Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
2.
Int J Obes (Lond) ; 44(9): 1946-1957, 2020 09.
Article En | MEDLINE | ID: mdl-32719434

BACKGROUND: Though it is well established that neonatal nutrition plays a major role in lifelong offspring health, the mechanisms underpinning this have not been well defined. Early postnatal accelerated growth resulting from maternal nutritional status is associated with increased appetite and body weight. Likewise, slow growth correlates with decreased appetite and body weight. Food consumption and food-seeking behaviour are directly modulated by central serotonergic (5-hydroxytryptamine, 5-HT) pathways. This study examined the effect of a rat maternal postnatal low protein (PLP) diet on 5-HT receptor mediated food intake in offspring. METHODS: Microarray analyses, in situ hybridization or laser capture microdissection of the ARC followed by RT-PCR were used to identify genes up- or down-regulated in the arcuate nucleus of the hypothalamus (ARC) of 3-month-old male PLP rats. Third ventricle cannulation was used to identify altered sensitivity to serotonin receptor agonists and antagonists with respect to food intake. RESULTS: Male PLP offspring consumed less food and had lower growth rates up to 3 months of age compared with Control offspring from dams fed a normal diet. In total, 97 genes were upregulated including the 5-HT5A receptor (5-HT5AR) and 149 downregulated genes in PLP rats compared with Controls. The former obesity medication fenfluramine and the 5-HT receptor agonist 5-Carboxamidotryptamine (5-CT) significantly suppressed food intake in both groups, but the PLP offspring were more sensitive to d-fenfluramine and 5-CT compared with Controls. The effect of 5-CT was antagonized by the 5-HT5AR antagonist SB699551. 5-CT also reduced NPY-induced hyperphagia in both Control and PLP rats but was more effective in PLP offspring. CONCLUSIONS: Postnatal low protein programming of growth in rats enhances the central effects of serotonin on appetite by increasing hypothalamic 5-HT5AR expression and sensitivity. These findings provide insight into the possible mechanisms through which a maternal low protein diet during lactation programs reduced growth and appetite in offspring.


Appetite/physiology , Body Weight/physiology , Gene Expression Regulation, Developmental/genetics , Hypothalamus/metabolism , Receptors, Serotonin , Animals , Diet , Female , Male , Obesity/metabolism , Rats , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Transcriptome/genetics
3.
Diabetologia ; 59(6): 1266-75, 2016 06.
Article En | MEDLINE | ID: mdl-26965244

AIMS/HYPOTHESIS: Individuals with a low birthweight have an increased risk of developing type 2 diabetes mellitus in adulthood. This is associated with peripheral insulin resistance. Here, we aimed to determine whether changes in insulin signalling proteins in white adipose tissue (WAT) can be detected prior to the onset of impaired glucose tolerance, determine whether these changes are cell-autonomous and identify the underlying mechanisms involved. METHODS: Fourteen-month-old male rat offspring born to dams fed a standard protein (20%) diet or a low (8%) protein diet throughout gestation and lactation were studied. Fat distribution and adipocyte size were determined. Protein content and mRNA expression of key insulin signalling molecules were analysed in epididymal WAT and in pre-adipocytes that had undergone in vitro differentiation. RESULTS: The offspring of low protein fed dams (LP offspring) had reduced visceral WAT mass, altered fat distribution and a higher percentage of small adipocytes in epididymal WAT. This was associated with reduced levels of IRS1, PI3K p110ß, Akt1 and PKCζ proteins and of phospho-Akt Ser473. Corresponding mRNA transcript levels were unchanged. Similarly, in vitro differentiated adipocytes from LP offspring showed reduced protein levels of IRß, IRS1, PI3K p85α and p110ß subunits, and Akt1. Levels of Akt Ser473 and IRS1 Tyr612 phosphorylation were reduced, while IRS1 Ser307 phosphorylation was increased. CONCLUSIONS/INTERPRETATION: Maternal protein restriction during gestation and lactation changes the distribution and morphology of WAT and reduces the levels of key insulin signalling proteins in the male offspring. This phenotype is retained in in vitro differentiated adipocytes, suggesting that programming occurs via cell-autonomous mechanism(s).


Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue/cytology , Signal Transduction/physiology , Animals , Cell Size , Cells, Cultured , Diet, Protein-Restricted , Female , Insulin/metabolism , Insulin Resistance/physiology , Male , Maternal Nutritional Physiological Phenomena/physiology , MicroRNAs/genetics , Pregnancy , Rats
4.
Dis Model Mech ; 9(4): 401-12, 2016 Apr.
Article En | MEDLINE | ID: mdl-26769798

Although obesity is a global epidemic, the physiological mechanisms involved are not well understood. Recent advances reveal that susceptibility to obesity can be programmed by maternal and neonatal nutrition. Specifically, a maternal low-protein diet during pregnancy causes decreased intrauterine growth, rapid postnatal catch-up growth and an increased risk for diet-induced obesity. Given that the synthesis of the neurotransmitter 5-hydroxytryptamine (5-HT) is nutritionally regulated and 5-HT is a trophic factor, we hypothesised that maternal diet influences fetal 5-HT exposure, which then influences development of the central appetite network and the subsequent efficacy of 5-HT to control energy balance in later life. Consistent with our hypothesis, pregnant rats fed a low-protein diet exhibited elevated serum levels of 5-HT, which was also evident in the placenta and fetal brains at embryonic day 16.5. This increase was associated with reduced levels of 5-HT2CR, the primary 5-HT receptor influencing appetite, in the fetal, neonatal and adult hypothalamus. As expected, a reduction of 5-HT2CR was associated with impaired sensitivity to 5-HT-mediated appetite suppression in adulthood. 5-HT primarily achieves effects on appetite by 5-HT2CR stimulation of pro-opiomelanocortin (POMC) peptides within the arcuate nucleus of the hypothalamus (ARC). We show that 5-HT2ARs are also anatomically positioned to influence the activity of ARC POMC neurons and that mRNA encoding 5-HT2AR is increased in the hypothalamus ofin uterogrowth-restricted offspring that underwent rapid postnatal catch-up growth. Furthermore, these animals at 3 months of age are more sensitive to appetite suppression induced by 5-HT2AR agonists. These findings not only reveal a 5-HT-mediated mechanism underlying the programming of susceptibility to obesity, but also provide a promising means to correct it, by treatment with a 5-HT2AR agonist.


Growth and Development , Hypothalamus/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Animals , Animals, Newborn , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Body Weight/drug effects , Dietary Proteins/pharmacology , Feeding Behavior/drug effects , Female , Fenfluramine/administration & dosage , Fenfluramine/pharmacology , Fetus/drug effects , Fetus/metabolism , Growth and Development/drug effects , Hypothalamus/anatomy & histology , Hypothalamus/drug effects , Hypothalamus/growth & development , Laser Capture Microdissection , Male , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Organ Size/drug effects , Pregnancy , Rats, Wistar , Reproducibility of Results , Serotonin/metabolism , Time Factors , Tryptophan/metabolism
5.
Am J Clin Nutr ; 103(2): 579-88, 2016 Feb.
Article En | MEDLINE | ID: mdl-26718412

BACKGROUND: It is well established that low birth weight and accelerated postnatal growth increase the risk of liver dysfunction in later life. However, molecular mechanisms underlying such developmental programming are not well characterized, and potential intervention strategies are poorly defined. OBJECTIVES: We tested the hypotheses that poor maternal nutrition and accelerated postnatal growth would lead to increased hepatic fibrosis (a pathological marker of liver dysfunction) and that postnatal supplementation with the antioxidant coenzyme Q10 (CoQ10) would prevent this programmed phenotype. DESIGN: A rat model of maternal protein restriction was used to generate low-birth-weight offspring that underwent accelerated postnatal growth (termed "recuperated"). These were compared with control rats. Offspring were weaned onto standard feed pellets with or without dietary CoQ10 (1 mg/kg body weight per day) supplementation. At 12 mo, hepatic fibrosis, indexes of inflammation, oxidative stress, and insulin signaling were measured by histology, Western blot, ELISA, and reverse transcriptase-polymerase chain reaction. RESULTS: Hepatic collagen deposition (diameter of deposit) was greater in recuperated offspring (mean ± SEM: 12 ± 2 µm) than in controls (5 ± 0.5 µm) (P < 0.001). This was associated with greater inflammation (interleukin 6: 38% ± 24% increase; P < 0.05; tumor necrosis factor α: 64% ± 24% increase; P < 0.05), lipid peroxidation (4-hydroxynonenal, measured by ELISA: 0.30 ± 0.02 compared with 0.19 ± 0.05 µg/mL per µg protein; P < 0.05), and hyperinsulinemia (P < 0.05). CoQ10 supplementation increased (P < 0.01) hepatic CoQ10 concentrations and ameliorated liver fibrosis (P < 0.001), inflammation (P < 0.001), some measures of oxidative stress (P < 0.001), and hyperinsulinemia (P < 0.01). CONCLUSIONS: Suboptimal in utero nutrition combined with accelerated postnatal catch-up growth caused more hepatic fibrosis in adulthood, which was associated with higher indexes of oxidative stress and inflammation and hyperinsulinemia. CoQ10 supplementation prevented liver fibrosis accompanied by downregulation of oxidative stress, inflammation, and hyperinsulinemia.


Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Dietary Supplements , Fetal Growth Retardation/diet therapy , Hepatitis/prevention & control , Liver Cirrhosis/prevention & control , Oxidative Stress , Ubiquinone/analogs & derivatives , Animals , Cytokines/antagonists & inhibitors , Cytokines/blood , Cytokines/metabolism , Diet, Protein-Restricted/adverse effects , Female , Fetal Development , Fetal Growth Retardation/etiology , Fetal Growth Retardation/immunology , Fetal Growth Retardation/physiopathology , Hepatitis/etiology , Hepatitis/metabolism , Hepatitis/pathology , Hyperinsulinism/etiology , Hyperinsulinism/prevention & control , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Malnutrition/physiopathology , Maternal Nutritional Physiological Phenomena , Pregnancy , Pregnancy Complications/physiopathology , Rats, Wistar , Specific Pathogen-Free Organisms , Ubiquinone/therapeutic use , Weaning
6.
FASEB J ; 28(12): 5398-405, 2014 Dec.
Article En | MEDLINE | ID: mdl-25172893

Low birth weight and rapid postnatal growth increases risk of cardiovascular-disease (CVD); however, underlying mechanisms are poorly understood. Previously, we demonstrated that rats exposed to a low-protein diet in utero that underwent postnatal catch-up growth (recuperated) have a programmed deficit in cardiac coenzyme Q (CoQ) that was associated with accelerated cardiac aging. It is unknown whether this deficit occurs in all tissues, including those that are clinically accessible. We investigated whether aortic and white blood cell (WBC) CoQ is programmed by suboptimal early nutrition and whether postweaning dietary supplementation with CoQ could prevent programmed accelerated aging. Recuperated male rats had reduced aortic CoQ [22 d (35±8.4%; P<0.05); 12 m (53±8.8%; P<0.05)], accelerated aortic telomere shortening (P<0.01), increased DNA damage (79±13% increase in nei-endonucleaseVIII-like-1), increased oxidative stress (458±67% increase in NAPDH-oxidase-4; P<0.001), and decreased mitochondrial complex II-III activity (P<0.05). Postweaning dietary supplementation with CoQ prevented these detrimental programming effects. Recuperated WBCs also had reduced CoQ (74±5.8%; P<0.05). Notably, WBC CoQ levels correlated with aortic telomere-length (P<0.0001) suggesting its potential as a diagnostic marker of vascular aging. We conclude that early intervention with CoQ in at-risk individuals may be a cost-effective and safe way of reducing the global burden of CVDs.


Cardiovascular Diseases/prevention & control , Dietary Supplements , Ubiquinone/metabolism , Animals , Cardiovascular Diseases/enzymology , Female , Oxidative Stress , Pregnancy , Rats, Wistar , Telomerase/metabolism , Ubiquinone/administration & dosage
7.
Am J Physiol Regul Integr Comp Physiol ; 307(1): R26-34, 2014 Jul 01.
Article En | MEDLINE | ID: mdl-24789994

Changes in the maternal nutritional environment during fetal development can influence offspring's metabolic risk in later life. Animal models have demonstrated that offspring of diet-induced obese dams develop metabolic complications, including nonalcoholic fatty liver disease. In this study we investigated the mechanisms in young offspring that lead to the development of nonalcoholic fatty liver disease (NAFLD). Female offspring of C57BL/6J dams fed either a control or obesogenic diet were studied at 8 wk of age. We investigated the roles of oxidative stress and lipid metabolism in contributing to fatty liver in offspring. There were no differences in body weight or adiposity at 8 wk of age; however, offspring of obese dams were hyperinsulinemic. Oxidative damage markers were significantly increased in their livers, with reduced levels of the antioxidant enzyme glutathione peroxidase-1. Mitochondrial complex I and II activities were elevated, while levels of mitochondrial cytochrome c were significantly reduced and glutamate dehydrogenase was significantly increased, suggesting mitochondrial dysfunction. Offspring of obese dams also had significantly greater hepatic lipid content, associated with increased levels of PPARγ and reduced triglyceride lipase. Liver glycogen and protein content were concomitantly reduced in offspring of obese dams. In conclusion, offspring of diet-induced obese dams have disrupted liver metabolism and develop NAFLD prior to any differences in body weight or body composition. Oxidative stress may play a mechanistic role in the progression of fatty liver in these offspring.


