Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Clin Exp Allergy ; 48(7): 773-786, 2018 07.
Article in English | MEDLINE | ID: mdl-29772098

ABSTRACT

Outdoor air pollution is a major environmental health problem throughout the world. In particular, exposure to particulate matter (PM) has been associated with the development and exacerbation of several respiratory diseases, including asthma. Although the adverse health effects of PM have been demonstrated for many years, the underlying mechanisms have not been fully identified. In this review, we focus on the role of the lung epithelium and specifically highlight multiple cytokines in PM-induced respiratory responses. We describe the available literature on the topic including in vitro studies, findings in humans (ie observations in human cohorts, human controlled exposure and ex vivo studies) and in vivo animal studies. In brief, it has been shown that exposure to PM modulates the airway epithelium and promotes the production of several cytokines, including IL-1, IL-6, IL-8, IL-25, IL-33, TNF-α, TSLP and GM-CSF. Further, we propose that PM-induced type 2-promoting cytokines are important mediators in the acute and aggravating effects of PM on airway inflammation. Targeting these cytokines could therefore be a new approach in the treatment of asthma.


Subject(s)
Air Pollutants/adverse effects , Air Pollution/adverse effects , Allergens/immunology , Bronchial Hyperreactivity/immunology , Hypersensitivity/immunology , Particulate Matter/adverse effects , Animals , Asthma/immunology , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Humans , Hypersensitivity/genetics , Hypersensitivity/metabolism , Hypersensitivity/pathology , Inflammation Mediators/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology
2.
J Neuroendocrinol ; 29(6)2017 06.
Article in English | MEDLINE | ID: mdl-28561903

ABSTRACT

The underlying hypothalamic neurocircuitry by which metabolism and feeding regulates reproductive function has been well-studied in the rodent; however, recent data have demonstrated significant neuroanatomical differences in the human brain. The present study had three objectives, centred on arcuate nucleus neuropeptides regulating feeding and reproduction: (i) to characterise coexpression patterns in the female nonhuman primate; (ii) to establish whether these neuronal populations make potential contacts with gonadotophin-releasing hormone (GnRH) neurones; and (iii) to determine whether these contacts differ between the low and high GnRH-releasing states of pre-puberty and adulthood, respectively. Female nonhuman primates have several coexpression patterns of hypothalamic neuropeptides that differ from those reported in rodents. Cocaine- and amphetamine-regulated transcript (CART) is not coexpressed with pro-opiomelanocortin but instead with neuropeptide Y (NPY). CART is also expressed in a subpopulation of kisspeptin cells in the nonhuman primate, similar to observations in humans but diverging from findings in rodents. Very few GnRH-expressing neurones received close appositions from double-labelled kisspeptin/CART fibres; however, both single-labelled kisspeptin and CART fibres were in frequent apposition with GnRH neurones, with no differences between prepubertal and adult animals. NPY/agouti-related peptide (AgRP) coexpressing fibres contacted significantly more GnRH neurones in prepubertal animals than adults, consistent with increased NPY and AgRP mRNA observed in prepubertal animals. The findings of the present study detail significant differences in arcuate nucleus neuropeptide coexpression in the monkey compared to the rodent and are consistent with the hypothesis that arcuate nucleus NPY/AgRP neurones play an inhibitory role in controlling GnRH neuronal regulation in the prepubertal primate.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Gonadotropin-Releasing Hormone/metabolism , Macaca mulatta , Age Factors , Agouti-Related Protein/metabolism , Animals , Cell Count , Female , Kisspeptins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuropeptide Y/metabolism
3.
Nutr Diabetes ; 7(1): e242, 2017 01 16.
Article in English | MEDLINE | ID: mdl-28092346

ABSTRACT

BACKGROUND/OBJECTIVE: Intake of high-energy foods and maternal nutrient overload increases the risk of metabolic diseases in the progeny such as obesity and diabetes. We hypothesized that maternal and postnatal intake of chocolate and soft drink will affect leptin sensitivity and hypothalamic astrocyte morphology in adult rat offspring. METHODS: Pregnant Sprague-Dawley rats were fed ad libitum chow diet only (C) or with chocolate and high sucrose soft drink supplement (S). At birth, litter size was adjusted into 10 male offspring per mother. After weaning, offspring from both dietary groups were assigned to either S or C diet, giving four groups until the end of the experiment at 26 weeks of age. RESULTS: As expected, adult offspring fed the S diet post weaning became obese (body weight: P<0.01, %body fat per kg: P<0.001) and this was due to the reduced energy expenditure (P<0.05) and hypothalamic astrogliosis (P<0.001) irrespective of maternal diet. Interesting, offspring born to S-diet-fed mothers and fed the S diet throughout postnatal life became obese despite lower energy intake than controls (P<0.05). These SS offspring showed increased feed efficiency (P<0.001) and reduced fasting pSTAT3 activity (P<0.05) in arcuate nucleus (ARC) compared with other groups. The findings indicated that the combination of the maternal and postnatal S-diet exposure induced persistent changes in leptin signalling, hence affecting energy balance. Thus, appetite regulation was more sensitive to the effect of leptin than energy expenditure, suggesting differential programming of leptin sensitivity in ARC in SS offspring. Effects of the maternal S diet were normalized when offspring were fed a chow diet after weaning. CONCLUSIONS: Maternal intake of chocolate and soft drink had long-term consequences for the metabolic phenotype in the offspring if they continued on the S diet in postnatal life. These offspring displayed obesity despite lowered energy intake associated with alterations in hypothalamic leptin signalling.


