan. In PNOF mice aged 4, 6, and 12 months, glucose intolerance and insulin resistance are observed, as well as a 27-34% increase in infarct size. This is accompanied by a higher PAT mass with increased inflammatory status. CONCLUSION Short-term PNOF results in nutritional programming, inducing long-lasting alterations in glucose metabolism and cardiac vulnerability in male mice, lasting up to 12 months.
Collapse
Affiliation(s)
- Eve Rigal
- Research Team: Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, Dijon, 21000, France
| | - Marie Josse
- Research Team: Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, Dijon, 21000, France
| | - Camille Greco
- Research Team: Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, Dijon, 21000, France
| | - Nathalie Rosenblatt
- Division of Angiology, Heart and Vessel Department, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, 1011, Switzerland
| | - Luc Rochette
- Research Team: Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, Dijon, 21000, France
| | - Charles Guenancia
- Research Team: Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, Dijon, 21000, France
- Cardiology Department, University Hospital of Dijon, Dijon, 21000, France
| | - Catherine Vergely
- Research Team: Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, Dijon, 21000, France
| |
Collapse
|
2
|
Iskakov NG, Anikina TA, Nikolaev TI, Krylova AV, Zefirov TL. Effect of Adrenoreceptor Stimulation on Peptidergic Regulation of Cardiac Activity in Newborn Rats. Bull Exp Biol Med 2023; 176:9-13. [PMID: 38091133 DOI: 10.1007/s10517-023-05957-1] [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: 02/14/2023] [Indexed: 12/19/2023]
Abstract
We studied the effect of adrenoreceptor stimulation on the frequency of spontaneous activity and amplitude-time parameters of isometric contraction of the atrial myocardial strips from newborn rats, as well as the effect of Y receptor stimulation against the background of adrenoreceptor activation. After addition of Y1,5 receptor agonist [Leu31, Pro34] NPY (10-7 M), a tendency to a decrease in the effect of β1,2-adrenoreceptor agonist isoproterenol (10-5 M) on the frequency of spontaneous activity and atrial myocardial contractility was observed. The age-related features of the effect of NPY on the frequency of spontaneous activity and contractility of myocardial strips from newborn and adult rats were revealed.
Collapse
Affiliation(s)
- N G Iskakov
- Department of Human Health Protection, Kazan (Volga region) Federal University, Kazan, Republic of Tatarstan, Russia.
- Department of Medical and Biological Disciplines, Volga Region State University of Physical Culture, Sports and Tourism, Kazan, Republic of Tatarstan, Russia.
| | - T A Anikina
- Department of Human Health Protection, Kazan (Volga region) Federal University, Kazan, Republic of Tatarstan, Russia
| | - T I Nikolaev
- Department of Human Health Protection, Kazan (Volga region) Federal University, Kazan, Republic of Tatarstan, Russia
| | - A V Krylova
- Department of Human Health Protection, Kazan (Volga region) Federal University, Kazan, Republic of Tatarstan, Russia
| | - T L Zefirov
- Department of Human Health Protection, Kazan (Volga region) Federal University, Kazan, Republic of Tatarstan, Russia
| |
Collapse
|
3
|
Tejera-Muñoz A, Guerra-Menéndez L, Amor S, González-Hedström D, García-Villalón ÁL, Granado M. Postnatal Overfeeding during Lactation Induces Endothelial Dysfunction and Cardiac Insulin Resistance in Adult Rats. Int J Mol Sci 2023; 24:14443. [PMID: 37833890 PMCID: PMC10572650 DOI: 10.3390/ijms241914443] [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: 08/14/2023] [Revised: 09/13/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023] Open
Abstract
Early overnutrition is associated with cardiometabolic alterations in adulthood, likely attributed to reduced insulin sensitivity due to its crucial role in the cardiovascular system. This study aimed to assess the long-term effects of early overnutrition on the development of cardiovascular insulin resistance. An experimental childhood obesity model was established using male Sprague Dawley rats. Rats were organized into litters of 12 pups/mother (L12-Controls) or 3 pups/mother (L3-Overfed) at birth. After weaning, animals from L12 and L3 were housed three per cage and provided ad libitum access to food for 6 months. L3 rats exhibited elevated body weight, along with increased visceral, subcutaneous, and perivascular fat accumulation. However, heart weight at sacrifice was reduced in L3 rats. Furthermore, L3 rats displayed elevated serum levels of glucose, leptin, adiponectin, total lipids, and triglycerides compared to control rats. In the myocardium, overfed rats showed decreased IL-10 mRNA levels and alterations in contractility and heart rate in response to insulin. Similarly, aortic tissue exhibited modified gene expression of TNFα, iNOS, and IL-6. Additionally, L3 aortas exhibited endothelial dysfunction in response to acetylcholine, although insulin-induced relaxation remained unchanged compared to controls. At the molecular level, L3 rats displayed reduced Akt phosphorylation in response to insulin, both in myocardial and aortic tissues, whereas MAPK phosphorylation was elevated solely in the myocardium. Overfeeding during lactation in rats induces endothelial dysfunction and cardiac insulin resistance in adulthood, potentially contributing to the cardiovascular alterations observed in this experimental model.
Collapse
Affiliation(s)
- Antonio Tejera-Muñoz
- Research Support Unit, Hospital General La Mancha Centro, 13600 Alcázar de San Juan, Spain;
- Instituto de Investigación de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Lucía Guerra-Menéndez
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain;
| | - Sara Amor
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (S.A.); (D.G.-H.); (Á.L.G.-V.)
| | - Daniel González-Hedström
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (S.A.); (D.G.-H.); (Á.L.G.-V.)
| | - Ángel Luis García-Villalón
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (S.A.); (D.G.-H.); (Á.L.G.-V.)
| | - Miriam Granado
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (S.A.); (D.G.-H.); (Á.L.G.-V.)
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
4
|
Mechanisms of Modulation of Adrenergic Regulation of Spontaneous Activity Rate and Atrial Myocardial Contractility in Early Postnatal Ontogeny in Rats. Bull Exp Biol Med 2023; 174:295-298. [PMID: 36723731 DOI: 10.1007/s10517-023-05694-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2022] [Indexed: 02/02/2023]
Abstract
We studied combined effect of the β1,2-adrenoreceptor agonist isoproterenol and the Y1,5 receptor agonist [Leu31, Pro34]neuropeptide Y on the frequency of spontaneous activity and myocardial contractility in 21- and 100-day-old rats. Isoproterenol increased the frequency of spontaneous activity and reduced the main parameters of isometric contraction of the atrial myocardium. When [Leu31, Pro34]neuropeptide Y was added, the frequency of spontaneous activity and the negative inotropic and the positive chronotropic effects of isoproterenol were reduced in 100-day-old rats. In 21-day-olds rats, a tendency to a decrease in the effect of isoproterenol was observed.
Collapse
|
5
|
Litter Size Reduction as a Model of Overfeeding during Lactation and Its Consequences for the Development of Metabolic Diseases in the Offspring. Nutrients 2022; 14:nu14102045. [PMID: 35631188 PMCID: PMC9145223 DOI: 10.3390/nu14102045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 02/06/2023] Open
Abstract
Overfeeding during lactation has a deleterious impact on the baby’s health throughout life. In humans, early overnutrition has been associated with higher susceptibility to obesity and metabolic disorders in childhood and adulthood. In rodents, using a rodent litter size reduction model (small litter) to mimic early overfeeding, the same metabolic profile has been described. Therefore, the rodent small litter model is an efficient tool to investigate the adaptive mechanisms involved in obesogenesis. Besides central and metabolic dysfunctions, studies have pointed to the contribution of the endocrine system to the small litter phenotype. Hormones, especially leptin, insulin, and adrenal hormones, have been associated with satiety, glucose homeostasis, and adipogenesis, while hypothyroidism impairs energy metabolism, favoring obesity. Behavioral modifications, hepatic metabolism changes, and reproductive dysfunctions have also been reported. In this review, we update these findings, highlighting the interaction of early nutrition and the adaptive features of the endocrine system. We also report the sex-related differences and epigenetic mechanisms. This model highlights the intense plasticity during lactation triggering many adaptive responses, which are the basis of the developmental origins of health and disease (DOHaD) concept. Our review demonstrates the complexity of the adaptive mechanisms involved in the obesity phenotype promoted by early overnutrition, reinforcing the necessity of adequate nutritional habits during lactation.
Collapse
|
6
|
Rodrigues VST, Moura EG, Peixoto TC, Soares P, Lopes BP, Bertasso IM, Silva BS, Cabral S, Kluck GEG, Atella GC, Trindade PL, Daleprane JB, Oliveira E, Lisboa PC. The model of litter size reduction induces long-term disruption of the gut-brain axis: An explanation for the hyperphagia of Wistar rats of both sexes. Physiol Rep 2022; 10:e15191. [PMID: 35146951 PMCID: PMC8831958 DOI: 10.14814/phy2.15191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 12/10/2021] [Accepted: 01/04/2022] [Indexed: 04/26/2023] Open
Abstract
The gut microbiota affects the host's metabolic phenotype, impacting health and disease. The gut-brain axis unites the intestine with the centers of hunger and satiety, affecting the eating behavior. Deregulation of this axis can lead to obesity onset. Litter size reduction is a well-studied model for infant obesity because it causes overnutrition and programs for obesity. We hypothesize that animals raised in small litters (SL) have altered circuitry between the intestine and brain, causing hyperphagia. We investigated vagus nerve activity, the expression of c-Fos, brain-derived neurotrophic factor (BDNF), gastrointestinal (GI) hormone receptors, and content of bacterial phyla and short-chain fatty acids (SCFAs) in the feces of adult male and female Wistar rats overfed during lactation. On the 3rd day after birth, litter size was reduced to 3 pups/litter (SL males or SL females) until weaning. Controls had normal litter size (10 pups/litter: 5 males and 5 females). The rats were killed at 5 months of age. The male and female offspring were analyzed separately. The SL group of both sexes showed higher food consumption and body adiposity than the respective controls. SL animals presented dysbiosis (increased Firmicutes, decreased Bacteroidetes) and had increased vagus nerve activity. Only the SL males had decreased hypothalamic GLP-1 receptor expression, while only the SL females had lower acetate and propionate in the feces and higher CCK receptor expression in the hypothalamus. Thus, overfeeding during lactation differentially changes the gut-brain axis, contributing to hyperphagia of the offspring of both sexes.
