1501
|
Feinle-Bisset C, Vozzo R, Horowitz M, Talley NJ. Diet, food intake, and disturbed physiology in the pathogenesis of symptoms in functional dyspepsia. Am J Gastroenterol 2004; 99:170-81. [PMID: 14687160 DOI: 10.1111/j.1572-0241.2004.04003.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Functional dyspepsia (FD) remains a relatively poorly characterized gastrointestinal disorder of unknown etiology that is frequently difficult to manage. A systematic review of the literature relating to food intake and FD is summarized here. Many patients with FD report symptoms after meal ingestion, including fullness, bloating, epigastric pain, nausea, and vomiting, and this has been interpreted as indicative of an underlying "motor disorder of the stomach or small intestine." Such hypotheses are, however, still largely unsubstantiated, and the data that do exist are inconclusive, particularly as few studies have directly examined the temporal relationships between dyspeptic symptoms, meal ingestion, and disordered gastric motility. Moreover, studies attempting to relate symptoms to specific disturbances in gastric motor function have, in most cases, not evaluated symptoms concurrently with the function test, and/or have used suboptimal symptom scoring to quantify symptoms. Furthermore, the term "early satiety" has been used loosely as a symptom, rather than a quantitative measure of food intake. Currently, the most widely accepted mechanism underlying FD is visceral hypersensitivity, which may contribute to both enhanced motor and symptomatic responses to food ingestion. However, the possible contribution of food and dietary habits to the induction and/or exacerbation of dyspeptic symptoms represents a relatively new area-despite frequent reports by patients that their symptoms are often related to food ingestion; this association has not been formally assessed. Dietary assessments have frequently implicated fatty foods in symptom induction, and these findings are supported by laboratory-based studies, particularly the demonstration that FD patients more often experience symptoms after intraduodenal infusions of fat, than glucose. Further studies into the potential role of dietary factors in the induction of dyspeptic symptom are required to establish whether dietary therapies have any place in the management of FD.
Collapse
Affiliation(s)
- Christine Feinle-Bisset
- Department of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | | | | | | |
Collapse
|
1502
|
Riediger T, Bothe C, Becskei C, Lutz TA. Peptide YY directly inhibits ghrelin-activated neurons of the arcuate nucleus and reverses fasting-induced c-Fos expression. Neuroendocrinology 2004; 79:317-26. [PMID: 15256809 DOI: 10.1159/000079842] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2004] [Accepted: 05/24/2004] [Indexed: 01/27/2023]
Abstract
The hypothalamic arcuate nucleus (Arc) monitors and integrates hormonal and metabolic signals involved in the maintenance of energy homeostasis. The orexigenic peptide ghrelin is secreted from the stomach during negative status of energy intake and directly activates neurons of the medial arcuate nucleus (ArcM) in rats. In contrast to ghrelin, peptide YY (PYY) is released postprandially from the gut and reduces food intake when applied peripherally. Neurons in the ArcM express ghrelin receptors and neuropeptide Y receptors. Thus, PYY may inhibit feeding by acting on ghrelin-sensitive Arc neurons. Using extracellular recordings, we (1) characterized the effects of PYY on the electrical activity of ghrelin-sensitive neurons in the ArcM of rats. In order to correlate the effect of PYY on neuronal activity with the energy status, we (2) investigated the ability of PYY to reverse fasting-induced c-Fos expression in Arc neurons of mice. In addition, we (3) sought to confirm that PYY reduces food intake under our experimental conditions. Superfusion of PYY reversibly inhibited 94% of all ArcM neurons by a direct postsynaptic mechanism. The PYY-induced inhibition was dose-dependent and occurred at a threshold concentration of 10(-8)M. Consistent with the opposite effects of ghrelin and PYY on food intake, a high percentage (50%) of Arc neurons was activated by ghrelin and inhibited by PYY. In line with this inhibitory action, peripherally injected PYY partly reversed the fasting-induced c-Fos expression in Arc neurons of mice. Similarly, refeeding of food-deprived mice reversed the fasting-induced activation in the Arc. Furthermore, peripherally injected PYY reduced food intake in 12-hour fasted mice. Thus the activity of Arc neurons correlated with the feeding status and was not only reduced by feeding but also by administration of PYY in non-refed mice. In conclusion, our current observations suggest that PYY may contribute to signaling a positive status of energy intake by inhibiting Arc neurons, which are activated under a negative status of energy intake by signals such as ghrelin.