Subject(s)
Carbonated Beverages , Chocolate , Hypothalamus/metabolism , Leptin/metabolism , Maternal Nutritional Physiological Phenomena , Prenatal Exposure Delayed Effects/metabolism , Animals , Energy Metabolism/drug effects , Feeding Behavior , Female , Hypothalamus/drug effects , Leptin/pharmacology , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Leptin/metabolism , Signal Transduction/drug effects
4.
Int J Obes (Lond) ; 40(10): 1600-1603, 2016 10.
Article in English | MEDLINE | ID: mdl-27357159

ABSTRACT

The inability to augment capillary blood volume (CBV) in response to insulin or glucose is thought to contribute to insulin resistance (IR) by limiting glucose uptake in key storage sites. Understanding the mechanisms that contribute to impaired CBV augmentation early in the onset of IR may lead to new future therapies. We hypothesized that inactivity alters the balance of vasoactive eicosanoids and contributes to microvascular IR. In ten activity-restricted (AR) and six normal activity adult male rhesus macaques, contrast-enhanced ultrasound of skeletal muscle blood flow and CBV was performed at baseline and during intravenous glucose tolerance test (IVGTT). Plasma was analyzed for vasoconstrictor hydroxyeicosatetraenoic acids (HETEs) and the ratio of vasodilatory epoxyeicosatrienoic acids (EETs) to their less biologically active dihydroxyeicosatrienoic acids (DHETs) as an indirect measure of soluble epoxide hydrolase activity. AR primates were IR during IVGTT and had a 45% lower glucose-stimulated CBV response. Vasoconstrictor 18-HETE and 19-HETE and the DHET/EET ratio were markedly elevated in the AR group and correlated inversely with the CBV response. In addition, levels of 18-HETE and 19-HETE correlated directly with microvascular IR. We conclude that a shift toward increased eicosanoid vasoconstrictor tone correlates with abnormal skeletal muscle vascular recruitment and may contribute to IR.


Subject(s)
Hydroxyeicosatetraenoic Acids/pharmacology , Insulin Resistance/physiology , Microcirculation/drug effects , Muscle, Skeletal/blood supply , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Blood Volume/drug effects , Capillaries/drug effects , Cytochrome P-450 Enzyme System/metabolism , Disease Models, Animal , Glucose Tolerance Test , Macaca mulatta , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Regional Blood Flow/drug effects
5.
Placenta ; 36(8): 903-10, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26145226

ABSTRACT

INTRODUCTION: Adequate maternal supply and placental delivery of long chain polyunsaturated fatty acids (LCPUFA) is essential for normal fetal development. In humans, maternal obesity alters placental FA uptake, though the impact of diet remains uncertain. The fatty fetal liver observed in offspring of Japanese macaques fed a high fat diet (HFD) was prevented with resveratrol supplementation during pregnancy. We sought to determine the effect of HFD and resveratrol, a supplement with insulin-sensitizing properties, on placental LCPUFA uptake in this model. METHODS: J. macaques were fed control chow (15% fat, n = 5), HFD (35% fat, n = 10) or HFD containing 0.37% resveratrol (n = 5) prior to- and throughout pregnancy. At ∼ 130 d gestation (term = 173 d), placentas were collected by caesarean section. Fatty acid uptake studies using (14)C-labeled oleic acid, arachidonic acid (AA) and docosahexanoic acid (DHA) were performed in placental explants. RESULTS: Resveratrol supplementation increased placental uptake of DHA (P < 0.05), while HFD alone had no measurable effect. Resveratrol increased AMP-activated protein kinase activity and mRNA expression of the fatty acid transporters FATP-4, CD36 and FABPpm (P < 0.05). Placental DHA content was decreased in HFD dams; resveratrol had no effect on tissue fatty acid profiles. DISCUSSION: Maternal HFD did not significantly affect placental LCPUFA uptake. Furthermore, resveratrol stimulated placental DHA uptake capacity, AMPK activation and transporter expression. Placental handling of DHA is particularly sensitive to the dramatic alterations in the maternal metabolic phenotype and placental AMPK activity associated with resveratrol supplementation.