Animal Nutritional Physiological Phenomena , Fatty Liver/etiology , Lipid Metabolism , Liver/metabolism , Obesity/complications , Oxidative Stress , Prenatal Exposure Delayed Effects , Prenatal Nutritional Physiological Phenomena , Adiposity , Age Factors , Animals , Body Weight , Cytochromes c/metabolism , Electron Transport Complex I/metabolism , Electron Transport Complex II/metabolism , Fatty Liver/metabolism , Female , Glutamate Dehydrogenase/metabolism , Glutathione Peroxidase/metabolism , Glycogen/metabolism , Homeostasis , Insulin/metabolism , Lipase/metabolism , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Obesity/metabolism , Obesity/physiopathology , PPAR gamma/metabolism , Phenotype , Pregnancy , Risk Factors , Signal Transduction , Glutathione Peroxidase GPX1
8.
Mol Metab ; 3(3): 325-33, 2014 Jun.
Article En | MEDLINE | ID: mdl-24749062

We determined the effects of maternal diet-induced obesity on offspring adipose tissue insulin signalling and miRNA expression in the aetiology of insulin resistance in later life. Although body composition and glucose tolerance of 8-week-old male offspring of obese dams were not dysregulated, serum insulin was significantly (p<0.05) elevated. Key insulin signalling proteins in adipose tissue were down-regulated, including the insulin receptor, catalytic (p110ß) and regulatory (p85α) subunits of PI3K as well as AKT1 and 2 (all p<0.05). The largest reduction observed was in IRS-1 protein (p<0.001), which was regulated post-transcriptionally. Concurrently, miR-126, which targets IRS-1, was up-regulated (p<0.05). These two features were maintained in isolated primary pre-adipocytes and differentiated adipocytes in-vitro. We have therefore established that maternal diet-induced obesity programs adipose tissue insulin resistance. We hypothesise that maintenance of the phenotype in-vitro strongly suggests that this mechanism is cell autonomous and may drive insulin resistance in later life.

9.
Mol Metab ; 2(4): 480-90, 2013.
Article En | MEDLINE | ID: mdl-24327963

Studies in human and animals have demonstrated that nutritionally induced low birth-weight followed by rapid postnatal growth increases the risk of metabolic syndrome and cardiovascular disease. Although the mechanisms underlying such nutritional programming are not clearly defined, increased oxidative-stress leading to accelerated cellular aging has been proposed to play an important role. Using an established rodent model of low birth-weight and catch-up growth, we show here that post-weaning dietary supplementation with coenzyme Q10, a key component of the electron transport chain and a potent antioxidant rescued many of the detrimental effects of nutritional programming on cardiac aging. This included a reduction in nitrosative and oxidative-stress, telomere shortening, DNA damage, cellular senescence and apoptosis. These findings demonstrate the potential for postnatal antioxidant intervention to reverse deleterious phenotypes of developmental programming and therefore provide insight into a potential translatable therapy to prevent cardiovascular disease in at risk humans.

10.
FASEB J ; 27(9): 3786-96, 2013 Sep.
Article En | MEDLINE | ID: mdl-23729590

Our aim was to determine the effect of exposure to maternal obesity or to maternal weight loss around conception on the programming of hepatic insulin signaling in the offspring. We used an embryo transfer model in sheep to investigate the effects of exposure to either maternal obesity or to weight loss in normal and obese mothers preceding and for 1 wk after conception on the expression of hepatic insulin-signaling and gluconeogenic factors and key miRNAs involved in insulin signaling in the offspring. We found that exposure to maternal obesity resulted in increased hepatic miR-29b (P<0.05), miR-103 (P<0.01), and miR-107 (P<0.05) expression, a decrease in IR (P<0.05), phopsho-Akt (P<0.01), and phospho-FoxO1 (P<0.01) abundance, and a paradoxical decrease in 11ßHSD1 (P<0.05), PEPCK-C (P<0.01), and PEPCK-M (P<0.05) expression in lambs. These changes were ablated by a period of moderate dietary restriction imposed during the periconceptional period. Maternal dietary restriction alone also resulted in decreased abundance of a separate subset of hepatic insulin-signaling molecules, namely, IRS1 (P<0.05), PDK1 (P<0.01), phospho-PDK1 (P<0.05), and aPKCζ (P<0.05) and in decreased PEPCK-C (P<0.01) and G6Pase (P<0.01) expression in the lamb. Our findings highlight the sensitivity of the epigenome to maternal nutrition around conception and the need for dietary interventions that maximize metabolic benefits and minimize metabolic costs for the next generation.