Collapse
Affiliation(s)
- Vanessa S. T. Rodrigues
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - Egberto G. Moura
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - Thamara C. Peixoto
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - Patricia N. Soares
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - Bruna P. Lopes
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - Iala M. Bertasso
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - Beatriz S. Silva
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - S. S. Cabral
- Laboratory of Lipids and Lipoprotein BiochemistryBiochemistry InstituteFederal University of Rio de JaneiroRio de JaneiroBrazil
| | - G. E. G. Kluck
- Laboratory of Lipids and Lipoprotein BiochemistryBiochemistry InstituteFederal University of Rio de JaneiroRio de JaneiroBrazil
| | - G. C. Atella
- Laboratory of Lipids and Lipoprotein BiochemistryBiochemistry InstituteFederal University of Rio de JaneiroRio de JaneiroBrazil
| | - P. L. Trindade
- Laboratory for studies of Interactions between Nutrition and GeneticsNutrition InstituteRio de Janeiro State UniversityRio de JaneiroBrazil
| | - J. B. Daleprane
- Laboratory for studies of Interactions between Nutrition and GeneticsNutrition InstituteRio de Janeiro State UniversityRio de JaneiroBrazil
| | - Elaine Oliveira
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - Patricia Cristina Lisboa
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| |
Collapse
|
7
|
Huang F, Zhu P, Wang J, Chen J, Lin W. Postnatal overfeeding induces hepatic microRNA-221 expression and impairs the PI3K/AKT pathway in adult male rats. Pediatr Res 2021; 89:143-149. [PMID: 32305038 DOI: 10.1038/s41390-020-0877-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 03/14/2020] [Accepted: 03/19/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Increasing evidence suggests that postnatal overfeeding induces childhood obesity, which is strongly associated with metabolic syndrome. Insulin resistance is a risk factor for metabolic syndrome. MicroRNA-221 (miR-221) is involved in the development of obesity and has been reported to negatively regulate insulin sensitivity. However, the underlying mechanism remains unclear. METHODS Rats raised in small litters (SLs, three pups/dam, n = 10) and normal litters (NLs, 10 pups/dam, n = 10) were used to model early postnatal overfeeding and act as controls, respectively. miR-221 and proteins related to the phosphoinositide 3-kinases (PI3K)/protein kinase B (AKT) pathway were assessed in the liver. RESULTS Early postnatal overfeeding significantly increased body weight, visceral fat index, blood glucose, serum triglycerides, and the homeostasis model assessment of insulin resistance at 9 weeks. Real-time polymerase chain reaction (PCR) and western blot analysis revealed that postnatal overfeeding induced insulin receptor and insulin receptor substrate 2 expression, but decreased PI3K and AKT phosphorylation in the liver. Quantitative real-time PCR showed that hepatic miR-221 was significantly overexpressed in the SL group. CONCLUSIONS These results indicate that postnatal overfeeding induces hepatic miR-221 overexpression and impairs the PI3K/AKT signal pathway, which may cause insulin resistance. IMPACT We first report postnatal overfeeding induces hepatic miR-221 expression. Postnatal overfeeding impairs PI3K/AKT pathway in the liver of adult rats. Postnatal overfeeding induces obesity and high blood glucose. Avoidance of overfeeding during early postnatal life may prevent obesity and T2DM.
Collapse
Affiliation(s)
- Fang Huang
- School of Public Health, Fujian Medical University, Fuzhou, Fujian, China.
| | - Pingping Zhu
- School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Jingwen Wang
- School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Jie Chen
- School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Wenting Lin
- School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
8
|
Programming of Cardiovascular Dysfunction by Postnatal Overfeeding in Rodents. Int J Mol Sci 2020; 21:ijms21249427. [PMID: 33322275 PMCID: PMC7763005 DOI: 10.3390/ijms21249427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 11/17/2022] Open
Abstract
Nutritional environment in the perinatal period has a great influence on health and diseases in adulthood. In rodents, litter size reduction reproduces the effects of postnatal overnutrition in infants and reveals that postnatal overfeeding (PNOF) not only permanently increases body weight but also affects the cardiovascular function in the short- and long-term. In addition to increased adiposity, the metabolic status of PNOF rodents is altered, with increased plasma insulin and leptin levels, associated with resistance to these hormones, changed profiles and levels of circulating lipids. PNOF animals present elevated arterial blood pressure with altered vascular responsiveness to vasoactive substances. The hearts of overfed rodents exhibit hypertrophy and elevated collagen content. PNOF also induces a disturbance of cardiac mitochondrial respiration and produces an imbalance between oxidants and antioxidants. A modification of the expression of crucial genes and epigenetic alterations is reported in hearts of PNOF animals. In vivo, a decreased ventricular contractile function is observed during adulthood in PNOF hearts. All these alterations ultimately lead to an increased sensitivity to cardiac pathologic challenges such as ischemia-reperfusion injury. Nevertheless, caloric restriction and physical exercise were shown to improve PNOF-induced cardiac dysfunction and metabolic abnormalities, drawing a path to the potential therapeutic correction of early nutritional programming.
Collapse
|
9
|
Overweight during lactation and its implications for biometric, nutritional and cardiovascular parameters of young and adult male and female rats. J Nutr Sci 2020; 9:e27. [PMID: 32742644 PMCID: PMC7372176 DOI: 10.1017/jns.2020.21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 05/26/2020] [Accepted: 06/03/2020] [Indexed: 01/08/2023] Open
Abstract
Litter size reduction can induce early overnourishment, being an attractive experimental model to study short- and long-term consequences of childhood obesity. Epidemiological data indicate sex differences regarding cardiometabolic disorders and hypertrophic cardiomyopathy. The present study aimed to describe biometric, nutritional and cardiovascular changes related to neonatal overweight promoted by litter size reduction in young and adult Wistar rats of both sexes. Litter adjustment to eight or four pups/mother (1:1 male-to-female ratio) gave, respectively, control and overweight groups. Body mass, food intake, haemodynamic and echocardiographic parameters and cardiorespiratory capacity were evaluated at postnatal days 30 and 150. Diminished litters were correlated with higher body mass and weight gain (12 %) during lactation, validating the experimental model of neonatal overweight. Soon after weaning male (16 %) and female (25 %) offspring of these litters presented a lower food intake than their respective control, without differences in body mass. Adult males from reduced litters presented higher abdominal circumference (7 %), systolic blood pressure (10 %), interventricular septum thickness (15 %) and relative wall thickness (15 %) compared with their respective control. Rats' performance on the maximal effort ergometer test was not affected by neonatal overweight. Data suggest the occurrence of catch-down growth and hypophagia in male and female rats submitted to neonatal overweight. However, only male rats presented haemodynamic and cardiac structural changes. These findings are crucial to personalised/gender medicine.
Collapse
Key Words
- AC, abdominal circumference
- Cardiovascular system
- Child development
- IVS, interventricular septum thickness
- IVSd, interventricular septum thickness diastole
- IVSs, interventricular septum thickness systole
- LVID, left ventricle internal diameter
- LVIDd, left ventricle internal diameter diastole
- LVPW, left ventricle posterior wall thickness
- LVPWd, left ventricle posterior wall thickness diastole
- LVPWs, left ventricle posterior wall thickness systole
- Lactation
- NAL, nose-to-anus length
- Overweight
- Sex characteristics
- TC, thoracic circumference
Collapse
|
10
|
Feeding circuit development and early-life influences on future feeding behaviour. Nat Rev Neurosci 2019; 19:302-316. [PMID: 29662204 DOI: 10.1038/nrn.2018.23] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A wide range of maternal exposures - undernutrition, obesity, diabetes, stress and infection - are associated with an increased risk of metabolic disease in offspring. Developmental influences can cause persistent structural changes in hypothalamic circuits regulating food intake in the service of energy balance. The physiological relevance of these alterations has been called into question because maternal impacts on daily caloric intake do not persist to adulthood. Recent behavioural and epidemiological studies in humans provide evidence that the relative contribution of appetitive traits related to satiety, reward and the emotional aspects of food intake regulation changes across the lifespan. This Opinion article outlines a neurodevelopmental framework to explore the possibility that crosstalk between developing circuits regulating different modalities of food intake shapes future behavioural responses to environmental challenges.
Collapse
|
11
|
de Moura Freitas C, Nascimento LCPD, Braz GRF, Andrade-Silva SC, Lima-Junior NC, de Araujo Silva T, Fernandes MP, Ferreira DJS, Lagranha CJ. Mitochondrial impairment following neonatal overfeeding: A comparison between normal and ischemic-reperfused hearts. J Cell Biochem 2019; 120:7341-7352. [PMID: 30368910 DOI: 10.1002/jcb.28009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 10/10/2018] [Indexed: 01/24/2023]
Abstract
Overweight and obesity are established factors underpin several metabolic impairments, including the cardiovascular. Although the diversity of factors involved in overweight/obesity-induced cardiovascular diseases, mitochondria has been highlighted due to its role in cardiac metabolism. As obesity can be originated in early postnatal life, the current study evaluates the effects of neonatal overfeeding on the cardiac mitochondrial bioenergetics and oxidative balance in rats that underwent an ischemia-reperfusion insult. Seventy-two hours after delivery, Wistar rat litters were randomly assigned into the control (C; nine pups per mother) and the Overfed (OF; three pups per mother) groups throughout the lactation period. At weaning, male offspring were fed with laboratory chow ad libitum until sacrifice at 30 and 60 days of life. Mitochondrial heart bioenergetics and oxidative balance showed to be deeply affected by neonatal overfeeding at both ages. Interestingly, after ischemia-reperfusion insult I/R (Langendorff or mineral oil incubation), most parameters evaluated in OF animals were not influenced by additional ischemic-reperfusion injury. Our findings demonstrated that suckling overfeeding deregulates cardiac mitochondrial alike to ischemia-reperfusion insult by disengaging electrical mitochondrial coupling and potentiate oxidative stress, wherein the neonatal overfeeding shows to be so detrimental as I/R. Our findings support the concept that nutritional insults in the critical development periods increase the risk for cardiovascular disease and mitochondria impairments throughout life while oxidative damage change between molecular targets.
Collapse
Affiliation(s)
- Cristiane de Moura Freitas
- Laboratory of Biochemistry and Exercise Biochemistry, Biochemistry and Physiology Graduate Program, CAV-Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Luciana Caroline Paulino do Nascimento
- Laboratory of Biochemistry and Exercise Biochemistry, Biochemistry and Physiology Graduate Program, CAV-Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Glauber Rudá Feitoza Braz
- Laboratory of Biochemistry and Exercise Biochemistry, Neuropsychiatry and Behavioral Science Graduate Program, CAV-Federal University of Pernambuco, Recife, Brazil
| | - Severina Cassia Andrade-Silva
- Laboratory of Biochemistry and Exercise Biochemistry, Biochemistry and Physiology Graduate Program, CAV-Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Nelson C Lima-Junior
- Department of Physical Education and Sports Science, Laboratory of Biochemistry and Exercise Biochemistry, CAV- Federal University of Pernambuco, Brazil
| | - Tercya de Araujo Silva
- Laboratory of Biochemistry and Exercise Biochemistry, Neuropsychiatry and Behavioral Science Graduate Program, CAV-Federal University of Pernambuco, Recife, Brazil
| | - Mariana Pinheiro Fernandes
- Department of Physical Education and Sports Science, Laboratory of Biochemistry and Exercise Biochemistry, CAV- Federal University of Pernambuco, Brazil
| | | | - Claudia Jacques Lagranha
- Laboratory of Biochemistry and Exercise Biochemistry, Biochemistry and Physiology Graduate Program, CAV-Federal University of Pernambuco, Recife, Pernambuco, Brazil.,Laboratory of Biochemistry and Exercise Biochemistry, Neuropsychiatry and Behavioral Science Graduate Program, CAV-Federal University of Pernambuco, Recife, Brazil.,Department of Physical Education and Sports Science, Laboratory of Biochemistry and Exercise Biochemistry, CAV- Federal University of Pernambuco, Brazil
| |
Collapse
|
12
|
Aylwin CF, Toro CA, Shirtcliff E, Lomniczi A. Emerging Genetic and Epigenetic Mechanisms Underlying Pubertal Maturation in Adolescence. JOURNAL OF RESEARCH ON ADOLESCENCE : THE OFFICIAL JOURNAL OF THE SOCIETY FOR RESEARCH ON ADOLESCENCE 2019; 29:54-79. [PMID: 30869843 DOI: 10.1111/jora.12385] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The adolescent transition begins with the onset of puberty which, upstream in the brain, is initiated by the gonadotropin-releasing hormone (GnRH) pulse generator that activates the release of peripheral sex hormones. Substantial research in human and animal models has revealed a myriad of cellular networks and heritable genes that control the GnRH pulse generator allowing the individual to begin the process of reproductive competence and sexual maturation. Here, we review the latest knowledge in neuroendocrine pubertal research with emphasis on genetic and epigenetic mechanisms underlying the pubertal transition.