Subject(s)
Diet, High-Fat , Fatty Acids/metabolism , Placenta/metabolism , Stilbenes/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Dietary Supplements , Female , Fetus/metabolism , Macaca , Maternal-Fetal Exchange/physiology , Phosphorylation , Placenta/drug effects , Pregnancy , Resveratrol
6.
Mol Metab ; 4(6): 437-60, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26042199

ABSTRACT

BACKGROUND: The gastrointestinal peptide hormone ghrelin was discovered in 1999 as the endogenous ligand of the growth hormone secretagogue receptor. Increasing evidence supports more complicated and nuanced roles for the hormone, which go beyond the regulation of systemic energy metabolism. SCOPE OF REVIEW: In this review, we discuss the diverse biological functions of ghrelin, the regulation of its secretion, and address questions that still remain 15 years after its discovery. MAJOR CONCLUSIONS: In recent years, ghrelin has been found to have a plethora of central and peripheral actions in distinct areas including learning and memory, gut motility and gastric acid secretion, sleep/wake rhythm, reward seeking behavior, taste sensation and glucose metabolism.

7.
Int J Obes (Lond) ; 38(2): 183-91, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23736354

ABSTRACT

OBJECTIVE: Fibroblast growth factor 21 (FGF21) is a metabolic regulator of glucose and lipid metabolism. The physiological role of FGF21 is not yet fully elucidated, however, administration of FGF21 lowers blood glucose in diabetic animals. Moreover, increased levels of FGF21 are found in obese and diabetic rodents and humans compared with lean/non-diabetic controls. METHODS: Adult male rhesus macaque monkeys were chronically maintained on a high-fat diet (HFD) or a standard diet (control, CTR). Plasma levels of FGF21, triglycerides and cholesterol were measured and body weight was record. Glucose-stimulated insulin secretion (GSIS) and glucose clearance was determined during an intravenous glucose tolerance test. Furthermore, expression of FGF21 and its receptors were determined in liver, pancreas, three white adipose tissues (WATs) and two skeletal muscles. RESULTS: A cohort of the high-fat fed monkeys responded to the HFD with increasing body weight, plasma lipids, total cholesterol, GSIS and decreased glucose tolerance. These monkeys were termed HFD sensitive. Another cohort of monkeys did not become obese and maintained normal insulin sensitivity. These animals were defined as HFD resistant. Plasma FGF21 levels were significantly increased in all HFD fed monkeys compared with the CTR group. The HFD-sensitive monkeys showed a significant increase in FGF21 mRNA expression in all examined tissues compared with CTR, whereas FGF21 expression in the HFD-resistant group was only increased in the liver, pancreas and the retroperitoneal WAT. In the WAT, the co-receptor ß-klotho was downregulated in the HFD-sensitive monkeys compared with the HFD-resistant group. CONCLUSION: This study demonstrates that HFD changes FGF21 and FGF21 receptor expression in a tissue-specific manner in rhesus monkeys; differential regulation is moreover observed between HFD-sensitive and -resistant monkeys. Monkeys that maintain normal levels of the FGF21 co-receptor ß-klotho in the WAT on HFD were protected toward development of dyslipidemia and hyperglycemia.


Subject(s)
Adipose Tissue, White/metabolism , Fibroblast Growth Factors/metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Animals , Blood Glucose/metabolism , Body Weight , Cholesterol/blood , Diet, High-Fat , Dyslipidemias/blood , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation , Glucose Tolerance Test , Hyperglycemia/blood , Insulin Resistance , Macaca mulatta , Male , Mice , Mice, Inbred NOD , Promoter Regions, Genetic , RNA, Messenger , Real-Time Polymerase Chain Reaction , Triglycerides/blood
8.
J Endocrinol ; 217(2): 207-13, 2013 May.
Article in English | MEDLINE | ID: mdl-23420316

ABSTRACT

Chronic high caloric intake has contributed to the increased prevalence of pediatric obesity and related morbidities. Most overweight or obese children, however, do not present with frank metabolic disease but rather insulin resistance or subclinical precursors. The innate immune system plays a role in the pathophysiology of type 2 diabetes but how it contributes to early metabolic dysfunction in children on chronic high-fat diet (HFD) is unclear. We hypothesize that such inflammation is present in the pancreas of children and is associated with early insulin resistance. We used nonhuman primate (NHP) juveniles exposed to chronic HFD as a model of early pediatric metabolic disease to demonstrate increased pancreatic inflammatory markers before the onset of significant obesity or glucose dysregulation. Pancreata from 13-month-old Japanese macaques exposed to a HFD from in utero to necropsy were analyzed for expression of cytokines and islet-associated macrophages. Parameters from an intravenous glucose tolerance test were correlated with cytokine expression. Before significant glucose dysregulation, the HFD cohort had a twofold increase in interleukin 6 (IL6), associated with decreased first-phase insulin response and a sexually dimorphic (male) increase in IL1ß correlating with increased fasting glucose levels. The number of islet-associated macrophages was also increased. Pancreata from juvenile NHP exposed to HFD have increased inflammatory markers and evidence of innate immune infiltration before the onset of significant obesity or glucose dysregulation. Given the parallel development of metabolic disease between humans and NHPs, these findings have strong relevance to the early metabolic disease driven by a chronic HFD in children.