Epigenesis, Genetic/genetics , Insulin/metabolism , Liver/metabolism , MicroRNAs/genetics , Obesity/genetics , Weight Loss/physiology , Animals , Female , Pregnancy , Sheep , Weight Loss/genetics
11.
Med Wieku Rozwoj ; 17(1): 7-12, 2013.
Article En | MEDLINE | ID: mdl-23749690

As the prevalence of obesity increases across the globe, vast efforts are being directed towards understanding the origins of obesity and mechanisms underlying this rapid increase. It is well known that the current environment of an individual can affect body weight, however, growing evidence suggests that the environment in very early life may be particularly important in determining long term obesity risk. This was prompted by a series of epidemiological studies demonstrating a relationship between suboptimal early growth and later risk of obesity. Evidence from human studies as well as animal models have shown that alterations in nutrition and growth in utero and during early postnatal life can have permanent effects on systems mediating regulation of energy balance. Rapid postnatal growth in particular has been associated with increased risk of developing obesity while slower postnatal growth lowers this risk. Alterations in pathways mediating energy homeostasis have been associated with both patterns of early growth. These include changes in structure and function of neuronal pathways in the brain which lead to deregulation of pathways mediating energy balance. In addition to the alterations at the central level, early nutrition can have detrimental long-lasting effects on peripheral physiological systems, for example the storage of fat and utilization of nutrients that make an individual more prone to development of obesity. The fundamental mechanisms underlying these programmed changes are still to be fully defined, although epigenetic mechanisms may play an important role.


Obesity/epidemiology , Obesity/physiopathology , Adipose Tissue/metabolism , Animals , Disease Models, Animal , Energy Metabolism , Epigenesis, Genetic , Homeostasis , Humans , Risk Factors
12.
FASEB J ; 27(1): 379-90, 2013 Jan.
Article En | MEDLINE | ID: mdl-23024373

Low birth weight and accelerated postnatal growth lead to increased risk of cardiovascular disease. We reported previously that rats exposed to a low-protein diet in utero and postnatal catch-up growth (recuperated) develop metabolic dysfunction and have reduced life span. Here we explored the hypothesis that cardiac oxidative and nitrosative stress leading to DNA damage and accelerated cellular aging could contribute to these phenotypes. Recuperated animals had a low birth weight (P<0.001) but caught up in weight to controls during lactation. At weaning, recuperated cardiac tissue had increased (P<0.05) protein nitrotyrosination and DNA single-stranded breaks. This condition was preceded by increased expression of DNA damage repair molecules 8-oxoguanine-DNA-glycosylase-1, nei-endonuclease-VIII-like, X-ray-repair-complementing-defective-repair-1, and Nthl endonuclease III-like-1 on d 3. These differences were maintained on d 22 and became more pronounced in the case of 8-oxoguanine-DNA-glycosylase-1 and nei-endonuclease-VIII-like. This was accompanied by increases in xanthine oxidase (P<0.001) and NADPH oxidase (P<0.05), major sources of reactive oxygen species (ROS). The detrimental effects of increased ROS in recuperated offspring may be exaggerated at 22 d by reductions (P<0.001) in the antioxidant enzymes peroxiredoxin-3 and CuZn-superoxide-dismutase. We conclude that poor fetal nutrition followed by accelerated postnatal growth results in increased cardiac nitrosative and oxidative-stress and DNA damage, which could contribute to age-associated disease risk.


DNA Damage , DNA Repair , Growth , Malnutrition/metabolism , Myocardium/metabolism , Nitrosation , Oxidative Stress , Animals , Base Sequence , Body Weight , DNA Primers , DNA, Mitochondrial/genetics , Female , Gene Expression Profiling , Malnutrition/genetics , Organ Size , Pregnancy , Rats , Rats, Wistar , Telomere
13.
PLoS One ; 8(12): e84594, 2013.
Article En | MEDLINE | ID: mdl-24386400