Collapse
|
13
|
High maternal milk intake in the postnatal life reduces the incidence of breast cancer during adulthood in rats. J Dev Orig Health Dis 2019; 10:479-487. [DOI: 10.1017/s2040174418001071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
AbstractEnvironmental factors during perinatal life can lead to changes in the mammary gland, making it susceptible to cancer in adulthood. Breastfeeding has a special importance since it takes place at a critical period of growth and development of the newborn. We aimed to analyze if an appropriate lactation protects the offspring against mammary carcinogenesis during adult life and explore the mechanisms involved in the protective effect. One-day-old Sprague-Dawley female rats were randomly distributed in litters of three (L3), eight (L8) or 12 (L12) pups per dam, to induce a differential consumption of breast milk. At 55 days of age, the animals were treated with a single dose of dimethylbenzanthracene to study tumor latency, incidence and progression. Histological, immunohistochemical and Western blot studies were performed. We observed lower incidence and higher latency in L3 compared to the other groups. The mitotic index and expression of proliferating cell nuclear antigen (PCNA) was significantly augmented in tumors of L12 rats compared to L3 and L8, while the apoptotic index was augmented in tumors of L3 v. L12. Cleaved caspase 8 was significantly higher in tumors from L3 compared to L12. Tumors developed in L3 have a greater number of apoptotic bodies and a greater expression of caspase 8. These results demonstrate that the animals that maintained a higher intake of maternal milk (L3) presented lower incidence and greater tumor latency. Lower consumption of breast milk (L12) would increase tumor mitosis and the expression of PCNA, explaining the higher tumor incidence observed in this group.
Collapse
|
14
|
Enes-Marques S, Giusti-Paiva A. Litter size reduction accentuates maternal care and alters behavioral and physiological phenotypes in rat adult offspring. J Physiol Sci 2018; 68:789-798. [PMID: 29380149 PMCID: PMC10717135 DOI: 10.1007/s12576-018-0594-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/18/2018] [Indexed: 01/12/2023]
Abstract
Maternal behavior has a substantial impact on the behavioral, endocrine, and neural development of the pups. This study investigated the effect of altering the neonatal nutritional environment by modifying the litter size on maternal care and anxiety- and fear-like behaviors in rats during adulthood. On postnatal day (PND) 2, litters were adjusted to a small litter (SL) size of three pups per dam or normal litter (NL) size of 12 pups per dam. Maternal behaviors were scored daily during lactation (PND2-21). The weight gain, food intake, adiposity, and biochemical landmarks of offspring rats were evaluated. On PND60, performances in the open field, elevated plus-maze (EPM), and fear conditioning test were measured. The reduction of the litter size enhanced maternal care in lactating rats, increasing the arched-back posture and licking pups. SL offspring exhibited accelerated weight gain, hyperphagia, increased visceral fat mass, dyslipidemia, and hyperleptinemia in adulthood. The SL offspring of both sexes showed an increase in the anti-thigmotactic effect in the open field, an intact anxious-phenotype in the EPM, and a decrease in the time spent freezing during the fear-conditioning test, compared to NL. The neonatal environment as determined by litter size plays a crucial role in programming the adult metabolic phenotype as well as behavioral responses to stressful stimuli, with an impact on anxiety-like and fear behaviors. These behavioral changes in offspring may be, at least in part, a result of increased maternal care.
Collapse
Affiliation(s)
- Silvia Enes-Marques
- Laboratório de Fisiologia Translacional, Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas-MG, Campus Santa Clara, Av Jovino Fernandes Sales 2600, Prédio E Sala 300, Alfenas, MG, 37130-000, Brazil
| | - Alexandre Giusti-Paiva
- Laboratório de Fisiologia Translacional, Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas-MG, Campus Santa Clara, Av Jovino Fernandes Sales 2600, Prédio E Sala 300, Alfenas, MG, 37130-000, Brazil.
| |
Collapse
|
15
|
Tan CMJ, Green P, Tapoulal N, Lewandowski AJ, Leeson P, Herring N. The Role of Neuropeptide Y in Cardiovascular Health and Disease. Front Physiol 2018; 9:1281. [PMID: 30283345 PMCID: PMC6157311 DOI: 10.3389/fphys.2018.01281] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/24/2018] [Indexed: 12/20/2022] Open
Abstract
Neuropeptide Y (NPY) is an abundant sympathetic co-transmitter, widely found in the central and peripheral nervous systems and with diverse roles in multiple physiological processes. In the cardiovascular system it is found in neurons supplying the vasculature, cardiomyocytes and endocardium, and is involved in physiological processes including vasoconstriction, cardiac remodeling, and angiogenesis. It is increasingly also implicated in cardiovascular disease pathogenesis, including hypertension, atherosclerosis, ischemia/infarction, arrhythmia, and heart failure. This review will focus on the physiological and pathogenic role of NPY in the cardiovascular system. After summarizing the NPY receptors which predominantly mediate cardiovascular actions, along with their signaling pathways, individual disease processes will be considered. A thorough understanding of these roles may allow therapeutic targeting of NPY and its receptors.
Collapse
Affiliation(s)
- Cheryl M J Tan
- Oxford Cardiovascular Clinical Research Facility, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Peregrine Green
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre, University of Oxford, Oxford, United Kingdom
| | - Nidi Tapoulal
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre, University of Oxford, Oxford, United Kingdom
| | - Adam J Lewandowski
- Oxford Cardiovascular Clinical Research Facility, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Paul Leeson
- Oxford Cardiovascular Clinical Research Facility, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Neil Herring
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
16
|
Pinos H, Carrillo B, Díaz F, Chowen JA, Collado P. Differential vulnerability to adverse nutritional conditions in male and female rats: Modulatory role of estradiol during development. Front Neuroendocrinol 2018; 48:13-22. [PMID: 28754628 DOI: 10.1016/j.yfrne.2017.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/07/2017] [Accepted: 07/23/2017] [Indexed: 01/21/2023]
Abstract
Many studies have shown the importance of an adequate nutritional environment during development to optimally establish the neurohormonal circuits that regulate feeding behavior. Under- or over-nutrition during early stages of life can lead to alterations in the physiology and brain networks that control food intake, resulting in a greater vulnerability to suffer maladjustments in energy metabolism in adulthood. These alterations produced by under- or over-nourishment during development differ between males and females, as does the modulatory action that estradiol exerts on the alterations produced by malnutrition. Estradiol regulates metabolism and brain metabolic circuits through the same transcription factor pathway, STAT3, that leptin and ghrelin use to program feeding circuits. Although more research is needed to disentangle the actual role of estradiol during development on the programming of feeding circuits, a synergistic role together with leptin and/or ghrelin might be hypothesized.
Collapse
Affiliation(s)
- Helena Pinos
- Departamento de Psicobiología, Universidad Nacional de Educación a Distancia (UNED), C/ Juan del Rosal n° 10, 28040 Madrid, Spain; Instituto Mixto de Investigación-Escuela Nacional de Sanidad (IMIENS), Spain
| | - Beatriz Carrillo
- Departamento de Psicobiología, Universidad Nacional de Educación a Distancia (UNED), C/ Juan del Rosal n° 10, 28040 Madrid, Spain; Instituto Mixto de Investigación-Escuela Nacional de Sanidad (IMIENS), Spain
| | - Francisca Díaz
- Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Investigación Biomédica en Red (CIBER) de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Avda. Menéndez Pelayo, N° 65, 28009 Madrid, Spain
| | - Julie A Chowen
- Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Investigación Biomédica en Red (CIBER) de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Avda. Menéndez Pelayo, N° 65, 28009 Madrid, Spain
| | - Paloma Collado
- Departamento de Psicobiología, Universidad Nacional de Educación a Distancia (UNED), C/ Juan del Rosal n° 10, 28040 Madrid, Spain; Instituto Mixto de Investigación-Escuela Nacional de Sanidad (IMIENS), Spain.
| |
Collapse
|
17
|
Sánchez-García G, Del Bosque-Plata L, Hong E. Postnatal overnutrition affects metabolic and vascular function reflected by physiological and histological changes in the aorta of adult Wistar rats. Clin Exp Hypertens 2017; 40:452-460. [PMID: 29115861 DOI: 10.1080/10641963.2017.1392557] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Rigorous nutritional care during early life leads to healthy adulthood. Cardiovascular and metabolic disorders, the most prevalent clinical challenges worldwide, are epidemiologically linked to poor nutritional habits throughout life. We aimed to understand whether postnatal overnutrition (PO) initiated during lactation affects metabolic markers and vascular function later in life. To test this hypothetical effect, we studied a PO Wistar rat model based on adjusting litter size at the third day of age to three pups and eight for the control group (C). Systemic parameters such as body weight and food intake were significantly increased in adult rats, measured up to 36 weeks. Moreover, fat mass, triglycerides, insulin and systolic blood pressure were all significantly increased in the PO group. Furthermore, we assessed whether these alterations would affect morphological and functional parameters in isolated vessels. Consistent with systemic alterations of the vasculature, contraction of thoracic aortic rings, determined by dose-response curves to norepinephrine (NE), was significantly reduced in PO rats. Histological stains revealed that the relative area of collagen was higher and the elastic fiber density was lower in the distal rings of PO rats. Altogether, our results highlight the critical importance of having a healthy neonatal nutrition to prevent harmful metabolic and vascular alterations during adulthood.
Collapse
Affiliation(s)
- Gerardo Sánchez-García
- a Department of Pharmacobiology , Center for Research and Advanced Studies of the National Polytechnic Institute , Mexico City , Mexico.,b Department of Nutrigenetic and Nutrigenomic , National Institute of Genomic Medicine , Mexico City , Mexico
| | - Laura Del Bosque-Plata
- b Department of Nutrigenetic and Nutrigenomic , National Institute of Genomic Medicine , Mexico City , Mexico
| | - Enrique Hong
- a Department of Pharmacobiology , Center for Research and Advanced Studies of the National Polytechnic Institute , Mexico City , Mexico
| |
Collapse
|
18
|
Granado M, Amor S, Fernández N, Carreño-Tarragona G, Iglesias-Cruz MC, Martín-Carro B, Monge L, García-Villalón AL. Effects of early overnutrition on the renal response to Ang II and expression of RAAS components in rat renal tissue. Nutr Metab Cardiovasc Dis 2017; 27:930-937. [PMID: 28958692 DOI: 10.1016/j.numecd.2017.06.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 06/16/2017] [Accepted: 06/29/2017] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND AIMS The aim of this study was to analyze the effects of early overnutrition (EON) on the expression of the renin angiotensin aldosterone system (RAAS) components in renal cortex, renal arteries and renal perivascular adipose tissue (PVAT), as well as the vascular response of renal arteries to Angiotensin II (Ang II). METHODS AND RESULTS On birth day litters were adjusted to twelve (L12-control) or three (L3-overfed) pups per mother. Half of the animals were sacrificed at weaning (21 days old) and the other half at 5 months of age. Ang II-induced vasoconstriction of renal artery segments increased in young overfed rats and decreased in adult overfed rats. EON decreased the gene expression of angiotensinogen (Agt), Ang II receptors AT1 and AT2 and eNOS in renal arteries of young rats, while it increased the mRNA levels of AT-2 and ET-1 in adult rats. In renal PVAT EON up-regulated the gene expression of COX-2 and TNF-α in young rats and the mRNA levels of renin receptor both in young and in adult rats. On the contrary, Ang II receptors mRNA levels were downregulated at both ages. Renal cortex of overfed rats showed increased gene expression of Agt in adult rats and of AT1 in young rats. However the mRNA levels of AT1 were decreased in the renal cortex of overfed adult rats. CONCLUSION EON is associated with alterations in the vascular response of renal arteries to Ang II and changes in the gene expression of RAAS components in renal tissue.