Subject(s)
Insulin Resistance/physiology , Islets of Langerhans/pathology , Macrophages/pathology , Pancreatitis/pathology , Pancreatitis/physiopathology , Animals , Blood Glucose/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Interleukin-1beta/blood , Interleukin-6/blood , Macaca , Male , Pancreatitis/etiology , Sex Factors
9.
Int J Obes (Lond) ; 37(2): 254-62, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22450853

ABSTRACT

OBJECTIVE: The link between maternal under-nutrition and cardiovascular disease (CVD) in the offspring later in life is well recognized, but the impact of maternal over-nutrition on the offspring's cardiovascular function and subsequent risk for CVD later in life remains unclear. Here, we investigated the impact of maternal exposure to a high-fat/calorie diet (HFD) during pregnancy and early postnatal period on endothelial function of the offspring in a nonhuman primate model. METHODS: Offspring, naturally born to either a control (CTR) diet (14% fat calories) or a HFD (36% fat calories) consumption dam, were breast-fed until weaning at about 8 months of age. After weaning, the offspring were either maintained on the same diet (CTR/CTR, HFD/HFD), or underwent a diet switch (CTR/HFD, HFD/CTR). Blood samples and arterial tissues were collected at necropsy when the animals were about 13 months of age. RESULTS: HFD/HFD juveniles displayed an increased plasma insulin level and glucose-stimulated insulin secretion in comparison with CTR/CTR. In abdominal aorta, but not the renal artery, acetylcholine-induced vasorelaxation was decreased remarkably for HFD/HFD juveniles compared with CTR/CTR. HFD/HFD animals also showed a thicker intima wall and an abnormal vascular-morphology, concurrent with elevated expression levels of several markers related to vascular inflammation and fibrinolytic function. Diet-switching animals (HFD/CTR and CTR/HFD) displayed modest damage on the abdominal vessel. CONCLUSION: Our data indicate that maternal HFD exposure impairs offspring's endothelial function. Both early programming events and postweaning diet contribute to the abnormalities that could be reversed partially by diet intervention.


Subject(s)
Diet, High-Fat/adverse effects , Fetal Growth Retardation/metabolism , Liver/metabolism , Obesity/blood , Overnutrition/blood , Prenatal Exposure Delayed Effects/blood , Animals , Animals, Newborn , Carotid Intima-Media Thickness , Disease Models, Animal , Endothelium, Vascular/pathology , Fasting/blood , Female , Fetal Growth Retardation/physiopathology , Gene Expression Regulation, Developmental , Macaca , Male , Maternal Nutritional Physiological Phenomena , Maternal-Fetal Exchange , Obesity/complications , Obesity/pathology , Overnutrition/complications , Placental Insufficiency/pathology , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Primates , Real-Time Polymerase Chain Reaction , Weaning
10.
Int J Obes (Lond) ; 37(8): 1064-70, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23207405

ABSTRACT

BACKGROUND: The intake of a Western diet enriched in animal fat has been shown to be a major risk factor for Type 2 diabetes and obesity. Previous rodent studies have indicated that these conditions may be triggered by the accumulation of the sphingolipid ceramide in insulin-sensitive tissues. However, data are lacking in this regard from both humans and non-human primates. OBJECTIVE: Here we have investigated the relationship between plasma ceramides and metabolic syndrome in Rhesus macaques fed a high-fat and high-fructose (HFFD) 'western' diet. METHODS: We investigated this relationship in cohorts of monkeys fed a HFFD for a period of 8 months to 5 years. Animals were classified as control, pre-diabetic or diabetic based on fasting plasma parameters and insulin sensitivity. RESULTS: HFFD treatment produced significant increases in body weight and body fat and also resulted in a decline in insulin sensitivity. In parallel to the reduction in insulin sensitivity, significant increases in both plasma ceramide and dihydroceramide levels were observed, which further increased as animals progressed to the diabetic state. Plasma levels of the rare sphingolipid C18:0 deoxysphinganine, a marker of increased metabolic flux through serine palmitoyl transferase (SPT), were also elevated in both pre- and diabetic animals. Furthermore, plasma serine levels were significantly elevated in diabetic monkeys, which may indicate a shift in SPT substrate selectivity from serine to alanine or glycine. In contrast, branch chain amino acids were unchanged in pre-diabetic non-human primates, and only plasma valine levels were elevated in diabetic animals. CONCLUSION: Together, these data indicate that HFFD induces de novo synthesis of ceramides in non-human primates, and that increased production of plasma ceramides is significantly correlated with the decline in insulin sensitivity.