Exposure to maternal obesity before and/or throughout pregnancy may increase the risk of obesity and insulin resistance in the offspring in childhood and adult life, therefore, resulting in its transmission into subsequent generations. We have previously shown that exposure to maternal obesity around the time of conception alone resulted in increased adiposity in female lambs. Changes in the abundance of insulin signalling molecules in skeletal muscle and adipose tissue precede the development of insulin resistance and type 2 diabetes. It is not clear, however, whether exposure to maternal obesity results in insulin resistance in her offspring as a consequence of the impact of increased adiposity on skeletal muscle or as a consequence of the programming of specific changes in the abundance of insulin signalling molecules in this tissue. We have used an embryo transfer model in the sheep to investigate the effects of exposure to either maternal obesity or to weight loss in normal and obese mothers preceding and for one week after conception on the expression and abundance of insulin signalling molecules in muscle in the offspring. We found that exposure to maternal obesity resulted in lower muscle GLUT-4 and Ser 9 phospho-GSK3α and higher muscle GSK3α abundance in lambs when compared to lambs conceived in normally nourished ewes. Exposure to maternal weight loss in normal or obese mothers, however, resulted in lower muscle IRS1, PI3K, p110ß, aPKCζ, Thr 642 phospho-AS160 and GLUT-4 abundance in the offspring. In conclusion, maternal obesity or weight loss around conception have each programmed specific changes on subsets of molecules in the insulin signalling, glucose transport and glycogen synthesis pathways in offspring. There is a need for a stronger evidence base to ensure that weight loss regimes in obese women seeking to become pregnant minimize the metabolic costs for the next generation.


Fertilization , Insulin Resistance , Insulin/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Prenatal Exposure Delayed Effects/metabolism , Signal Transduction , Weight Loss , Animals , Female , Muscle Proteins/metabolism , Muscle, Skeletal/pathology , Obesity/pathology , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Sheep
14.
Proteomics ; 12(22): 3386-92, 2012 Nov.
Article En | MEDLINE | ID: mdl-22997143

Studies of neuronal, endocrine, and metabolic disorders would be facilitated by characterization of the hypothalamus proteome. Protein extracts prepared from 16 whole rat hypothalami were measured by data-independent label-free nano LC-MS/MS. Peptide features were detected, aligned, and searched against a rat Swiss-Prot database using ProteinLynx Global Server v.2.5. The final combined dataset comprised 21 455 peptides, corresponding to 622 unique proteins, each identified by a minimum of two distinct peptides. The majority of the proteins (69%) were cytosolic, and 16% were membrane proteins. Important proteins involved in neurological and synaptic function were identified including several members of the Ras-related protein family and proteins involved in glutamate biosynthesis.


Chromatography, Liquid/methods , Hypothalamus/chemistry , Nerve Tissue Proteins/analysis , Proteome/analysis , Tandem Mass Spectrometry/methods , Animals , Biomarkers/analysis , Databases, Protein , Hypothalamus/metabolism , Isoelectric Point , Nerve Tissue Proteins/chemistry , Peptide Fragments/analysis , Proteome/chemistry , Proteomics , Rats , Rats, Wistar
15.
Rev Endocr Metab Disord ; 13(2): 85-92, 2012 Jun.
Article En | MEDLINE | ID: mdl-22430227

It is well established that there is a relationship between patterns of early growth and subsequent risk of development of metabolic diseases such as type 2 diabetes and cardiovascular disease. Studies in both humans and in animal models have provided strong evidence that the early environment plays an important role in mediating these relationships. The concept of the developmental origins of health and disease is therefore widely accepted. The mechanisms by which an event in very early life can have a permanent effect on the long-term health of an individual are still relatively poorly understood. However a growing body of evidence has implicated a number of candidate mechanisms. These include permanent changes in an organ structure, programmed changes in gene expression through epigenetic modifications and persistent effects on regulation of cellular ageing. Understanding the extent and nature of these processes may enable the identification of individuals at risk of metabolic disease as well as providing insight into potential preventative and intervention strategies.


Child Development/physiology , Disease , Fetal Development/physiology , Animals , Cellular Senescence/physiology , Child , Epidemiologic Studies , Epigenesis, Genetic/physiology , Female , Gene Expression Regulation, Developmental , Growth/physiology , Humans , Infant, Low Birth Weight/physiology , Infant, Newborn , Mortality , Pregnancy , Risk Assessment
16.
PLoS One ; 7(12): e52363, 2012.
Article En | MEDLINE | ID: mdl-23300651

Before birth, glucocorticoids retard growth, although the extent to which this is mediated by changes in insulin signalling pathways in the skeletal muscle of the fetus is unknown. The current study determined the effects of endogenous and synthetic glucocorticoid exposure on insulin signalling proteins in skeletal muscle of fetal sheep during late gestation. Experimental manipulation of fetal plasma glucocorticoid concentration was achieved by fetal cortisol infusion and maternal dexamethasone treatment. Cortisol infusion significantly increased muscle protein levels of Akt2 and phosphorylated Akt at Ser473, and decreased protein levels of phosphorylated forms of mTOR at Ser2448 and S6K at Thr389. Muscle GLUT4 protein expression was significantly higher in fetuses whose mothers were treated with dexamethasone compared to those treated with saline. There were no significant effects of glucocorticoid exposure on muscle protein abundance of IR-ß, IGF-1R, PKCζ, Akt1, calpastatin or muscle glycogen content. The present study demonstrated that components of the insulin signalling pathway in skeletal muscle of the ovine fetus are influenced differentially by naturally occurring and synthetic glucocorticoids. These findings may provide a mechanism by which elevated concentrations of endogenous glucocorticoids retard fetal growth.