Collapse
MESH Headings
- Adipose Tissue/drug effects
- Adipose Tissue/metabolism
- Adipose Tissue/physiopathology
- Age Factors
- Angiotensin II/pharmacology
- Animal Nutritional Physiological Phenomena
- Animals
- Cyclooxygenase 2/genetics
- Cyclooxygenase 2/metabolism
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Gene Expression Regulation
- Kidney/blood supply
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- Nutritional Status
- Overnutrition/genetics
- Overnutrition/metabolism
- Overnutrition/physiopathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
- Renal Artery/drug effects
- Renal Artery/metabolism
- Renal Artery/physiopathology
- Renin-Angiotensin System/drug effects
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
- Vasoconstriction/drug effects
- Vasoconstrictor Agents/pharmacology
Collapse
Affiliation(s)
- M Granado
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain.
| | - S Amor
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - N Fernández
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - G Carreño-Tarragona
- Servicio de Hematología, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - M C Iglesias-Cruz
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - B Martín-Carro
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - L Monge
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - A L García-Villalón
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
19
|
Carrillo B, Collado P, Díaz F, Chowen JA, Pérez-Izquierdo MÁ, Pinos H. Physiological and brain alterations produced by high-fat diet in male and female rats can be modulated by increased levels of estradiol during critical periods of development. Nutr Neurosci 2017; 22:29-39. [PMID: 28696162 DOI: 10.1080/1028415x.2017.1349574] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Overnutrition due to a high-fat diet (HFD) can increase the vulnerability of the metabolic system to maladjustments. Estradiol has an inhibitory role on food intake and this hormone has demonstrated to be a crucial organizer during brain development. OBJECTIVE Our aim was to determine whether increased levels of estradiol in the early postnatal period modulate the alterations in metabolism and brain metabolic circuits produced by overnutrition. METHODS Twenty-four male and 24 female Wistar rats were submitted to a HFD (34.9% fat) or a control diet (5% fat) from gestational day 6. From postnatal (P) 6 to P13, both control and HFD groups were administered a s.c. injection of vehicle or estradiol benzoate (0.4 mg/kg), resulting in eight experimental groups (n = 6 in each group). Body weight, food intake and subcutaneous, visceral, and brown fat pads were measured. Agouti-related peptide, neuropeptide Y, orexin, and proopiomelanocortin (POMC) were analyzed by quantitative real-time polymerase chain reaction assay and plasma estradiol levels were measured by ELISA. RESULTS Males fed a HFD showed an increase in body weight and the amount of visceral and subcutaneous fat, which was coincident with an increase in the number of kilocalories ingested. Neonatal estradiol treatment restored the body weight and subcutaneous fat of HFD males to control levels. Hypothalamic POMC mRNA levels in HFD females were increased with respect to control females. This increase was reverted with estradiol treatment during development. DISCUSSION HFD and estradiol treatment have different effects on males and females. Overnutrition affects physiological parameters, such as body weight, visceral, and subcutaneous fat content, in males, while females present alterations in hypothalamic POMC mRNA levels. Hence, the increase in estradiol levels during a period that is critical for the programing of the feeding system can modulate some of the alterations produced by the continuous intake of high-fat content food.
Collapse
Affiliation(s)
- Beatriz Carrillo
- a Departamento de Psicobiología , Universidad Nacional de Educación a Distancia (UNED) , C/ Juan del Rosal n° 10, 28040 Madrid , Spain
| | - Paloma Collado
- a Departamento de Psicobiología , Universidad Nacional de Educación a Distancia (UNED) , C/ Juan del Rosal n° 10, 28040 Madrid , Spain
| | - Francisca Díaz
- b Departamento de Endocrinología , Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Investigación Biomédica en Red (CIBER) de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III , Avda. Menéndez Pelayo, N° 65, 28009 , Madrid
| | - Julie A Chowen
- b Departamento de Endocrinología , Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Investigación Biomédica en Red (CIBER) de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III , Avda. Menéndez Pelayo, N° 65, 28009 , Madrid
| | - Mª Ángeles Pérez-Izquierdo
- a Departamento de Psicobiología , Universidad Nacional de Educación a Distancia (UNED) , C/ Juan del Rosal n° 10, 28040 Madrid , Spain
| | - Helena Pinos
- a Departamento de Psicobiología , Universidad Nacional de Educación a Distancia (UNED) , C/ Juan del Rosal n° 10, 28040 Madrid , Spain
| |
Collapse
|
20
|
Maternal dipyrone treatment during lactation in mice reduces maternal behavior and increases anxiety‐like behavior in offspring. Int J Dev Neurosci 2017; 58:74-81. [DOI: 10.1016/j.ijdevneu.2017.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 02/15/2017] [Accepted: 02/15/2017] [Indexed: 11/23/2022] Open
|
21
|
Early life overnutrition induced by litter size manipulation decreases social play behavior in adolescent male rats. Int J Dev Neurosci 2016; 53:75-82. [DOI: 10.1016/j.ijdevneu.2016.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/19/2016] [Accepted: 07/23/2016] [Indexed: 11/23/2022] Open
|
22
|
Argente-Arizón P, Ros P, Díaz F, Fuente-Martin E, Castro-González D, Sánchez-Garrido MÁ, Barrios V, Tena-Sempere M, Argente J, Chowen JA. Age and sex dependent effects of early overnutrition on metabolic parameters and the role of neonatal androgens. Biol Sex Differ 2016; 7:26. [PMID: 27195103 PMCID: PMC4870809 DOI: 10.1186/s13293-016-0079-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 05/09/2016] [Indexed: 12/28/2022] Open
Abstract
Background Males and females respond differently to diverse metabolic situations. Being raised in a small litter is reported to cause overnutrition that increases weight gain and predisposes an individual to metabolic disturbances in adulthood; however, existing data are inconsistent. Indeed, significant weight gain and/or metabolic disturbances, such as hyperinsulinemia and hyperleptinemia, are sometimes not encountered. We hypothesized that these inconsistencies could be due to the animal’s sex and/or age at which metabolic parameters are measured. Methods To analyze the effects of neonatal overnutrition, male and female Wistar rats were raised in litters of 4 or 12 pups/dam and killed at postnatal days (PND) 10, 21, 30, 50, 85, or 150. In a second study to determine if neonatal sex steroid levels influence sex differences in metabolic parameters, female rats were treated with testosterone on PND1. Effects on weight, length, fat pads, adipokine production, and serum levels of glucose, metabolic hormones, and cytokines were analyzed in both studies. Results By PND10, both males and females raised in small litters had increased body weight, body length, adiposity, and serum glucose, insulin, leptin, and adiponectin levels. Females had a greater increase in inguinal fat, and males had higher expression of leptin messenger RNA (mRNA) and serum insulin, as well as increased testosterone levels. Most of the litter size effects diminished or disappeared after weaning and reappeared during adulthood in males, with sex differences in body size and adiposity being apparent postpubertally. Treatment of females with testosterone on PND1 tended to masculinize some metabolic parameters in adulthood such as increased body weight and serum leptin levels. Conclusions Our results indicate that (1) both sex and age determine the response to neonatal overnutrition; (2) differences in neonatal sex steroid levels may participate in the development of sex differences in metabolic parameters in adulthood and possibly in the response to neonatal overnutrition; and (3) the comparison of circulating hormone and cytokine levels, even in normal control animals, should take into consideration the early neonatal nutritional environment.
Collapse
Affiliation(s)
- Pilar Argente-Arizón
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Avenida Menéndez Pelayo 65, Madrid, 28009 Spain ; Department of Pediatrics, Universidad Autónoma of Madrid, Madrid, Spain ; Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Purificación Ros
- Department of Pediatrics, Universidad Autónoma of Madrid, Madrid, Spain ; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Francisca Díaz
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Avenida Menéndez Pelayo 65, Madrid, 28009 Spain ; Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Esther Fuente-Martin
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Avenida Menéndez Pelayo 65, Madrid, 28009 Spain ; Department of Pediatrics, Universidad Autónoma of Madrid, Madrid, Spain ; Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - David Castro-González
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Avenida Menéndez Pelayo 65, Madrid, 28009 Spain ; Department of Pediatrics, Universidad Autónoma of Madrid, Madrid, Spain ; Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel Ángel Sánchez-Garrido
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain ; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC/HURS), Córdoba, 14004 Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Avenida Menéndez Pelayo 65, Madrid, 28009 Spain ; Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Tena-Sempere
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain ; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC/HURS), Córdoba, 14004 Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Avenida Menéndez Pelayo 65, Madrid, 28009 Spain ; Department of Pediatrics, Universidad Autónoma of Madrid, Madrid, Spain ; Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Avenida Menéndez Pelayo 65, Madrid, 28009 Spain ; Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
23
|
Teixeira VP, Cervilha DAB, Cabral LDM, Oliveira LM, Incerpi EK, Novaes RD, Ionta M, Soncini R. Postnatal overnutrition in mice leads to impaired pulmonary mechanics in response to salbutamol. J Physiol Sci 2016; 66:221-8. [PMID: 26497334 PMCID: PMC10717942 DOI: 10.1007/s12576-015-0417-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 10/03/2015] [Indexed: 01/10/2023]
Abstract
Obesity increases the risk of respiratory disease, which is associated with airway hyperresponsiveness. Although the molecular underpinnings of this phenomenon are not well established, lung remodeling is known as an important factor in this process and could potentially explain compromised lung functions. In the present study, the obesity was induced by postnatal overnutrition in Swiss mice and we investigated the pulmonary mechanics after aerosolization of saline, methacholine, and salbutamol. The lungs were prepared for morphometric analysis. Obese animals showed bronchoconstriction in response to methacholine, as evidenced by airway and tissue resistance, tissue elastance, and hysteresivity. Salbutamol was effective at recovering the response only for airway resistance but not for tissue mechanics. We suggest that this impaired response in obese mice is related to collapsed alveolar, to inflammatory cells, and to elevated deposition collagen fibers in parenchymal tissue.