Subject(s)
Diabetes Mellitus, Type 2/blood , Diet, High-Fat/adverse effects , Metabolic Syndrome/blood , Obesity/blood , Serine C-Palmitoyltransferase/blood , Sphingolipids/blood , Animals , Biomarkers/blood , Ceramides/blood , Diabetes Mellitus, Type 2/etiology , Disease Models, Animal , Disease Progression , Fructose/adverse effects , Insulin Resistance , Macaca mulatta , Male , Metabolic Syndrome/etiology , Obesity/complications , Risk Factors , Valine/blood
11.
J Neuroendocrinol ; 23(11): 1099-112, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21518032

ABSTRACT

Low levels of the adipocyte hormone leptin are considered to be the key signal contributing to inhibited gonadotrophin-releasing hormone (GnRH) release and reproductive acyclicity during negative energy balance. Hypoleptinaemia-induced inhibition of GnRH may be initiated with upstream inhibition of the secretagogue kisspeptin (Kiss1) because GnRH neurones do not express leptin receptors. The present study aimed to determine whether eliminating the hypoleptinaemia associated with caloric restriction (CR), by restoring leptin to normal basal levels, could reverse the suppression of the reproductive neuroendocrine axis. Fifty percent CR resulted in significant suppression of anteroventral periventricular Kiss1 mRNA, arcuate nucleus (ARH) Kiss1 and neurokinin B (NKB) mRNA levels and serum luteinising hormone (LH). Restoring leptin to normal basal levels did not restore Kiss1 or NKB mRNA or LH levels. Surprisingly, leptin did not activate expression of phosphorylated signal-transducer and activator of transcription-3 in ARC Kiss1 neurones, indicating that these neurones may not relay leptin signalling to GnRH neurones. Previous work in fasting models showing restoration of LH used a pharmacological dose of leptin. Therefore, in a 48-h fast study, replacement of leptin to pharmacological levels was compared with replacement of leptin to normal basal levels. Maintaining leptin at normal basal levels during the fast did not prevent inhibition of LH. By contrast, pharmacological levels of leptin did maintain LH at control values. These results suggest that, although leptin may be a permissive signal for reproductive function, hypoleptinaemia is unlikely to be the critical signal responsible for ARC Kiss1 and LH inhibition during negative energy balance.


Subject(s)
Energy Metabolism , Kisspeptins/metabolism , Leptin/physiology , Luteinizing Hormone/metabolism , Animals , Caloric Restriction , Female , Hypothalamus/metabolism , Immunohistochemistry , Kisspeptins/genetics , Leptin/pharmacology , Luteinizing Hormone/blood , RNA, Messenger/genetics , Radioimmunoassay , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
12.
Brain Res ; 1364: 139-52, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20727862

ABSTRACT

Lactation is an important physiological model of the integration of energy balance and reproduction, as it involves activation of potent appetitive neuropeptide systems coupled to a profound inhibition of pulsatile GnRH/LH secretion. There are multiple systems that contribute to the chronic hyperphagia of lactation: 1) suppression of the metabolic hormones, leptin and insulin, 2) activation of hypothalamic orexigenic neuropeptide systems NPY, AGRP, orexin (OX) and melanin concentrating hormone (MCH), 3) special induction of NPY expression in the dorsomedial hypothalamus, and 4) suppression of anorexigenic systems POMC and CART. These changes ensure adequate energy intake to meet the metabolic needs of milk production. There is significant overlap in all of the systems that regulate food intake with the regulation of GnRH, suggesting there could be several redundant factors acting to suppress GnRH/LH during lactation. In addition to an overall increase in inhibitory tone acting directly on GnRH cell bodies that is brought about by increases in orexigenic systems, there are also effects at the ARH to disrupt Kiss1/neurokinin B/dynorphin neuronal function through inhibition of Kiss1 and NKB. These changes could lead to an increase in inhibitory auto-regulation of the Kiss1 neurons and a possible disruption of pulsatile GnRH release. While the low levels of leptin and insulin contribute to the changes in ARH appetitive systems, they do not appear to contribute to the suppression of ARH Kiss1 or NKB. The inhibition of Kiss1 may be the key factor in the suppression of GnRH during lactation, although the mechanisms responsible for its inhibition are unknown.


Subject(s)
Gonadotropin-Releasing Hormone/physiology , Lactation/physiology , Leptin/physiology , Neurosecretory Systems/physiology , Tumor Suppressor Proteins/physiology , Animals , Appetitive Behavior/physiology , Energy Metabolism/physiology , Female , Humans , Hyperphagia , Hypothalamus/physiology , Insulin/metabolism , Insulin/physiology , Kisspeptins , Neurons/physiology , Neuropeptides/physiology , Reproduction/physiology
13.
Endocrinology ; 151(4): 1622-32, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20176722

ABSTRACT

The hypothalamic melanocortin system, which controls appetite and energy expenditure, develops during the third trimester in primates. Thus, maternal nutrition and health may have a profound influence on the development of this system. To study the effects of chronic maternal high-fat diet (HFD) on the development of the melanocortin system in the fetal nonhuman primate, we placed adult female macaques on either a control (CTR) diet or a HFD for up to 4 yr. A subgroup of adult female HFD animals was also switched to CTR diet during the fifth year of the study (diet reversal). Third-trimester fetuses from mothers on HFD showed increases in proopiomelanocortin mRNA expression, whereas agouti-related protein mRNA and peptide levels were decreased in comparison with CTR fetuses. Proinflammatory cytokines, including IL-1beta and IL-1 type 1 receptor, and markers of activated microglia were elevated in the hypothalamus, suggesting an activation of the local inflammatory response. Fetuses of diet-reversal mothers had normal melanocortin levels. These results raise the concern that chronic consumption of a HFD during pregnancy, independent of maternal obesity and diabetes, can lead to widespread activation of proinflammatory cytokines that may alter the development of the melanocortin system. The abnormalities in the fetal POMC system, if maintained into the postnatal period, could impact several systems, including body weight homeostasis, stress responses, and cardiovascular function. Indeed, the HFD offspring develop early-onset excess weight gain. These abnormalities may be prevented by healthful nutrient consumption during pregnancy even in obese and severely insulin-resistant individuals.