Dexamethasone/pharmacology , Fetus/cytology , Hydrocortisone/pharmacology , Insulin/metabolism , Muscle, Skeletal/cytology , Sheep , Signal Transduction/drug effects , Animals , Dexamethasone/administration & dosage , Dexamethasone/blood , Female , Fetus/drug effects , Hydrocortisone/administration & dosage , Hydrocortisone/blood , Mothers , Muscle, Skeletal/drug effects , Pregnancy
17.
Endocrinology ; 152(2): 476-82, 2011 Feb.
Article En | MEDLINE | ID: mdl-21209019

Low birth weight and rapid postnatal weight gain are independent and additive risk factors for the subsequent development of metabolic disease. Despite an abundance of evidence for these associations, mechanistic data are lacking. The hormone leptin has received significant interest as a potential programming factor, because differences in the profile of leptin in early life have been associated with altered susceptibility to obesity. Whether leptin alone is a critical factor for programming obesity has, until now, remained unclear. Using the leptin-deficient ob/ob mouse, we show that low birth weight followed by rapid catch-up growth during lactation (recuperated offspring) leads to a persistent increase in body weight in adult life, both in wild-type and ob/ob animals. Furthermore, recuperated offspring are hyperphagic and epididymal fat pad weights are significantly increased, reflecting greater adiposity. These results show definitively that factors other than leptin are crucial in the programming of energy homeostasis in this model and are powerful enough to alter adiposity in a genetically obese strain.


Adiposity/physiology , Body Weight/physiology , Leptin/metabolism , Adiposity/genetics , Animals , Body Weight/genetics , Eating/genetics , Eating/physiology , Enzyme-Linked Immunosorbent Assay , Female , Leptin/genetics , Lipids/blood , Male , Mice , Mice, Mutant Strains
18.
J Hepatol ; 52(6): 913-20, 2010 Jun.
Article En | MEDLINE | ID: mdl-20413174

BACKGROUND & AIMS: Obesity induced, non-alcoholic fatty liver disease (NAFLD), is now the major cause in affluent countries, of the spectrum of steatosis-to-cirrhosis. Obesity and NAFLD rates in reproductive age women, and adolescents, are rising worldwide. Our hypothesis was that maternal obesity and lactation transmit to the offspring a pre-disposition to dysmetabolism, obesity and NAFLD. METHODS: Female mice were fed standard or obesogenic chow, before, throughout pregnancy, and during lactation. The critical developmental period was studied by cross-fostering offspring of lean and obese dams. Offspring were then weaned onto standard chow and studied at 3months. Read-outs included markers of metabolic dysfunction, biochemical and histological indicators of NAFLD, induction of liver fibrogenesis, and activation of pro-fibrotic pathways. Mechanisms involved in programming a dysmetabolic and NAFLD phenotype were investigated by assaying breast milk components. RESULTS: Offspring of obese dams had a dysmetabolic, insulin resistant and NAFLD phenotype compared to offspring of lean dams. Offspring of lean dams that were suckled by obese dams showed an exaggerated dysmetabolic and NAFLD phenotype, with increased body weight, as well as increased levels of insulin, leptin, aspartate transaminase, interleukin-6, tumour necrosis factor-alpha, liver triglycerides, steatosis, hepatic fibrogenesis, renal norepinephrine, and liver alpha1-D plus beta1-adrenoceptors, indicative of sympathetic nervous system activation. Obese dams also had raised breast milk leptin levels compared to lean dams. CONCLUSIONS: Maternal obesity programs development of a dysmetabolic and NAFLD phenotype, which is critically dependent on the early postnatal period and possibly involving alteration of hypothalamic appetite nuclei signalling by maternal breast milk and neonatal adipose tissue derived, leptin.