Collapse
Affiliation(s)
- Vanessa P Teixeira
- Department of Physiology, Institute of Biomedical Science, Federal University of Alfenas, Rua Gabriel Monteiro da Silva, 700, 37130-000, Alfenas, MG, Brazil
| | - Daniela A B Cervilha
- Department of Physiology, Institute of Biomedical Science, Federal University of Alfenas, Rua Gabriel Monteiro da Silva, 700, 37130-000, Alfenas, MG, Brazil
| | - Layla D M Cabral
- Department of Physiology, Institute of Biomedical Science, Federal University of Alfenas, Rua Gabriel Monteiro da Silva, 700, 37130-000, Alfenas, MG, Brazil
| | - Luiz M Oliveira
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, 05508-000, São Paulo, SP, Brazil
| | - Erika K Incerpi
- Department of Physiology, Institute of Biomedical Science, Federal University of Alfenas, Rua Gabriel Monteiro da Silva, 700, 37130-000, Alfenas, MG, Brazil
| | - Rômulo D Novaes
- Integrative Animal Biology Laboratory, Institute of Biomedical Science, Federal University of Alfenas, 37130-000, Alfenas, MG, Brazil
| | - Marisa Ionta
- Integrative Animal Biology Laboratory, Institute of Biomedical Science, Federal University of Alfenas, 37130-000, Alfenas, MG, Brazil
| | - Roseli Soncini
- Department of Physiology, Institute of Biomedical Science, Federal University of Alfenas, Rua Gabriel Monteiro da Silva, 700, 37130-000, Alfenas, MG, Brazil.
| |
Collapse
|
24
|
Reid DT, Eller LK, Nettleton JE, Reimer RA. Postnatal prebiotic fibre intake mitigates some detrimental metabolic outcomes of early overnutrition in rats. Eur J Nutr 2015; 55:2399-2409. [PMID: 26407845 DOI: 10.1007/s00394-015-1047-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 09/14/2015] [Indexed: 11/29/2022]
Abstract
PURPOSE Overnutrition during early development has been linked to metabolic disease and obesity in adulthood. Interventions to ameliorate this metabolic malprogramming are needed. Our objective was to determine whether prebiotic fibre would reduce weight gain and improve satiety hormone profiles in rats overnourished during the suckling period. METHODS Male Sprague-Dawley rats reared in small litter (SL 3 pups) or normal litter (NL 12 pups) were randomized at weaning to AIN-93 (control) or a 10 % oligofructose (OFS) diet for 16 weeks. Body composition, an oral glucose tolerance test for glucose and gut hormones, and gut microbiota were assessed. RESULTS At weaning, body weight was higher in SL than in NL rats (P < 0.03). At 19 weeks, body weight was lower with OFS than control (P < 0.04). There was a diet × litter size interaction wherein OFS in SL rats reduced body fat (%) to levels seen in NL rats (P < 0.05). OFS attenuated the glucose response in SL but not in NL rats (P < 0.015). Independent of litter size, OFS decreased total AUC for glucose-dependent insulinotropic polypeptide (P < 0.002) and increased total AUC for peptide YY (P < 0.01) and glucagon-like peptide-1 (P < 0.04) when compared to control. OFS, not litter size, played the predominant role in altering gut microbiota which included increased bifidobacteria and Akkermansia muciniphila with OFS. CONCLUSIONS Postnatal consumption of OFS by rats raised in SL was able to attenuate body fat and glycaemia to levels seen in NL rats. OFS appears to influence satiety hormone and gut microbiota response similarly in overnourished and control rats.
Collapse
Affiliation(s)
- Danielle T Reid
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Lindsay K Eller
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada.,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Jodi E Nettleton
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Raylene A Reimer
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada. .,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
25
|
Lin X, Qi Q, Zheng Y, Huang T, Lathrop M, Zelenika D, Bray GA, Sacks FM, Liang L, Qi L. Neuropeptide Y genotype, central obesity, and abdominal fat distribution: the POUNDS LOST trial. Am J Clin Nutr 2015; 102:514-9. [PMID: 26156739 PMCID: PMC4515864 DOI: 10.3945/ajcn.115.107276] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 06/11/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Neuropeptide Y is a key peptide affecting adiposity and has been related to obesity risk. However, little is known about the role of NPY variations in diet-induced change in adiposity. OBJECTIVE The objective was to examine the effects of NPY variant rs16147 on central obesity and abdominal fat distribution in response to dietary interventions. DESIGN We genotyped a functional NPY variant rs16147 among 723 participants in the Preventing Overweight Using Novel Dietary Strategies trial. Changes in waist circumference (WC), total abdominal adipose tissue, visceral adipose tissue, and subcutaneous adipose tissue (SAT) from baseline to 6 and 24 mo were evaluated with respect to the rs16147 genotypes. Genotype-dietary fat interaction was also examined. RESULTS The rs16147 C allele was associated with a greater reduction in WC at 6 mo (P < 0.001). In addition, the genotypes showed a statistically significant interaction with dietary fat in relation to WC and SAT (P-interaction = 0.01 and 0.04): the association was stronger in individuals with high-fat intake than in those with low-fat intake. At 24 mo, the association remained statistically significant for WC in the high-fat diet group (P = 0.02), although the gene-dietary fat interaction became nonsignificant (P = 0.30). In addition, we found statistically significant genotype-dietary fat interaction on the change in total abdominal adipose tissue, visceral adipose tissue, and SAT at 24 mo (P = 0.01, 0.05, and 0.04): the rs16147 T allele appeared to associate with more adverse change in the abdominal fat deposition in the high-fat diet group than in the low-fat diet group. CONCLUSION Our data indicate that the NPY rs16147 genotypes affect the change in abdominal adiposity in response to dietary interventions, and the effects of the rs16147 single-nucleotide polymorphism on central obesity and abdominal fat distribution were modified by dietary fat.
Collapse
Affiliation(s)
- Xiaochen Lin
- Departments of Epidemiology, Department of Epidemiology, School of Public Health, Brown University, Providence, RI
| | - Qibin Qi
- Nutrition, and Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | | | | | - Mark Lathrop
- Departments of Human and Medical Genetics, McGill University and Québec Innovation Centre, Montréal, Canada; Fondation Jean Dausset-CEPH, Paris, France
| | - Diana Zelenika
- Commissariat à l'Energie Atomique, Institut Génomique, Centre National de Génotypage, Evry, France
| | - George A Bray
- Pennington Biomedical Research Center of the Louisiana State University System, Baton Rouge, LA
| | | | - Liming Liang
- Departments of Epidemiology, Biostatistics, Harvard School of Public Health, Boston, MA;
| | - Lu Qi
- Nutrition, and Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
26
|
Head GA, Lim K, Barzel B, Burke SL, Davern PJ. Central nervous system dysfunction in obesity-induced hypertension. Curr Hypertens Rep 2015; 16:466. [PMID: 25090962 DOI: 10.1007/s11906-014-0466-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The activation of the sympathetic nervous system is a major mechanism underlying both human and experimental models of obesity-related hypertension. While insulin and the adipokine leptin have long been thought to contribute to obesity-related neurogenic mechanisms, the evidence is now very strong that they play a major role, shown particularly in animal studies using selective receptor antagonists. There is not just maintenance of leptin's sympatho-excitatory actions as previously suggested but considerable amplification particularly in renal sympathetic nervous activity. Importantly, these changes are not dependent on short-term elevation or reduction in plasma leptin or insulin, but require some weeks to develop indicating a slow "neural adaptivity" within hypothalamic signalling. These effects can be carried across generations even when offspring are raised on a normal diet. A better understanding of the underlying mechanism should be a high research priority given the prevalence of obesity not just in the current population but also for future generations.
Collapse
Affiliation(s)
- Geoffrey A Head
- Neuropharmacology Laboratory, Baker IDI Heart and Diabetes Institute, P.O. Box 6492, Melbourne, Victoria, 3004, Australia,
| | | | | | | | | |
Collapse
|
27
|
Gugusheff JR, Ong ZY, Muhlhausler BS. The early origins of food preferences: targeting the critical windows of development. FASEB J 2014; 29:365-73. [PMID: 25466884 DOI: 10.1096/fj.14-255976] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The nutritional environment to which an individual is exposed during the perinatal period plays a crucial role in determining his or her future metabolic health outcomes. Studies in rodent models have demonstrated that excess maternal intake of high-fat and/or high-sugar "junk foods" during pregnancy and lactation can alter the development of the central reward pathway, particularly the opioid and dopamine systems, and program an increased preference for junk foods in the offspring. More recently, there have been attempts to define the critical windows of development during which the opioid and dopamine systems within the reward pathway are most susceptible to alteration and to determine whether it is possible to reverse these effects through nutritional interventions applied later in development. This review discusses the progress made to date in these areas, highlights the apparent importance of sex in determining these effects, and considers the potential implications of the findings from rodent models in the human context.
Collapse
Affiliation(s)
- Jessica Rose Gugusheff
- *FOODplus Research Centre, School of Agriculture Food and Wine, The University of Adelaide, Adelaide, South Australia, Australia; Sansom Institute for Health Research, School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia; and Department of Psychology, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zhi Yi Ong
- *FOODplus Research Centre, School of Agriculture Food and Wine, The University of Adelaide, Adelaide, South Australia, Australia; Sansom Institute for Health Research, School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia; and Department of Psychology, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Beverly Sara Muhlhausler
- *FOODplus Research Centre, School of Agriculture Food and Wine, The University of Adelaide, Adelaide, South Australia, Australia; Sansom Institute for Health Research, School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia; and Department of Psychology, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
28
|
Alles MS, Eussen SRBM, van der Beek EM. Nutritional challenges and opportunities during the weaning period and in young childhood. ANNALS OF NUTRITION AND METABOLISM 2014; 64:284-93. [PMID: 25300272 DOI: 10.1159/000365036] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The early years of life are a period of very rapid growth and development. In this critical phase, food preferences are formed which carry over into childhood and beyond and foundations are laid for a healthy adult life. Excess energy, imbalances in macronutrient quality, and nutritional deficiencies may form inappropriate nutritional signals, leading to metabolic disturbances and affecting the obesity risk. For instance, the intake of protein and sugar-sweetened beverages in young children has been associated with an increased risk of overweight and obesity. In reality, scientific reports have shown that the dietary intakes of vegetables, α-linolenic acid, docosahexaenoic acid, iron, vitamin D, and iodine are low and the intakes of protein, saturated fatty acids, and added sugar are high in young children living in Europe. A focus on improving feeding habits and approaches to support more balanced nutritional intakes early in life may have significant public health benefits.
Collapse
Affiliation(s)
- Martine S Alles
- Nutricia Research, Danone Nutricia Early Life Nutrition, Utrecht, The Netherlands
| | | | | |
Collapse
|
29
|
Martínez AC, Hernández M, Novella S, Martínez MP, Pagán RM, Hermenegildo C, García-Sacristán A, Prieto D, Benedito S. Diminished neurogenic femoral artery vasoconstrictor response in a Zucker obese rat model: differential regulation of NOS and COX derivatives. PLoS One 2014; 9:e106372. [PMID: 25216050 PMCID: PMC4162543 DOI: 10.1371/journal.pone.0106372] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 08/03/2014] [Indexed: 01/21/2023] Open
Abstract
Objective Peripheral arterial disease is one of the macrovascular complications of type 2 diabetes mellitus. This study addresses femoral artery regulation in a prediabetic model of obese Zucker rats (OZR) by examining cross-talk between endothelial and neural factors. Methods and Results Arterial preparations from lean (LZR) and OZR were subjected to electrical field stimulation (EFS) on basal tone. Nitric oxide synthase (NOS) and cyclooxygenase (COX) isoform expression patterns were determined by immunohistochemical labelling and Western blotting. Results indicate significantly reduced noradrenergic contractions in preparations from OZR compared with those of LZR. Functional inhibition of endothelial NOS (eNOS) indicated a predominant role of this isoform in LZR and its modified activity in OZR. Neural (nNOS) and inducible NOS (iNOS) were activated and their expression was higher in femoral arteries from OZR. Neurotransmission modulated by large-conductance Ca2+-activated (BKCa) or voltage-dependent (KV) K+ channels did not seem compromised in the obese animals. Endothelial COX-1 and COX-2 were expressed in LZR and an additional adventitial location of COX-2 was also observed in OZR, explaining the higher COX-2 protein levels detected in this group. Prostanoids derived from both isoforms helped maintain vasoconstriction in LZR while in OZR only COX-2 was active. Superoxide anion inhibition reduced contractions in endothelium-intact arteries from OZR. Conclusions Endothelial dysfunction led to reduced neurogenic vasoconstriction in femoral arteries from OZR. In a setting of obesity, NO-dependent nNOS and iNOS dilation activity could be an alternative mechanism to offset COX-2- and reactive oxygen species-mediated vasoconstriction, along with impaired endothelial NO relaxation.