Subject(s)
Dietary Fats/metabolism , Hypothalamus/metabolism , Inflammation/metabolism , Melanocortins/metabolism , Prenatal Nutritional Physiological Phenomena/physiology , Adrenocorticotropic Hormone/metabolism , Animal Nutritional Physiological Phenomena , Animals , Female , Fetus/metabolism , Immunohistochemistry , In Situ Hybridization , Interleukin-1beta/metabolism , Macaca , Melanocortins/genetics , Microglia/metabolism , Microscopy, Confocal , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin-1 Type I/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
Int J Obes (Lond) ; 34(6): 989-1000, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20157318

ABSTRACT

OBJECTIVE: A sexual dimorphism exists in body fat distribution; females deposit relatively more fat in subcutaneous/inguinal depots whereas males deposit more fat in the intra-abdominal/gonadal depot. Our objective was to systematically document depot- and sex-related differences in the accumulation of adipose tissue and gene expression, comparing differentially expressed genes in diet-induced obese mice with mice maintained on a chow diet. RESEARCH DESIGN AND METHODS: We used a microarray approach to determine whether there are sexual dimorphisms in gene expression in age-matched male, female or ovariectomized female (OVX) C57/BL6 mice maintained on a high-fat (HF) diet. We then compared expression of validated genes between the sexes on a chow diet. RESULTS: After exposure to a high fat diet for 12 weeks, females gained less weight than males. The microarray analyses indicate in intra-abdominal/gonadal adipose tissue in females 1642 genes differ by at least twofold between the depots, whereas 706 genes differ in subcutaneous/inguinal adipose tissue when compared with males. Only 138 genes are commonly regulated in both sexes and adipose tissue depots. Inflammatory genes (cytokine-cytokine receptor interactions and acute-phase protein synthesis) are upregulated in males when compared with females, and there is a partial reversal after OVX, where OVX adipose tissue gene expression is more 'male-like'. This pattern is not observed in mice maintained on chow. Histology of male gonadal white adipose tissue (GWAT) shows more crown-like structures than females, indicative of inflammation and adipose tissue remodeling. In addition, genes related to insulin signaling and lipid synthesis are higher in females than males, regardless of dietary exposure. CONCLUSIONS: These data suggest that male and female adipose tissue differ between the sexes regardless of diet. Moreover, HF diet exposure elicits a much greater inflammatory response in males when compared with females. This data set underscores the importance of analyzing depot-, sex- and steroid-dependent regulation of adipose tissue distribution and function.


Subject(s)
Adipose Tissue/physiology , Adiposity/genetics , Obesity/genetics , Sex Characteristics , Adipose Tissue/metabolism , Animals , Body Fat Distribution , Dietary Fats/administration & dosage , Dietary Fats/metabolism , Female , Gene Expression , Male , Mice , Mice, Obese , Obesity/metabolism , Obesity/physiopathology , Ovariectomy , Tissue Array Analysis/methods , Weight Gain/genetics
15.
Forum Nutr ; 63: 186-194, 2010.
Article in English | MEDLINE | ID: mdl-19955786

ABSTRACT

Increasing evidence indicates that early metabolic programming contributes to escalating obesity rates in children and adults. Metabolic imprinting is involved in the establishment of set points for physiologic and metabolic responses in adulthood. Evidence from epidemiological studies and animal models indicates that maternal health and nutritional status during gestation and lactation have long-term effects on central and peripheral systems that regulate energy balance in the developing offspring. Perinatal nutrition also impacts susceptibility to developing metabolic disorders and plays a role in programming body weight set points. The states of maternal energy status and health that are implicated in predisposing offspring to increased risk of developing obesity include maternal overnutrition, diabetes, and undernutrition. This chapter discusses the evidence from epidemiologic studies and animal models that each of these states of maternal energy status results in metabolic imprinting of obesity in offspring. Also, the potential molecular mediators of metabolic imprinting of obesity by maternal energy status including glucose, insulin, leptin, inflammatory cytokines and epigenetic mechanisms are considered.