Collapse
Affiliation(s)
- Thomas Riediger
- Institute of Veterinary Physiology, University of Zurich, CH-8057 Zurich, Switzerland.
| | | | | | | |
Collapse
|
1503
|
Abstract
Prader-Willi syndrome (PWS) is a complex human genetic disease that arises from lack of expression of paternally inherited imprinted genes on chromosome 15q11-q13. Identification of the imprinting control centre, novel imprinted genes and distinct phenotypes in PWS patients and mouse models has increased interest in this human obesity syndrome. In this review I focus on: (i) the chromosomal region and candidate genes associated with PWS, and the possible links with individual PWS phenotypes identified using mouse models; (ii) the metabolic and hormonal phenotypes in PWS; (iii) postmortem studies of human PWS hypothalami; and (iv) current and potential advances in the management of PWS and its complications. This could have benefits for a wide spectrum of endocrine, paediatric and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Anthony P Goldstone
- Department of Endocrinology, St Bartholomew's Hospital, West Smithfield, London EC1A 7BE, UK.
| |
Collapse
|
1504
|
Woods SC. Gastrointestinal satiety signals I. An overview of gastrointestinal signals that influence food intake. Am J Physiol Gastrointest Liver Physiol 2004; 286:G7-13. [PMID: 14665437 DOI: 10.1152/ajpgi.00448.2003] [Citation(s) in RCA: 222] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
An overview is presented of those signals generated by the gastrointestinal (GI) tract during meals that interact with the central nervous system to create a sensation of fullness and satiety. Although dozens of enzymes, hormones, and other factors are secreted by the GI tract in response to food in the lumen, only a handful are able to influence food intake directly. Most of these cause meals to terminate and hence are called satiety signals, with CCK being the most investigated. Only one GI signal, ghrelin, that increases meal size has been identified. The administration of exogenous CCK or other satiety signals causes smaller meals to be consumed, whereas blocking the action of endogenous CCK or other satiety signals causes larger meals to be consumed. Satiety signals are relayed to the hindbrain, either indirectly via nerves such as the vagus from the GI tract or else directly via the blood. Most factors that influence how much food is eaten during individual meals act by changing the sensitivity to satiety signals. This includes adiposity signals as well as habits and learning, the social situation, and stressors.
Collapse
Affiliation(s)
- Stephen C Woods
- Dept. of Psychiatry, Univ. of Cincinnati, Cincinnati, OH 45267, USA.
| |
Collapse
|
1505
|
Abstract
Growing health problems related to obesity have focused considerable attention on a number of neurotransmitters, particularly hypothalamic neuropeptides, involved in regulating energy homeostasis and food intake. As the fast-acting transmitters GABA and glutamate underlie the majority of fast synaptic activity in the hypothalamus, understanding neuropeptide modulation of amino acid transmitter actions may be key to a full appreciation of how the brain controls caloric balances.