Collapse
Affiliation(s)
- Ana Cristina Martínez
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Medardo Hernández
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Susana Novella
- Departamento de Fisiología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - María Pilar Martínez
- Departamento de Anatomía y Anatomía Patológica Comparadas, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Rosa María Pagán
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Carlos Hermenegildo
- Departamento de Fisiología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Albino García-Sacristán
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Dolores Prieto
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Sara Benedito
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
- * E-mail:
| |
Collapse
|
30
|
Boersma GJ, Bale TL, Casanello P, Lara HE, Lucion AB, Suchecki D, Tamashiro KL. Long-term impact of early life events on physiology and behaviour. J Neuroendocrinol 2014; 26:587-602. [PMID: 24690036 DOI: 10.1111/jne.12153] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 03/14/2014] [Accepted: 03/25/2014] [Indexed: 01/12/2023]
Abstract
This review discusses the effects of stress and nutrition throughout development and summarises studies investigating how exposure to stress or alterations in nutrition during the pre-conception, prenatal and early postnatal periods can affect the long-term health of an individual. In general, the data presented here suggest that that anything signalling potential adverse conditions later in life, such as high levels of stress or low levels of food availability, will lead to alterations in the offspring, possibly of an epigenetic nature, preparing the offspring for these conditions later in life. However, when similar environmental conditions are not met in adulthood, these alterations may have maladaptive consequences, resulting in obesity and heightened stress sensitivity. The data also suggest that the mechanism underlying these adult phenotypes might be dependent on the type and the timing of exposure.
Collapse
Affiliation(s)
- G J Boersma
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Hou M, Chu Z, Liu T, Lv H, Sun L, Wang B, Huang J, Yan W. A high-fat maternal diet decreases adiponectin receptor-1 expression in offspring. J Matern Fetal Neonatal Med 2014; 28:216-21. [PMID: 24724805 DOI: 10.3109/14767058.2014.914489] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In early life, over-nutrition may increase the risk of insulin resistance in the adult stage. Adiponectin and its receptor may play a key role in this process. This study aimed to identify the effect of a high-fat (HF) maternal diet on metabolic parameters and muscle adiponectin signaling in young adult offspring. We found that offspring born to dams fed HF chow (HF; 31% of calories from fat) had elevated body and adipose tissue weight and higher serum glucose levels after glucose challenge at three weeks (W3) and eight weeks (W8) of age. Offspring exposed to a HF diet also had higher serum adiponectin levels at W3 compared to controls. However, adiponectin levels were significantly decreased compared to controls by W8. Adiponectin receptor 1 mRNA expression in skeletal muscle was decreased in the HF group at W3 and W8, and there was no difference between the two groups in adiponectin receptor 2 expression. Furthermore, glucose transporter 4 mRNA and protein expression was decreased in the skeletal muscle of the HF group at W3 and W8. Our results suggest that a HF maternal diet decreases adiponectin receptor 1 expression in the offspring, which could contribute to reduced sensitivity to adiponectin and to adverse nutritional programing outcomes.
Collapse
Affiliation(s)
- Miao Hou
- Department of Cardiology, Soochow University Affiliated Children's Hospital , Suzhou , China and
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Postnatal early overnutrition causes long-term renal decline in aging male rats. Pediatr Res 2014; 75:259-65. [PMID: 24232634 DOI: 10.1038/pr.2013.223] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 07/04/2013] [Indexed: 01/05/2023]
Abstract
BACKGROUND We evaluated the influence of postnatal early overnutrition on renal pathophysiological changes in aging rats. METHODS Three or 10 male pups per mother were assigned to either the small litter (SL) or normal litter (control) groups, respectively, during the first 21 d of life. The effects of early postnatal overnutrition were determined at 12 mo. RESULTS SL rats weighed more than controls between 4 d and 6 mo of age (P < 0.05). However, between 6 and 12 mo, body weights in both groups were not different. In the SL group, at 12 mo, systolic blood pressure was higher and creatinine clearance was lower than the same in controls (P < 0.05). Numbers of CD68 (ED1)-positive macrophages and apoptotic cells in renal cortex were higher in SL rats (P < 0.05). Furthermore, index scores for glomerulosclerosis and tubulointerstitial fibrosis were higher in the SL group (P < 0.05). Significantly less glomeruli per section area were found in aging SL rats (P < 0.05). Immunoblotting and immunohistochemistry showed decreased intrarenal renin expression in SL rats (P < 0.05). CONCLUSION Early postnatal overnutrition can potentiate structural and functional abnormalities in the aging kidney and can lead to systolic hypertension with reduced intrarenal renin activity.
Collapse
|
33
|
Kjaergaard M, Nilsson C, Rosendal A, Nielsen MO, Raun K. Maternal chocolate and sucrose soft drink intake induces hepatic steatosis in rat offspring associated with altered lipid gene expression profile. Acta Physiol (Oxf) 2014; 210:142-53. [PMID: 23782871 DOI: 10.1111/apha.12138] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 03/01/2013] [Accepted: 06/13/2013] [Indexed: 12/11/2022]
Abstract
AIM According to the World Diabetes Foundation, there is an urgent need to investigate the impact of maternal health and nutrition during pregnancy to understand the background for the accelerating incidence of obesity and type 2 diabetes. In this study, we specifically concentrated on the role of overfeeding during different developmental periods. METHODS Sprague-Dawley rats were offered chow or high-fat/high-sucrose diet (chow plus chocolate and soft drink) during gestation and lactation. At birth, offspring were randomly cross-fostered within each dietary group into small and normal litter sizes until weaning, giving four dietary groups. RESULTS At postnatal day 1, offspring from high-fat/high-sucrose-fed dams were heavier and had increased hepatic triglycerides (TG), hepatic glycogen, blood glucose and plasma insulin compared with offspring from chow-fed dams. Hepatic genes involved in lipid oxidation, VLDL transport and insulin receptor were down-regulated, whereas FGF21 expression was up-regulated. Independent of postnatal litter size, offspring from high-fat/high-sucrose-fed dams aged 21 days had still increased hepatic TG and up-regulated FGF21 expression, while plasma insulin started to decrease. Litter size reduction in offspring from high-fat/high-sucrose-fed dams further increased body weight and adiposity, and up-regulated genes involved in hepatic mitochondrial lipid oxidation and VLDL transport compared with all other groups. Litter size reduction did not have any impact on body weight gain and adiposity in offspring born to chow-fed dams. CONCLUSION Our results suggest that supplementation of chocolate and soft drink during gestation and lactation contributes to early onset of hepatic steatosis associated with changes in hepatic gene expression and lipid handling.
Collapse
Affiliation(s)
- M. Kjaergaard
- Department of Type 2 Diabetes Pharmacology; Novo Nordisk A/S; Maaloev Denmark
- Department of Veterinary Clinical and Animal Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Frederiksberg Denmark
| | - C. Nilsson
- Uppsala University Innovation, Uppsala Science Park; Uppsala Sweden
| | - A. Rosendal
- Department of Assay Technology; Novo Nordisk A/S; Maaloev Denmark
| | - M. O. Nielsen
- Department of Veterinary Clinical and Animal Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Frederiksberg Denmark
| | - K. Raun
- Department of Type 2 Diabetes Pharmacology; Novo Nordisk A/S; Maaloev Denmark
| |
Collapse
|
34
|
Ong ZY, Muhlhausler BS. Consuming a low-fat diet from weaning to adulthood reverses the programming of food preferences in male, but not in female, offspring of 'junk food'-fed rat dams. Acta Physiol (Oxf) 2014; 210:127-41. [PMID: 23746329 DOI: 10.1111/apha.12132] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 04/28/2013] [Accepted: 06/04/2013] [Indexed: 12/16/2022]
Abstract
AIM This study aimed to determine whether the negative effects of maternal 'junk food' feeding on food preferences and gene expression in the mesolimbic reward system could be reversed by weaning the offspring onto a low-fat diet. METHODS Offspring of control (n = 11) and junk food-fed (JF, n = 12) dams were weaned onto a standard rodent chow until 6 weeks (juvenile) or 3 months (adult). They were then given free access to both chow and junk food for 3 weeks and food preferences determined. mRNA expression of key components of the mesolimbic reward system was determined by qRT-PCR at 6 weeks, 3 and 6 months of age. RESULTS In the juvenile group, both male and female JF offspring consumed more energy and carbohydrate during the junk food exposure at 6 weeks of age and had a higher body fat mass at 3 months (P < 0.05). Female juvenile JF offspring had higher tyrosine hydroxylase, dopamine receptors and dopamine active transporter expression in the ventral tegmental area (P < 0.05). In the adult group, there was no difference between control and JF offspring in energy and macronutrient intakes during exposure to junk food; however, female JF offspring had a higher body fat mass at 6 months (P < 0.05). CONCLUSION These results suggest that the effects of perinatal junk food exposure on food preferences and fat mass can be reversed by consuming a low-fat diet from weaning to adulthood in males. Females, however, retain a higher propensity for diet-induced obesity even after consuming a low-fat diet for an extended period after weaning.
Collapse
Affiliation(s)
- Z. Y. Ong
- Sansom Institute for Health Research; School of Pharmacy and Medical Science; University of South Australia; Adelaide Australia
- FOODplus Research Centre; School of Agriculture Food and Wine; The University of Adelaide; Adelaide Australia
| | - B. S. Muhlhausler
- Sansom Institute for Health Research; School of Pharmacy and Medical Science; University of South Australia; Adelaide Australia
- FOODplus Research Centre; School of Agriculture Food and Wine; The University of Adelaide; Adelaide Australia
| |
Collapse
|
35
|
Long-term effect of altered nutrition induced by litter size manipulation and cross-fostering in suckling male rats on development of obesity risk and health complications. Eur J Nutr 2013; 53:1273-80. [DOI: 10.1007/s00394-013-0630-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 11/20/2013] [Indexed: 11/26/2022]
|
36
|
Liu Z, Lim CY, Su MYF, Soh SLY, Shui G, Wenk MR, Grove KL, Radda GK, Han W, Xiao X. Neonatal overnutrition in mice exacerbates high-fat diet-induced metabolic perturbations. J Endocrinol 2013; 219:131-43. [PMID: 23959078 DOI: 10.1530/joe-13-0111] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neonatal overnutrition results in accelerated development of high-fat diet (HFD)-induced metabolic defects in adulthood. To understand whether the increased susceptibility was associated with aggravated inflammation and dysregulated lipid metabolism, we studied metabolic changes and insulin signaling in a chronic postnatal overnutrition (CPO) mouse model. Male Swiss Webster pups were raised with either three pups per litter to induce CPO or ten pups per litter as control (CTR) and weaned to either low-fat diet (LFD) or HFD. All animals were killed on the postnatal day 150 (P150) except for a subset of mice killed on P15 for the measurement of stomach weight and milk composition. CPO mice exhibited accelerated body weight gain and increased body fat mass prior to weaning and the difference persisted into adulthood under conditions of both LFD and HFD. As adults, insulin signaling was more severely impaired in epididymal white adipose tissue (WAT) from HFD-fed CPO (CPO-HFD) mice. In addition, HFD-induced upregulation of pro-inflammatory cytokines was exaggerated in CPO-HFD mice. Consistent with greater inflammation, CPO-HFD mice showed more severe macrophage infiltration than HFD-fed CTR (CTR-HFD) mice. Furthermore, when compared with CTR-HFD mice, CPO-HFD mice exhibited reduced levels of several lipogenic enzymes in WAT and excess intramyocellular lipid accumulation. These data indicate that neonatal overnutrition accelerates the development of insulin resistance and exacerbates HFD-induced metabolic defects, possibly by worsening HFD-induced inflammatory response and impaired lipid metabolism.
