Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Int J Obes (Lond) ; 37(8): 1051-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23229735

ABSTRACT

BACKGROUND: It is now widely accepted that the early-life nutritional environment is important in determining susceptibility to metabolic diseases. In particular, intra-uterine growth restriction followed by accelerated postnatal growth is associated with an increased risk of obesity, type-2 diabetes and other features of the metabolic syndrome. The mechanisms underlying these observations are not fully understood. AIM: Using a well-established maternal protein-restriction rodent model, our aim was to determine if exposure to mismatched nutrition in early-life programmes adipose tissue structure and function, and expression of key components of the insulin-signalling pathway. METHODS: Offspring of dams fed a low-protein (8%) diet during pregnancy were suckled by control (20%)-fed dams to drive catch-up growth. This 'recuperated' group was compared with offspring of dams fed a 20% protein diet during pregnancy and lactation (control group). Epididymal adipose tissue from 22-day and 3-month-old control and recuperated male rats was studied using histological analysis. Expression and phosphorylation of insulin-signalling proteins and gene expression were assessed by western blotting and reverse-transcriptase PCR, respectively. RESULTS: Recuperated offspring at both ages had larger adipocytes (P<0.001). Fasting serum glucose, insulin and leptin levels were comparable between groups but increased with age. Recuperated offspring had reduced expression of IRS-1 (P<0.01) and PI3K p110ß (P<0.001) in adipose tissue. In adult recuperated rats, Akt phosphorylation (P<0.01) and protein levels of Akt-2 (P<0.01) were also reduced. Messenger RNA expression levels of these proteins were not different, indicating a post-transcriptional effect. CONCLUSION: Early-life nutrition programmes alterations in adipocyte cell size and impairs the protein expression of several insulin-signalling proteins through post-transcriptional mechanisms. These indices may represent early markers of insulin resistance and metabolic disease risk.


Subject(s)
Diabetes Mellitus, Type 2/pathology , Fetal Growth Retardation/pathology , Insulin Resistance , Metabolic Syndrome/pathology , Obesity/pathology , Proto-Oncogene Proteins c-akt/metabolism , Adipocytes , Adipose Tissue/pathology , Animals , Blotting, Western , Body Weight , Female , Fetal Development , Fetal Growth Retardation/metabolism , Gene Expression , Insulin , Male , Metabolic Syndrome/metabolism , Phenotype , Phosphorylation , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Wistar
2.
Diabetes Obes Metab ; 14 Suppl 3: 29-39, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22928562

ABSTRACT

Type 2 diabetes (T2D), also known as non-insulin dependent diabetes mellitus, arises as a consequence of peripheral insulin resistance in combination with an inability of pancreatic islet ß-cells to secrete adequate amounts of insulin. It is widely recognized that the current environment (e.g. an unhealthy diet and sedentary lifestyle) contributes to this process. In recent years, however, the role of the early environment, particularly nutrition, has emerged as an important factor capable of influencing health and disease risk of an individual, including risk of T2D. The impact of early environment on glucose metabolism has been extensively studied. Compelling evidence from epidemiological studies and animal models suggests that early nutrition can affect insulin action as a mediator of glucose homeostasis in peripheral tissues and as an important regulator of appetite and body weight. The early environment can also affect ß-cell mass and function, and hence insulin secretion. The molecular mechanisms underlying the relationship between a suboptimal early environment and impaired insulin action and secretion is thought to include epigenetic modifications of the foetal genome, oxidative stress and mitochondrial dysfunction.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Infant Nutritional Physiological Phenomena , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Maternal Nutritional Physiological Phenomena , Prenatal Exposure Delayed Effects/metabolism , Animals , Birth Weight , Blood Glucose/genetics , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Epigenesis, Genetic , Female , Humans , Infant, Newborn , Insulin Resistance , Insulin Secretion , Male , Pregnancy , Prenatal Exposure Delayed Effects/genetics
3.
Int J Obes (Lond) ; 36(8): 1040-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22124449