Collapse
Affiliation(s)
- A N van den Pol
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| |
Collapse
|
1506
|
Boutin P, Dina C, Vasseur F, Dubois S, Corset L, Séron K, Bekris L, Cabellon J, Neve B, Vasseur-Delannoy V, Chikri M, Charles MA, Clement K, Lernmark A, Froguel P. GAD2 on chromosome 10p12 is a candidate gene for human obesity. PLoS Biol 2003; 1:E68. [PMID: 14691540 PMCID: PMC270019 DOI: 10.1371/journal.pbio.0000068] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2003] [Accepted: 10/09/2003] [Indexed: 11/19/2022] Open
Abstract
The gene GAD2 encoding the glutamic acid decarboxylase enzyme (GAD65) is a positional candidate gene for obesity on Chromosome 10p11-12, a susceptibility locus for morbid obesity in four independent ethnic populations. GAD65 catalyzes the formation of gamma-aminobutyric acid (GABA), which interacts with neuropeptide Y in the paraventricular nucleus to contribute to stimulate food intake. A case-control study (575 morbidly obese and 646 control subjects) analyzing GAD2 variants identified both a protective haplotype, including the most frequent alleles of single nucleotide polymorphisms (SNPs) +61450 C>A and +83897 T>A (OR = 0.81, 95% CI [0.681-0.972], p = 0.0049) and an at-risk SNP (-243 A>G) for morbid obesity (OR = 1.3, 95% CI [1.053-1.585], p = 0.014). Furthermore, familial-based analyses confirmed the association with the obesity of SNP +61450 C>A and +83897 T>A haplotype (chi(2) = 7.637, p = 0.02). In the murine insulinoma cell line betaTC3, the G at-risk allele of SNP -243 A>G increased six times GAD2 promoter activity (p < 0.0001) and induced a 6-fold higher affinity for nuclear extracts. The -243 A>G SNP was associated with higher hunger scores (p = 0.007) and disinhibition scores (p = 0.028), as assessed by the Stunkard Three-Factor Eating Questionnaire. As GAD2 is highly expressed in pancreatic beta cells, we analyzed GAD65 antibody level as a marker of beta-cell activity and of insulin secretion. In the control group, -243 A>G, +61450 C>A, and +83897 T>A SNPs were associated with lower GAD65 autoantibody levels (p values of 0.003, 0.047, and 0.006, respectively). SNP +83897 T>A was associated with lower fasting insulin and insulin secretion, as assessed by the HOMA-B% homeostasis model of beta-cell function (p = 0.009 and 0.01, respectively). These data support the hypothesis of the orexigenic effect of GABA in humans and of a contribution of genes involved in GABA metabolism in the modulation of food intake and in the development of morbid obesity.
Collapse
Affiliation(s)
- Philippe Boutin
- 1Institute of Biology–Centre National de la Recherche Scientifique, Pasteur InstituteLilleFrance
| | - Christian Dina
- 1Institute of Biology–Centre National de la Recherche Scientifique, Pasteur InstituteLilleFrance
| | - Francis Vasseur
- 1Institute of Biology–Centre National de la Recherche Scientifique, Pasteur InstituteLilleFrance
- 2University Hospital of LilleLilleFrance
| | - Séverine Dubois
- 1Institute of Biology–Centre National de la Recherche Scientifique, Pasteur InstituteLilleFrance
| | - Laetitia Corset
- 1Institute of Biology–Centre National de la Recherche Scientifique, Pasteur InstituteLilleFrance
| | - Karin Séron
- 1Institute of Biology–Centre National de la Recherche Scientifique, Pasteur InstituteLilleFrance
| | - Lynn Bekris
- 3Department of Medicine, University of WashingtonSeattle, WashingtonUnited States of America
| | - Janice Cabellon
- 3Department of Medicine, University of WashingtonSeattle, WashingtonUnited States of America
| | - Bernadette Neve
- 1Institute of Biology–Centre National de la Recherche Scientifique, Pasteur InstituteLilleFrance
| | - Valérie Vasseur-Delannoy
- 1Institute of Biology–Centre National de la Recherche Scientifique, Pasteur InstituteLilleFrance
| | - Mohamed Chikri
- 1Institute of Biology–Centre National de la Recherche Scientifique, Pasteur InstituteLilleFrance
| | - M. Aline Charles
- 4Institut National de la Santé et de la Recherche Médicale (INSERM), Paul Brousse HospitalVillejuifFrance
| | - Karine Clement
- 5Paris VI University and INSERM “Avenir,” Department of Nutrition, Hôtel Dieu HospitalParisFrance
| | - Ake Lernmark
- 3Department of Medicine, University of WashingtonSeattle, WashingtonUnited States of America
| | - Philippe Froguel
- 1Institute of Biology–Centre National de la Recherche Scientifique, Pasteur InstituteLilleFrance
- 6Hammersmith Genome Centre and Department of Genomic Medicine, Imperial CollegeLondonUnited Kingdom
| |
Collapse
|
1507
|
Abstract