Collapse
Affiliation(s)
- Zhiguo Liu
- Laboratory of Lipid and Glucose Metabolism, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Yuzhong District, Chongqing 400016, People's Republic of China Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, Agency for Science Technology and Research (A*STAR), #02-02 Helios, 11 Biopolis Way, Singapore 138667, Singapore Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, USA Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119275, Singapore
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Habbout A, Li N, Rochette L, Vergely C. Postnatal overfeeding in rodents by litter size reduction induces major short- and long-term pathophysiological consequences. J Nutr 2013; 143:553-62. [PMID: 23446961 DOI: 10.3945/jn.112.172825] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Numerous studies have demonstrated that the early postnatal environment can influence body weight and energy homeostasis into adulthood. Rodents raised in small litters have been shown to be a useful experimental model to study the short- and long-term consequences of early overnutrition, which can lead to modifications not only in body weight but also of several metabolic features. Postnatal overfeeding (PNOF) induces early malprogramming of the hypothalamic system, inducing acquired persisting central leptin and insulin resistance and an increase in orexigenic signals. Visceral white adipose tissue, lipogenic activity, and inflammatory status are increased in PNOF rodents, while brown adipose tissue shows reduced thermogenic activity. Pancreatic and hepatic glucose responsiveness is persistently reduced in PNOF rodents, which also frequently present disturbances in plasma lipids. PNOF rodents present increased circulating concentrations of leptin, elevated corticosterone secretion, and significant changes in glucocorticoid sensitivity. PNOF also influences nephrogenesis and renal maturation. Increased oxidative stress is also described in circulating blood and in some tissues, such as the heart or liver. At the cardiovascular level, a moderate increase in arterial blood pressure is sometimes observed and rapid cardiac hypertrophy is observed at weaning; however, during maturation, impaired contractility and fibrosis are observed. Myocardial genome expression is rapidly modified in overfed mice. Moreover, hearts of PNOF rodents are more sensitive to ischemia-reperfusion injury. Together, these results suggest that the nutritional state in the immediate postnatal period should be taken into account, because it may have an impact on cardiometabolic risk in adulthood.
Collapse
Affiliation(s)
- Ahmed Habbout
- Inserm UMR866, LPPCM, Faculties of Medicine and Pharmacy, University of Burgundy, Dijon, France
| | | | | | | |
Collapse
|
38
|
Viana L, Lima C, Oliveira M, Borges R, Cardoso T, Almeida I, Diniz D, Bento-Torres J, Pereira A, Batista-de-Oliveira M, Lopes A, Silva R, Abadie-Guedes R, Amâncio dos Santos A, Lima D, Vasconcelos P, Cunningham C, Guedes R, Picanço-Diniz C. Litter size, age-related memory impairments, and microglial changes in rat dentate gyrus: Stereological analysis and three dimensional morphometry. Neuroscience 2013; 238:280-96. [DOI: 10.1016/j.neuroscience.2013.02.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 02/12/2013] [Accepted: 02/12/2013] [Indexed: 10/27/2022]
|
39
|
Habbout A, Guenancia C, Lorin J, Rigal E, Fassot C, Rochette L, Vergely C. Postnatal overfeeding causes early shifts in gene expression in the heart and long-term alterations in cardiometabolic and oxidative parameters. PLoS One 2013; 8:e56981. [PMID: 23468899 PMCID: PMC3582632 DOI: 10.1371/journal.pone.0056981] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 01/16/2013] [Indexed: 01/24/2023] Open
Abstract
Background Postnatal overfeeding (OF) in rodents induces a permanent moderate increase in body weight in adulthood. However, the repercussions of postnatal OF on cardiac gene expression, cardiac metabolism and nitro-oxidative stress are less well known. Methodology/Principal Findings Immediately after birth, litters of C57BL/6 mice were either maintained at 10 (normal-fed group, NF), or reduced to 3 in order to induce OF. At weaning, mice of both groups received a standard diet. The cardiac gene expression profile was determined at weaning and cardiac metabolism and oxidative stress were assessed at 7 months. The cardiac expression of several genes, including members of the extracellular matrix and apelin pathway, was modified in juvenile OF mice. In adult mice, OF led to an increase in body weight (+30%) and to significant increases in plasma cholesterol, insulin and leptin levels. Myocardial oxidative stress, SOD and catalase activity and mRNA expression were increased in OF mice. In vivo, diastolic and systolic blood pressures were significantly higher and LV shortening and ejection fraction were decreased in OF mice. Ex vivo, after 30 min of ischemia, hearts isolated from OF mice showed lower functional recovery and larger infarct size (31% vs. 54%, p<0.05). Increases in collagen deposition and expression/activity of matrix-metalloproteinase-2 were observed in adult OF mouse hearts. Moreover, an increase in the expression of SOCS-3 and a decrease in STAT-3 phosphorylation were observed in ventricular tissues from OF mice. Conclusions/Significance Our study emphasizes that over-nutrition during the immediate postnatal period in mice leads to early changes in cardiac gene expression, which may permanently modify the heart’s structural organization and metabolism and could contribute to a greater susceptibility to myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Ahmed Habbout
- Inserm UMR866, Laboratoire de Physiopathologie et Pharmacologie Cardio-Métaboliques (LPPCM), Faculties of Medicine and Pharmacy, University of Burgundy, Dijon, France
| | | | | | | | | | | | | |
Collapse
|
40
|
Granado M, Fernández N, Monge L, Figueras JC, Carreño-Tarragona G, Amor S, García-Villalón AL. Effects of coronary ischemia-reperfusion in a rat model of early overnutrition. Role of angiotensin receptors. PLoS One 2013; 8:e54984. [PMID: 23383303 PMCID: PMC3562319 DOI: 10.1371/journal.pone.0054984] [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] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 12/18/2012] [Indexed: 11/19/2022] Open
Abstract
Background Obesity during childhood has dramatically increased worldwide in the last decades. Environmental factors acting early in life, including nutrition, play an important role in the pathogenesis of obesity and cardiovascular diseases in adulthood. Aims To analyze the effects of early overfeeding on the heart and coronary circulation, the effect of ischemia-reperfusion (I/R) and the role of the renin-angiotensin system (RAS) were studied in isolated hearts from control and overfed rats during lactation. Methods and Results On the day of birth litters were adjusted to twelve pups per mother (control) or to three pups per mother (overfed). At weaning (21 days) the rats were killed and the heart perfused in a Langendorff system and subjected to 30 min of ischemia followed by 15 min of reperfusion. The contractility (left developed intraventricular pressure) was lower in the hearts from overfed rats, and was reduced by I/R in hearts from control but not from overfed rats. I/R also reduced the coronary vasoconstriction to angiotensin II more in hearts from control than from overfed rats, and the vasodilatation to bradykinin similarly in both experimental groups. The expression of both angiotensin AGTRa and AGTR2 receptors was increased in the myocardium of overfed rats, and I/R increased the expression of both receptors in control rats but reduced it in overfed rats. The expression of apoptotic and antiapoptotic markers was increased in hearts of overfed rats compared with control, and further increased by I/R. Conclusions These results suggest that both overfeeding and I/R impair cardiac and coronary function due, at least in part, to activation of the angiotensin pathway. However, overfeeding may reduce the impairment of ventricular contractility by I/R.
Collapse
Affiliation(s)
- Miriam Granado
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- CIBER Fisiopatología de Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Nuria Fernández
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Luis Monge
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan Carlos Figueras
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Sara Amor
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | | |
Collapse
|
41
|
Coutinho GVP, Coutinho FR, Faiad JZ, Taki MS, de Lima Reis SR, Ignácio-Souza LM, Paiva AA, Latorraca MQ, Gomes-da-Silva MHG, Martins MSF. Intrauterine protein restriction combined with early postnatal overfeeding was not associated with adult-onset obesity but produced glucose intolerance by pancreatic dysfunction. Nutr Metab (Lond) 2013; 10:5. [PMID: 23305533 PMCID: PMC3574039 DOI: 10.1186/1743-7075-10-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 12/19/2012] [Indexed: 12/19/2022] Open
Abstract
We investigated if whether intrauterine protein restriction in combination with overfeeding during lactation would cause adult-onset obesity and metabolic disorders. After birth, litters from dams fed with control (17% protein) and low protein (6% protein) diets were adjusted to a size of four (CO and LO groups, respectively) or eight (CC and LC groups, respectively) pups. All of the offspring were fed a diet containing 12% protein from the time of weaning until they were 90 d old. Compared to the CC and LC groups, the CO and LO groups had higher relative and absolute food intakes, oxygen consumption and carbon dioxide production; lower brown adipose tissue weight and lipid content and greater weight gain and absolute and relative white adipose tissue weight and absolute lipid content. Compared with the CO and CC rats, the LC and LO rats exhibited higher relative food intake, brown adipose tissue weight and lipid content, reduced oxygen consumption, carbon dioxide production and spontaneous activity, increased relative retroperitoneal adipose tissue weight and unaltered absolute white adipose tissue weight and lipid content. The fasting serum glucose was similar among the groups. The area under the glucose curve was higher in the LO and CO rats than in the LC and CC rats. The basal insulinemia and homeostasis model assessment of insulin resistance (HOMA-IR) were lower in the LO group than in the other groups. The total area under the insulin curve for the LO rats was similar to the CC rats, and both were lower than the CO and LC rats. Kitt was higher in the LO, LC and CO groups than in the CC group. Thus, intrauterine protein restriction followed by overfeeding during lactation did not induce obesity, but produced glucose intolerance by impairing pancreatic function in adulthood.