ABSTRACT

BACKGROUND: Pups of normally nourished dams that are cross-fostered after birth to dams fed a low-protein (8% by weight) diet (postnatal low protein (PLP)) grow slower during the suckling period and remain small and lean throughout adulthood. At weaning, they have increased expression in the arcuate nucleus (ARC) of the hypothalamus of the orexigenic neuropeptide Y (NPY) and decreased expression of pro-opiomelanocortin, the precursor of anorexigenic melanocortins. OBJECTIVES AND METHODS: We investigated, using third ventricle administration, whether 3-month-old male PLP rats display altered sensitivity to leptin with respect to food intake, NPY and the melanocortin 3/4-receptor agonist MTII, and using in situ hybridization or laser capture microdissection of the ARC followed by RT-PCR, whether the differences observed were associated with changes in the hypothalamic expression of NPY or the leptin receptor, NPY receptors and melanocortin receptors. RESULTS: PLP rats were smaller and had reduced percentage body fat content and plasma leptin concentration compared with control rats. Leptin (5 µg) reduced food intake over 0-48 h more in PLP than control rats (P<0.05). Submaximal doses of NPY increased the food intake less in PLP rats than in controls, whereas submaximal doses of MTII reduced the food intake more in PLP rats. Maximal responses did not differ between PLP and control rats. Leptin and melanocortin-3 receptor (MC3R) expression were increased in both ARC and ventromedial hypothalamic nuclei in PLP animals compared with the controls. MC4R, NPY Y1R, Y5R and NPY expression were unchanged. CONCLUSION: Postnatal undernourishment results in food intake in adult rats being more sensitive to reduction by leptin and melanocortins, and less sensitive to stimulation by NPY. We propose that this contributes to increased leptin sensitivity and resistance to obesity. Increased expression of ObRb and MC3R may partly explain these findings but other downstream mechanisms must also be involved.


Subject(s)
Animals, Newborn/growth & development , Arcuate Nucleus of Hypothalamus/pathology , Leptin/metabolism , Neuropeptide Y/metabolism , Obesity/genetics , Receptor, Melanocortin, Type 3/metabolism , Thinness/genetics , Animals , Arcuate Nucleus of Hypothalamus/physiology , Body Weight/genetics , Disease Susceptibility , Eating , Gene Expression Regulation , Leptin/pharmacology , Male , Neuropeptide Y/pharmacology , Obesity/metabolism , Rats , Rats, Wistar , Thinness/metabolism , Time Factors , Weight Gain/genetics
4.
FASEB J ; 25(1): 420-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20923964

ABSTRACT

Molecular mechanisms predisposing people to insulin resistance are starting to emerge. Altered insulin signaling for hepatic gluconeogenesis and muscle glucose uptake is thought to play a central role. Development under suboptimal conditions is also known to increase the risk of insulin resistance in adulthood. However, the partial contributions of reduced oxygen vs. nutrient delivery to the fetus, two common adverse conditions in utero, to developmental programming of insulin resistance remain unknown. The aim of this study was to determine the effects of developmental hypoxia or undernutrition on the expression of insulin-signaling proteins in liver and skeletal muscle in adult rat offspring. We show that the expression of hepatic phospho-Akt and muscle Akt2 were significantly reduced in offspring of hypoxic, relative to offspring from normoxic or undernourished, pregnancies. Hepatic Akt-1, Akt-2, and PKCζ protein expression was reduced in offspring from both hypoxic and undernourished pregnancies. Muscle GLUT4 expression was decreased in undernourished, and further decreased in hypoxic, offspring. These findings link prenatal hypoxia to down-regulation of components of hepatic and muscle Akt expression in adult offspring. Akt may represent a pharmaceutical target for clinical intervention against the developmental programming of metabolic disease resulting from prenatal hypoxia.


Subject(s)
Biomarkers/metabolism , Hypoxia/physiopathology , Insulin Resistance/physiology , Prenatal Exposure Delayed Effects , Animals , Blotting, Western , Body Weight , Female , Glucose Transporter Type 4/metabolism , Insulin/blood , Lipids/blood , Litter Size , Liver/metabolism , Male , Malnutrition/physiopathology , Muscle, Skeletal/metabolism , Pregnancy , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Time Factors
5.
Carcinogenesis ; 31(10): 1873-81, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20460357

ABSTRACT

We previously showed that offspring of rat dams receiving a protein-restricted (low protein) diet throughout pregnancy and lactation develop mammary tumors more quickly. Rapid post-weaning mammary growth and mammary tissue overexpression of insulin receptor, insulin-like growth factor-1 receptor (IGF-1R), estrogen receptor isoform alpha and v-erb-b2 erythroblastic leukemia viral oncogene homolog 2 (ERBB2), correlated with this risk. The objectives of this study were therefore (i) to identify underlying mechanisms of increased risk through candidate and global approaches; (ii) to determine if excessive calorie intake further increased risk and if so, (iii) to identify the molecular mechanisms mediating this. We provide evidence for transcriptional upregulation of IGF-1R by Sp1 in LP mammary tissue (P < 0.01). Cell cycle control and DNA damage repair gene cyclin-dependent kinase inhibitor 1A (CDKN1A) (p21waf1) was also upregulated (P < 0.05) as was transcription factor nuclear factor of kappa light polypeptide gene enhancer in B-cell (P < 0.05) and adhesion gene CDH1 (P < 0.05). Invasion and metastasis markers matrix metalloproteinase 9 and serpin peptidase inhibitor, clade E, member 1 (SERPIN1) were upregulated (both P < 0.05), whereas metastasis suppressor gene NME1 was downregulated (P < 0.01). Feeding a highly palatable diet (HPD) to increase calorie intake from puberty, additively and independently increased early mammary tumor risk, which correlated with increased serum insulin and triglyceride concentrations (P < 0.05). PTEN gene expression was reduced both by early protein restriction (P < 0.05) and HPD (P < 0.01), which may induce Akt in cell survival pathways. Progesterone receptor and ERBB2 (both P < 0.05) expression increased as an effect of an interaction between maternal diet and adult nutrition, with subsequent downstream activation of the mitogen-activated protein kinase pathway. We conclude that poor early growth and excessive calorie intake exert independent and additive effects on mitogenic growth factor signaling to influence mammary tumor susceptibility.