Fatty Liver/etiology , Lactation , Metabolic Syndrome/etiology , Obesity/complications , Pregnancy Complications/physiopathology , Prenatal Exposure Delayed Effects/physiopathology , Actins/genetics , Adipose Tissue/metabolism , Animals , Collagen/genetics , Collagen Type I , Fatty Liver/pathology , Fatty Liver/physiopathology , Female , Gene Expression/physiology , Interleukin-6/genetics , Leptin/metabolism , Metabolic Syndrome/pathology , Metabolic Syndrome/physiopathology , Mice , Mice, Inbred C57BL , Milk/metabolism , Obesity/metabolism , Obesity/physiopathology , Pregnancy , Pregnancy Complications/metabolism , Pregnancy Complications/pathology , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Receptors, Adrenergic, alpha-1/genetics , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/genetics
19.
J Nutr ; 140(3): 662-6, 2010 Mar.
Article En | MEDLINE | ID: mdl-20107142

Epidemiological studies have revealed a relationship between poor early growth and development of type 2 diabetes and other features of metabolic syndrome. The mechanistic basis of this relationship is not known. However, compelling evidence suggests that early environmental factors, including nutrition, play an important role. Studies of individuals in utero during a period of famine showed a direct relationship between maternal nutrition and glucose tolerance. Further evidence has come from studies of monozygotic twins who were discordant for type 2 diabetes. Nutrition during the early postnatal period has also been shown to have long-term consequences on metabolic health. Excess nutrition and accelerated growth during the neonatal period has been suggested to be particularly detrimental. Animal models, including maternal protein restriction, have been developed to elucidate mechanisms linking the early environment and future disease susceptibility. Maternal protein restriction in rats leads to a low birth weight and development of type 2 diabetes in the offspring. This is associated with beta cell dysfunction and insulin resistance. The latter is associated with changes in expression of key components of the insulin-signaling cascade in muscle and adipocytes similar to that observed in tissue from young men with a low birth weight. These differences occur prior to development of disease and thus may represent molecular markers of early growth restriction and disease risk. The fundamental mechanisms by which these programmed changes occur remain to be fully defined but are thought to involve epigenetic mechanisms.


Diabetes Mellitus, Type 2/complications , Infant Nutritional Physiological Phenomena , Obesity/complications , Animals , Diabetes Mellitus, Type 2/epidemiology , Female , Humans , Infant , Infant, Newborn , Male , Obesity/epidemiology , Pregnancy , Rats , Risk Factors
20.
Am J Physiol Regul Integr Comp Physiol ; 297(3): R675-81, 2009 Sep.
Article En | MEDLINE | ID: mdl-19535678

We recently reported insulin resistance in adult offspring of obese C57BL/6J mice. We have now evaluated whether parameters of skeletal muscle structure and function may play a role in insulin resistance in this model of developmental programming. Obesity was induced in female mice by feeding a highly palatable sugar and fat-rich diet for 6 wk prior to pregnancy, and during pregnancy and lactation. Offspring of obese dams were weaned onto standard laboratory chow. At 3 mo of age, skeletal muscle insulin signaling protein expression, mitochondrial electron transport chain activity (ETC), muscle fiber type, fiber density, and fiber cross-sectional area were compared with that of offspring of control dams weaned onto the chow diet. Female offspring of obese dams demonstrated decreased skeletal muscle expression of p110beta, the catalytic subunit of PI3K (P < 0.01), as well as reduced Akt phosphorylation at Serine residue 473 compared with control offspring. Male offspring of obese dams demonstrated increased skeletal muscle Akt2 and PKCzeta expression (P < 0.01; P < 0.001, respectively). A decrease in mitochondrial-linked complex II-III was observed in male offspring of obese dams (P < 0.01), which was unrelated to CoQ deficiency. This was not observed in females. There were no differences in muscle fiber density between offspring of obese dams and control offspring in either sex. Sex-related alterations in key insulin-signaling proteins and in mitochondrial ETC may contribute to a state of insulin resistance in offspring of obese mice.


Electron Transport Complex III/metabolism , Electron Transport Complex II/metabolism , Insulin Resistance , Insulin/metabolism , Mitochondria, Muscle/metabolism , Obesity/metabolism , Quadriceps Muscle/metabolism , Signal Transduction , Animal Nutritional Physiological Phenomena , Animals , Body Weight , Class I Phosphatidylinositol 3-Kinases , Disease Models, Animal , Female , Glucose Transporter Type 4/metabolism , Insulin Receptor Substrate Proteins/metabolism , Male , Maternal Nutritional Physiological Phenomena , Mice , Mice, Inbred C57BL , Mitochondria, Muscle/enzymology , Muscle Fibers, Skeletal/metabolism , Obesity/pathology , Obesity/physiopathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Pregnancy , Prenatal Exposure Delayed Effects , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quadriceps Muscle/enzymology , Quadriceps Muscle/pathology , Receptor, Insulin/metabolism , Sex Factors , Ubiquinone/metabolism
...