Subject(s)
Energy Metabolism/physiology , Nutritional Status/physiology , Obesity/embryology , Obesity/metabolism , Prenatal Nutritional Physiological Phenomena/physiology , Animals , Disease Models, Animal , Epigenesis, Genetic , Female , Genetic Predisposition to Disease , Humans , Male , Obesity/genetics , Pregnancy , Prenatal Exposure Delayed Effects
16.
Front Neuroendocrinol ; 31(1): 16-31, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19822169

ABSTRACT

Over the last decade there has been a striking increase in the early onset of metabolic disease, including obesity and diabetes. The regulation of energy homeostasis is complex and involves the intricate integration of peripheral and central systems, including the hypothalamus. This review provides an overview of the development of brain circuitry involved in the regulation of energy homeostasis as well as recent findings related to the impact of both prenatal and postnatal maternal environment on the development of these circuits. There is surprising evidence that both overnutrition and undernutrition impact the development of these circuits in a similar manner as well as having similar consequences of increased obesity and diabetes later in life. There is also a special focus on relevant species differences in the development of hypothalamic circuits. A deeper understanding of the mechanisms involved in the development of brain circuitry is needed to fully understand how the nutritional and/or maternal environments impact the functional circuitry as well as the behavior and physiological outcomes.


Subject(s)
Hypothalamus/growth & development , Neuropeptides/physiology , Nutritional Physiological Phenomena , Adolescent , Adult , Animals , Brain/physiology , Child , Child, Preschool , Energy Metabolism/physiology , Female , Fetal Development , Homeostasis/physiology , Humans , Hypothalamus/physiology , Maternal Nutritional Physiological Phenomena , Metabolic Syndrome/etiology , Obesity/epidemiology , Obesity/etiology , Obesity/genetics , Pregnancy , Prenatal Exposure Delayed Effects
17.
Aquat Toxicol ; 86(1): 91-8, 2008 Jan 20.
Article in English | MEDLINE | ID: mdl-18023888

ABSTRACT

The synthetic organic compound 4-nonylphenol (NP) has been detected in many human-impacted surface waters in North America. In this study, we examined the ability of NP to alter reproductive competence in male fathead minnows after a 28 day flow-through exposure in a range of environmentally relevant concentrations bracketing the U.S. Environmental Protection Agency toxicity-based NP chronic exposure criterion of 6.1 microg NP/L. Exposure to NP at and above the EPA chronic exposure criterion resulted in an induction of plasma vitellogenin (VTG) within 14 days. However, 7 days after the cessation of exposure, VTG concentrations had dropped more than 50% and few males expressed VTG above the detection threshold. All of the morphological endpoints, including gonadosomatic index, hepatosomatic index, secondary sexual characters, and histopathology, were unaltered by all NP treatments. However, when NP-exposed male fish were allowed to compete with control males for access to nest sites and females, most treatments altered the reproductive competence of exposed males. At lower NP concentrations, exposed males out-competed control males, possibly by being primed through the estrogenic NP exposure in a fashion similar to priming by pheromones released from female fathead minnows. At higher NP exposure concentrations, this priming effect was negated by the adverse effects of the exposure and control males out-competed treated males. Results of this study indicate the complexity of endocrine disrupting effects and the need for multiple analysis levels to assess the effects of these compounds on aquatic organisms.


Subject(s)
Cyprinidae , Environmental Exposure , Phenols/toxicity , Reproduction/drug effects , Water Pollutants, Chemical/toxicity , Animals , Female , Male , Phenols/analysis , Random Allocation , Sexual Behavior, Animal/drug effects , Time Factors , Vitellogenins/blood
18.
Neuroscience ; 143(4): 975-86, 2006 Dec 28.
Article in English | MEDLINE | ID: mdl-17029798

ABSTRACT

In the rodent, arcuate nucleus of the hypothalamus (ARH)-derived neuropeptide Y (NPY) and proopiomelanocortin (POMC) neurons have efferent projections throughout the hypothalamus that do not fully mature until the second and third postnatal weeks. Since this process is likely completed by birth in primates we characterized the ontogeny of NPY and melanocortin systems in the fetal Japanese macaque during the late second (G100), early third (G130) and late third trimesters (G170). NPY mRNA was expressed in the ARH, paraventricular nucleus (PVH), and dorsomedial nucleus of the hypothalamus (DMH) as early as G100. ARH-derived NPY projections to the PVH were initiated at G100 but were limited and variable; however, there was a modest increase in density and number by G130. ARH-NPY/agouti-related peptide (AgRP) fiber projections to efferent target sites were completely developed by G170, but the density continued to increase in the postnatal period. In contrast to NPY/AgRP projections, alphaMSH fibers were minimal at G100 and G130 but were moderate at G170. This study also revealed several significant species differences between rodent and the nonhuman primate (NHP). There were few NPY/catecholamine projections to the PVH and ARH prior to birth, while projections were increased in the adult. A substantial proportion of the catecholamine fibers did not coexpress NPY. In addition, cocaine and amphetamine-related transcript (CART) and alpha-melanocyte stimulating hormone (alphaMSH) were not colocalized in fibers or cell bodies. As a consequence of the prenatal development of these neuropeptide systems in the NHP, the maternal environment may critically influence these circuits. Additionally, because differences exist in the neuroanatomy of NPY and melanocortin circuitry the regulation of these systems may be different in primates than in rodents.