Subject(s)
Energy Intake , Growth Disorders/complications , Mammary Neoplasms, Animal/etiology , Signal Transduction , Animals , Body Weight , Disease Susceptibility , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Profiling , Mammary Glands, Animal/chemistry , Mammary Glands, Animal/metabolism , Rats , Rats, Wistar , Receptor, ErbB-2/genetics , Receptor, ErbB-2/physiology , Receptor, IGF Type 1/analysis , Receptor, IGF Type 1/genetics , Sp1 Transcription Factor/analysis , Sp1 Transcription Factor/genetics
6.
J Dev Orig Health Dis ; 1(3): 184-91, 2010 Jun.
Article in English | MEDLINE | ID: mdl-25141786

ABSTRACT

Individuals exposed in utero to maternal obesity have increased risk of developing type 2 diabetes mellitus and obesity in adulthood. The molecular mechanisms underlying this association are unknown. We have therefore used a murine model of maternal obesity, in which the offspring of obese dams develop hyperinsulinaemia by 3 months of age indicative of insulin resistance. Here, we investigate the effects of maternal diet-induced obesity on the expression/phosphorylation of key hepatic insulin signalling proteins and the expression of anti-oxidant enzymes in male offspring. At 3 months of age, offspring of obese dams had decreased levels of insulin receptor substrate (IRS) 1 (P < 0.01), whereas the ratio of phosphorylated IRS1 Ser307 to total IRS1 was significantly increased (P < 0.001), suggesting that it was less active. Protein expression of the PI3K p85α subunit was decreased (P < 0.01) and there was a tendency for phosphorylation of Akt at Ser473 to be reduced (P = 0.08) in the offspring of obese dams. protein kinase Cζ (P < 0.001) and glycogen synthase kinase 3ß (P < 0.05) levels were increased in these animals in comparison with controls. Maternal obesity also resulted in increased phosphorylation of p38 mitogen-activated protein kinase at Thr180/Tyr182 (P < 0.01) and raised c-Jun N-terminal kinase 1 expression (P < 0.5) in the offspring. The expression of antioxidant enzymes was also affected by maternal obesity with CuZnSOD (P < 0.001) and glutathione reductase (P < 0.05) being increased, whereas glutathione peroxidase 1 was reduced (P < 0.05) in the offspring. We conclude that maternal obesity leads to alterations in hepatic insulin signalling protein expression and phosphorylation. These molecular changes may contribute to the development of insulin resistance.

7.
FASEB J ; 22(6): 2037-44, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18230683

ABSTRACT

Low birth weight is associated with increased cardiovascular disease (CVD) in humans. Detrimental effects of low birth weight are amplified by rapid catch-up growth. Conversely, slow growth during lactation reduces CVD risk. Gestational protein restriction causes low birth weight, vascular dysfunction, and accelerated aging in rats. Atherosclerotic aortic tissue has shortened telomeres, and oxidative stress accelerates telomere shortening through generation of DNA single-strand breaks (ssbs). This study tested the hypothesis that maternal diet influences aortic telomere length through changes in DNA ssbs, antioxidant capacity, and oxidative stress. We used our models of gestational protein restriction followed by rapid catch-up growth (the recuperated group) and protein restriction during lactation (the postnatal low-protein [PLP] group). Southern blotting revealed fewer aortic DNA ssbs and subsequently fewer short telomeres (P<0.05) in the PLP group. This result was associated with reduced (P<0.01) 8-hydroxy-2-deoxyguanosine, a marker of oxidative stress. PLP animals expressed increased (P<0.01) manganese superoxide-dismutase, copper-zinc superoxide-dismutase, catalase, and glutathione-reductase. Age-dependent changes in antioxidant defense enzymes indicated more protection to oxidative stress in the PLP animals; conversely, recuperated animals demonstrated age-associated impairment of antioxidant defenses. We conclude that maternal diet has a major influence on aortic telomere length. This finding may provide a mechanistic link between early growth patterns and CVD.