Collapse
|
42
|
Conceição EPS, Moura EG, Trevenzoli IH, Peixoto-Silva N, Pinheiro CR, Younes-Rapozo V, Oliveira E, Lisboa PC. Neonatal overfeeding causes higher adrenal catecholamine content and basal secretion and liver dysfunction in adult rats. Eur J Nutr 2012; 52:1393-404. [DOI: 10.1007/s00394-012-0448-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 09/13/2012] [Indexed: 12/12/2022]
|
43
|
Zhang X, Qi Q, Liang J, Hu FB, Sacks FM, Qi L. Neuropeptide Y promoter polymorphism modifies effects of a weight-loss diet on 2-year changes of blood pressure: the preventing overweight using novel dietary strategies trial. Hypertension 2012; 60:1169-75. [PMID: 22966009 DOI: 10.1161/hypertensionaha.112.197855] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Neuropeptide Y (NPY) is implicated in the regulation of blood pressure (BP), and NPY pathways in the hypothalamus are sensitive to dietary fat. We evaluated the potential effect of a functional variant rs16147 located in the NPY gene promoter region on the association between 2-year diet intervention and change in multiple BP measures in the randomized Preventing Overweight Using Novel Dietary Strategies Trial. The NPY rs16147 was genotyped in 723 obese adults who were randomly assigned to 1 of 4 diets differing in the target percentages of energy derived from fat, protein, and carbohydrate. The changes of 4 BP phenotypes, including systolic BP, diastolic BP, pulse pressure, and mean arterial pressure, during 2-year diet intervention were analyzed. In the total participants and participants with hypertension, we observed significant and consistent interactions between rs16147 genotype and dietary fat intake on changes in multiple BP phenotypes at 2 years (all P for interactions <0.05). The risk allele (C allele) was associated with a greater reduction of BP phenotypes in response to low-fat diet, whereas an opposite genetic effect was observed in response to high-fat diet. In addition, the C allele was related to greater changes in 4 BP phenotypes in hypertensive compared with nonhypertensive participants. Our data suggest that NPY rs16147 may modulate the association between dietary fat intake and changes in BP phenotypes, and the C allele exerts a long-term beneficial effect on lowering BP in response to low-fat diet in obese and hypertensive subjects.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Department of Nutrition, Harvard School of Public Health, 665 Huntington Ave, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
44
|
Conceição EPS, Franco JG, Oliveira E, Resende AC, Amaral TAS, Peixoto-Silva N, Passos MCF, Moura EG, Lisboa PC. Oxidative stress programming in a rat model of postnatal early overnutrition--role of insulin resistance. J Nutr Biochem 2012; 24:81-7. [PMID: 22819562 DOI: 10.1016/j.jnutbio.2012.02.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 12/05/2011] [Accepted: 02/17/2012] [Indexed: 12/21/2022]
Abstract
Postnatal early overfeeding (EO) is related to later development of overweight and other metabolic disorders. As oxidative stress is implicated in most human diseases, as obesity and diabetes, we decided to study some parameters related to oxidative stress and insulin signaling in liver from EO animals in adult life. To induce EO, litter size was reduced to three pups per litter (SL: small litter) and groups with normal litter size (NL:10 pups per litter) were used as control. After weaning, rats had free access to standard diet and water. Body weight and food intake were monitored daily and offspring were killed at 180 days-old. Significant differences had P<.05 or less. As expected, SL rats had hyperphagia, higher body weight and higher visceral fat mass at weaning and adulthood. In liver, postnatal EO programmed for lower catalase (-42%), superoxide dismutase (-45%) and glutathione peroxidase (-65%) activities. The evaluation of liver injury in adult SL group showed lower nitrite content (-10%), higher liver and plasma malondialdehyde content (+25% and 1.1-fold increase, respectively). No changes of total protein bound carbonyl or Cu/Zn superoxide dismutase protein expression in liver were detected between the groups. Regarding insulin signaling pathway in liver, SL offspring showed lower IRβ (-66%), IRS1 (-50%), phospho-IRS1 (-73%), PI3-K (-30%) and Akt1 (-58%). Indeed, morphological analysis showed that SL rats presented focal areas of inflammatory cell infiltrate and lipid drops in their cytoplasm characterizing a microsteatosis. Thus, we evidenced that postnatal EO can program the oxidative stress in liver, maybe contributing for impairment of the insulin signaling.
Collapse
Affiliation(s)
- Ellen P S Conceição
- Department of Physiological Sciences, Roberto Alcântara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ 20551-030, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Fox EA, Biddinger JE. Early postnatal overnutrition: potential roles of gastrointestinal vagal afferents and brain-derived neurotrophic factor. Physiol Behav 2012; 106:400-12. [PMID: 22712064 PMCID: PMC3517218 DOI: 10.1016/j.physbeh.2012.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Abnormal perinatal nutrition (APN) results in a predisposition to develop obesity and the metabolic syndrome and thus may contribute to the prevalence of these disorders. Obesity, including that which develops in organisms exposed to APN, has been associated with increased meal size. Vagal afferents of the gastrointestinal (GI) tract contribute to regulation of meal size by transmitting satiation signals from gut-to-brain. Consequently, APN could increase meal size by altering this signaling, possibly through changes in expression of factors that control vagal afferent development or function. Here two studies that addressed these possibilities are reviewed. First, meal patterns, meal microstructure, and the structure and density of vagal afferents that innervate the intestine were examined in mice that experienced early postnatal overnutrition (EPO). These studies provided little evidence for EPO effects on vagal afferents as it did not alter meal size or vagal afferent density or structure. However, these mice exhibited modest hyperphagia due to a satiety deficit. In parallel, the possibility that brain-derived neurotrophic factor (BDNF) could mediate APN effects on vagal afferent development was investigated. Brain-derived neurotrophic factor was a strong candidate because APN alters BDNF levels in some tissues and BDNF knockout disrupts development of vagal sensory innervation of the GI tract. Surprisingly, smooth muscle-specific BDNF knockout resulted in early-onset obesity and hyperphagia due to increases in meal size and frequency. Microstructure analysis revealed decreased decay of intake rate during a meal in knockouts, suggesting that the loss of vagal negative feedback contributed to their increase in meal size. However, meal-induced c-Fos activation within the dorsal vagal complex suggested this effect could be due to augmentation of vago-vagal reflexes. A model is proposed to explain how high-fat diet consumption produces increased obesity in organisms exposed to APN, and may be required to reveal effects of EPO on vagal function.
Collapse
Affiliation(s)
- Edward A Fox
- Behavioral Neurogenetics Laboratory & Ingestive Behavior Research Center, Department of Psychological Sciences, Purdue University, West Lafayette, IN 47907, USA.
| | | |
Collapse
|
46
|
Maniam J, Morris MJ. The link between stress and feeding behaviour. Neuropharmacology 2012; 63:97-110. [PMID: 22710442 DOI: 10.1016/j.neuropharm.2012.04.017] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 04/11/2012] [Accepted: 04/20/2012] [Indexed: 02/06/2023]
Abstract
Exposure to stress is inevitable, and it may occur, to varying degrees, at different phases throughout the lifespan. The impact of stress experienced in later life has been well documented as many populations in modern society experience increasing socio-economic demands. The effects of stress early in life are less well known, partly as the impact of an early exposure may be difficult to quantify, however emerging evidence shows it can impact later in life. One of the major impacts of stress besides changes in psychosocial behaviour is altered feeding responses. The system that regulates stress responses, the hypothalamo-pituitary-adrenal axis, also regulates feeding responses because the neural circuits that regulate food intake converge on the paraventricular nucleus, which contains corticotrophin releasing hormone (CRH), and urocortin containing neurons. In other words the systems that control food intake and stress responses share the same anatomy and thus each system can influence each other in eliciting a response. Stress is known to alter feeding responses in a bidirectional pattern, with both increases and decreases in intake observed. Stress-induced bidirectional feeding responses underline the complex mechanisms and multiple contributing factors, including the levels of glucocorticoids (dependent on the severity of a stressor), the interaction between glucocorticoids and feeding related neuropeptides such as neuropeptide Y (NPY), alpha-melanocyte stimulating hormone (α-MSH), agouti-related protein (AgRP), melanocortins and their receptors, CRH, urocortin and peripheral signals (leptin, insulin and ghrelin). This review discusses the neuropeptides that regulate feeding behaviour and how their function can be altered through cross-talk with hormones and neuropeptides that also regulate the hypothalamo-pituitary-adrenal axis. In addition, long-term stress induced alterations in feeding behaviour, and changes in gene expression of neuropeptides regulating stress and food intake through epigenetic modifications will be discussed. This article is part of a Special Issue entitled 'SI: Central Control of Food Intake'.
Collapse
Affiliation(s)
- Jayanthi Maniam
- Pharmacology, School of Medical Sciences, University of New South Wales, Sydney NSW 2052, Australia
| | | |
Collapse
|
47
|
Jiang F, Lim HK, Morris MJ, Prior L, Velkoska E, Wu X, Dusting GJ. Systemic upregulation of NADPH oxidase in diet-induced obesity in rats. Redox Rep 2012; 16:223-9. [PMID: 22195989 DOI: 10.1179/174329211x13049558293713] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase is upregulated in a variety of tissues in obesity. It is still unclear as to whether NADPH oxidase upregulation in a specific tissue is part of a systemic response. Here we analyzed the expression pattern of NADPH oxidase in vascular, adipose, and kidney tissues in a rat model of diet-induced obesity. After weaning, rats were fed either a normal or high-fat diet for 12 weeks. The high-fat diet resulted in 20% increased body weight. In the aorta, Nox4 expression was increased by three-fold in obese rats. Upregulations of p22phox and p47phox in adipose, and Nox4, p22phox, and p47phox in kidney were observed in obesity. Marked increases in plasma leptin and insulin were observed, with more modest changes in adiponectin in obese rats. The average systolic blood pressure in the obese group was 11 mmHg higher than that of lean rats (P < 0.005). There was a significant correlation between blood pressure and aortic Nox4 expression (P < 0.01). In cultured vascular smooth muscle cells, adiponectin reduced the expression of Nox4 in a protein kinase A-dependent manner. Our results suggest that upregulation of NADPH oxidase in multiple tissues during obesity appears to be a systemic response. At least in vitro, adiponectin may have a protective antioxidant role by suppressing vascular NADPH oxidase expression. The association between NADPH oxidase Nox4 expression in the vasculature and the elevated blood pressure in obesity requires further investigation.
Collapse
Affiliation(s)
- Fan Jiang
- O'Brien Institute and Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
48
|
Alcazar MAA, Boehler E, Rother E, Amann K, Vohlen C, von Hörsten S, Plank C, Dötsch J. Early postnatal hyperalimentation impairs renal function via SOCS-3 mediated renal postreceptor leptin resistance. Endocrinology 2012; 153:1397-410. [PMID: 22253420 DOI: 10.1210/en.2011-1670] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Early postnatal hyperalimentation has long-term implications for obesity and developing renal disease. Suppressor of cytokine signaling (SOCS) 3 inhibits phosphorylation of signal transducer and activator of transcription (STAT) 3 and ERK1/2 and thereby plays a pivotal role in mediating leptin resistance. In addition, SOCS-3 is induced by both leptin and inflammatory cytokines. However, little is known about the intrinsic-renal leptin synthesis and function. Therefore, this study aimed to elucidate the implications of early postnatal hyperalimentation on renal function and on the intrinsic-renal leptin signaling. Early postnatal hyperalimentation in Wistar rats during lactation was induced by litter size reduction at birth (LSR) either to LSR10 or LSR6, compared with home cage control male rats. Assessment of renal function at postnatal day 70 revealed decreased glomerular filtration rate and proteinuria after LSR6. In line with this impairment of renal function, renal inflammation and expression as well as deposition of extracellular matrix molecules, such as collagen I, were increased. Furthermore, renal expression of leptin and IL-6 was up-regulated subsequent to LSR6. Interestingly, the phosphorylation of Stat3 and ERK1/2 in the kidney, however, was decreased after LSR6, indicating postreceptor leptin resistance. In accordance, neuropeptide Y (NPY) gene expression was down-regulated; moreover, SOCS-3 protein expression, a mediator of postreceptor leptin resistance, was strongly elevated and colocalized with NPY. Thus, our findings not only demonstrate impaired renal function and profibrotic processes but also provide compelling evidence of a SOCS-3-mediated intrinsic renal leptin resistance and concomitant up-regulated NPY expression as an underlying mechanism.
Collapse
|
49
|
Figueiredo MS, da Fonseca Passos MC, Trevenzoli IH, Troina AA, Carlos AS, Alves Nascimento-Saba CC, Fraga MC, Manhães AC, de Oliveira E, Lisboa PC, de Moura EG. Adipocyte morphology and leptin signaling in rat offspring from mothers supplemented with flaxseed during lactation. Nutrition 2012; 28:307-15. [DOI: 10.1016/j.nut.2011.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 07/03/2011] [Accepted: 07/04/2011] [Indexed: 10/16/2022]
|
50
|
South T, Westbrook F, Morris MJ. Neurological and stress related effects of shifting obese rats from a palatable diet to chow and lean rats from chow to a palatable diet. Physiol Behav 2012; 105:1052-7. [DOI: 10.1016/j.physbeh.2011.11.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 11/22/2011] [Accepted: 11/23/2011] [Indexed: 11/29/2022]
|