Subject(s)
Hypothalamus/embryology , Hypothalamus/metabolism , Macaca/embryology , Macaca/metabolism , Neuropeptides/metabolism , Agouti-Related Protein , Animals , Arcuate Nucleus of Hypothalamus/embryology , Arcuate Nucleus of Hypothalamus/metabolism , Catecholamines/metabolism , Dorsomedial Hypothalamic Nucleus/embryology , Dorsomedial Hypothalamic Nucleus/metabolism , Female , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Pathways/embryology , Neural Pathways/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Neuropeptides/genetics , Paraventricular Hypothalamic Nucleus/embryology , Paraventricular Hypothalamic Nucleus/metabolism , Pregnancy , RNA, Messenger/metabolism , Rodentia/embryology , Rodentia/metabolism , Species Specificity , alpha-MSH/metabolism
19.
Ann Oncol ; 17(2): 189-99, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16236756

ABSTRACT

In the absence of metastatic disease patients with localized or locally advanced pancreatic cancer can benefit from surgical resection or chemoradiation. Despite the advances of imaging technology, however, noninvasive staging modalities are still inaccurate in identifying small volume metastatic disease leading potentially to inappropriate treatment and avoidable morbidity in a subgroup of patients. Staging laparoscopy may identify those patients with unsuspected metastatic disease on preoperative imaging and prevent unnecessary laparotomy or chemoradiation. A controversy exists, however, as to whether the procedure should be used routinely or selectively in pancreatic cancer patients with no evidence of metastasis on noninvasive staging. This review aims to assess the current role of staging laparoscopy by examining its diagnostic accuracy and ability to prevent unnecessary treatment as well as its morbidity, oncologic effect and cost-effectiveness. The available literature will be evaluated critically, its limitations identified and exisiting controversies addressed.


Subject(s)
Adenocarcinoma/pathology , Laparoscopy , Pancreatic Neoplasms/pathology , Adenocarcinoma/economics , Cost-Benefit Analysis , Cytodiagnosis , Humans , Neoplasm Metastasis , Neoplasm Staging , Pancreatic Neoplasms/economics , Patient Selection , Peritoneal Lavage
20.
Neuroscience ; 116(2): 393-406, 2003.
Article in English | MEDLINE | ID: mdl-12559095

ABSTRACT

In the adult rat, arcuate-neuropeptide Y/agouti-related protein neurons have efferent projections throughout the hypothalamus and provide a potent orexigenic stimulus. At birth neuropeptide Y fibers are also present throughout the hypothalamus; however, the source of these fibers has been unknown. The present studies determined the postnatal ontogeny of arcuate-neuropeptide Y fibers into the paraventricular nucleus and dorsomedial hypothalamic nucleus, as well as the ontogeny of neuropeptide Y1 receptor expression within these areas. Agouti-related protein messenger RNA and protein expression was present exclusively in cell bodies in the arcuate throughout postnatal development, starting at P2, and was colocalized in the vast majority of arcuate-neuropeptide Y neurons. This exclusive colocalization of agouti-related protein with arcuate-neuropeptide Y neurons makes it an excellent marker for these neurons and their projections. Even though single-label neuropeptide Y fibers were abundant in the dorsomedial hypothalamic nucleus and paraventricular nucleus as early as P2, arcuate-neuropeptide Y/agouti-related protein fibers did not significantly innervate these areas until P5-6 and P10-11, respectively. In contrast, a portion of the neuropeptide Y fibers within the paraventricular nucleus as early as P2 originated from the brainstem, as indicated by their colocalization with dopamine beta hydroxylase. It remains to be determined if local sources of neuropeptide Y-expressing cells within the dorsomedial hypothalamic nucleus and paraventricular nucleus also contribute to the neuropeptide Y-immunoreactive fibers within these regions prior to the development of arcuate-neuropeptide Y/agouti-related protein projections. In addition to the dramatic change in arcuate-neuropeptide Y/agouti-related protein projections, there is also a striking change in Y1 protein expression in the hypothalamus during the first two postnatal weeks. Taken together these data suggest that the early postnatal period, during which there is a dynamic change in the hypothalamic neuropeptide Y system, may constitute a critical period in the development of this important feeding circuit.


Subject(s)
Arcuate Nucleus of Hypothalamus/growth & development , Arcuate Nucleus of Hypothalamus/physiology , Gene Expression Regulation, Developmental , Neuropeptide Y/genetics , Age Factors , Agouti-Related Protein , Animals , Arcuate Nucleus of Hypothalamus/cytology , Biomarkers , Brain Stem/cytology , Brain Stem/growth & development , Efferent Pathways , Female , Intercellular Signaling Peptides and Proteins , Nerve Fibers/chemistry , Nerve Fibers/physiology , Neuropeptide Y/analysis , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/growth & development , Pregnancy , Proteins/analysis , Proteins/genetics , RNA, Messenger/analysis , Rats , Receptors, Neuropeptide Y/analysis , Receptors, Neuropeptide Y/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...