Subject(s)
Antioxidants , DNA Damage , Diet , Oxidative Stress , Telomere/ultrastructure , Age Factors , Animals , Aorta/ultrastructure , Female , Models, Animal , Mothers , Rats
8.
J Intern Med ; 261(5): 437-52, 2007 May.
Article in English | MEDLINE | ID: mdl-17444883

ABSTRACT

It is widely accepted that an association exists between the intrauterine environment in which a fetus grows and develops and the subsequent development of type 2 diabetes. Any disturbance in maternal ability to provide nutrients and oxygen to the fetus can lead to fetal intrauterine growth restriction (IUGR). Here we will review IUGR in rodent models, in which maternal metabolism has been experimentally manipulated to investigate the molecular basis of the relationship between IUGR and development of type 2 diabetes in later life, and the identification of the molecular derangements in specific metabolically - sensitive organs/tissues.


Subject(s)
Diabetes Mellitus, Type 2/etiology , Fetal Growth Retardation/metabolism , Prenatal Exposure Delayed Effects , Animals , Caloric Restriction , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes, Gestational/metabolism , Dietary Proteins/administration & dosage , Disease Models, Animal , Epigenesis, Genetic/genetics , Female , Fetal Growth Retardation/genetics , Fetal Nutrition Disorders/metabolism , Glucocorticoids/adverse effects , Humans , Insulin/metabolism , Mitochondrial Diseases/metabolism , Placenta/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism , Signal Transduction/physiology , Uterus/metabolism
9.
Diabetologia ; 49(12): 2993-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17063325

ABSTRACT

AIMS/HYPOTHESIS: Individuals with low birthweight are at increased risk of type 2 diabetes mellitus. However, the underlying molecular mechanisms are unknown. Previously we have shown that low birthweight is associated with changes in muscle insulin signalling proteins. Here we determined whether low birthweight is associated with changes in insulin signalling proteins in adipose tissue. METHODS: Men (age 23 years) with either a low (bottom 10th percentile) (n = 17) or a normal (50th-90th percentile) (n = 17) birthweight were recruited from the Danish Medical Birth Registry and subcutaneous adipose biopsies were taken. RESULTS: Between the two groups there was no difference in protein level of the insulin receptor, protein kinase C zeta, glycogen synthase kinase-3 (GSK3) alpha, GSK3 beta, protein kinase B alpha and beta, peroxisome proliferative activated receptor gamma coactivator 1 or Src-homology-2-containing protein. However, the levels of GLUT4 (also known as solute carrier family 2 [facilitated glucose transporter], member 4 [SLC2A4]) (52 +/- 10.9% reduction, p < 0.01), p85alpha subunit of phosphoinositide 3-kinase (PI3K) (45 +/- 9% reduction, p < 0.01), p110ss subunit of PI3K (48 +/- 17% reduction, p = 0.06) and IRS1 (59 +/- 24% reduction, p < 0.05) were reduced in men of low birthweight. CONCLUSIONS/INTERPRETATION: These findings show that low birthweight is associated with reduced levels of adipose insulin signalling proteins, thus providing a potential molecular framework to explain why people with low birthweight are at increased risk of developing type 2 diabetes. These differences precede the development of diabetes and thus may help predict disease risk.


Subject(s)
Blood Proteins/metabolism , Diabetes Mellitus, Type 2/epidemiology , Infant, Low Birth Weight , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adult , Biopsy , Birth Weight , Glucose Transporter Type 4/genetics , Glycogen Synthase Kinase 3/genetics , Humans , Infant, Newborn , Male , Phosphatidylinositol 3-Kinases/genetics , RNA, Messenger/genetics , Risk Assessment
10.
Biochem Soc Trans ; 34(Pt 5): 779-82, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17052196

ABSTRACT

The ability of mother to provide nutrients and oxygen for her baby is a critical factor for fetal health and its survival. Failure in supplying the adequate amount of nutrients to meet fetal demand can lead to fetal malnutrition. The fetus responds and adapts to undernutrition but by doing so it permanently alters the structure and function of the body. Maternal overnutrition also has long-lasting and detrimental effects on the health of the offspring. There is growing evidence that maternal nutrition can induce epigenetic modifications of the fetal genome. Only relatively recently has evidence from epidemiological and animal studies emerged suggesting that fetal responses to the intrauterine environment may underlie the prevalence of many chronic diseases of adulthood including Type 2 (non-insulin-dependent) diabetes. It is now of crucial importance to gain the understanding of the molecular mechanisms underlying the relationship between fetal alterations to the intra-uterine environment and their long-term effects on the health of an individual.


Subject(s)
Child Welfare , Maternal Nutritional Physiological Phenomena , Child , Female , Fetus/physiology , Health Status , Humans , Infant, Newborn , Malnutrition/physiopathology , Pregnancy , Pregnancy Complications/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...