PURPOSE OF REVIEW Food intake is critical for survival and is a complex behavior with multiple levels of control. Short-term, meal-related signals arise from many sources including the gastrointestinal tract, the environment, and higher centers in the brain. As described in this review, inputs from the gastrointestinal tract can exert potent effects on meal initiation, meal termination, and meal frequency. The complex array of signals generated from the gastrointestinal system and from adipose tissue, which participate in the regulation of food intake, and specifically how these signals relate to satiety and hunger, is the focus of this review. RECENT FINDINGS Literature on the role of the well-studied gastrointestinal peptide, cholecystokinin, in satiety, in addition to its interaction with long-term adiposity signals in mediating food intake will be reviewed. In addition, literature on the gastrointestinal hormones glucagon-like-peptide 1, apolipoprotein A-IV and peptide YY, and how they may act to regulate satiety, is described. Finally, the newly discovered hormone, ghrelin, and how it relates to meal initiation and hunger is discussed. SUMMARY A better understanding of these systems and how they relate to body adiposity will prove to have important clinical applications. The available data suggest that interventions directed at multiple targets in the energy homeostasis system may be necessary to achieve and maintain weight loss.
Collapse
Affiliation(s)
- Deborah L Drazen
- Department of Psychiatry, University of Cincinnati College of Medicine, 231 Albert Sabin Way, MSB G059, Cincinnati, OH 45267-0559, USA.
| | | |
Collapse
|
1508
|
Abstract
PURPOSE OF REVIEW Obesity continues to increase around the globe, and creates a major health problem because of its comorbidities such as diabetes and cardiovascular disease. Considering that the major cause of most human obesities is the modern lifestyle in a rapidly changing environment, this suggests that the battle is between brain areas controlling internal metabolic homeostasis and those dealing with cognitive and emotional processing of external information. Thus, this review is intended to bridge this apparent neural dichotomy and highlight possible ways of interactions between these homeostatic and nonhomeostatic systems. RECENT FINDINGS Rapid progress has been made in identifying the molecular mechanisms underlying the neural circuits regulating food intake and energy balance. Specific populations of peptidergic neurons in the medial hypothalamus can be considered metabolic integrators sensing both short and long-term availability of fuels. These cell groups in turn connect with various other brain regions to orchestrate adaptive responses through changes in food intake, as well as endocrine and autonomic responses. Alternatively, the impact of the environmental changes on ingestive behavior have been documented, and many of the underlying cortico-limbic pathways are starting to be identified. It has been proposed that a switch from instinctual control of energy balance to a more cognitive control will be necessary to stop the obesity epidemic. SUMMARY The realization that there is nothing wrong with the homeostatic regulatory system in common obesity suggests that a systematic investigation of pathways that link the 'cognitive' with the 'metabolic' brain might be a more promising area of research. Such an investigation would reveal new molecular and cellular mechanisms providing new drug targets, leading to behavioral therapies that are directed more towards the causes of common obesity.
Collapse
Affiliation(s)
- Hans-Rudolf Berthoud
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| |
Collapse
|
1509
|
Koteja P, Carter PA, Swallow JG, Garland T. Food wasting by house mice: variation among individuals, families, and genetic lines. Physiol Behav 2003; 80:375-83. [PMID: 14637238 DOI: 10.1016/j.physbeh.2003.09.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Under ad libitum conditions, laboratory house mice (Mus domesticus) fragment considerable amounts of pelleted food and leave it scattered in their cages. The proportion of food thus wasted (in relation to food eaten) varies remarkably among individuals, from 2% to 40%, but is highly consistent in consecutive trials, even when the mice were moved from 22 to -10 degrees C and food consumption doubled. Food wasting did not differ either between the sexes or between genetic lines that had been selected (10 generations) for high voluntary wheel-running behavior (n=4) and their unselected control lines (n=4). However, it varied significantly among replicate lines within the selection groups and among families within the lines (coefficient of intraclass correlation for full sibs, rhof=0.41 in room temperature trials and rhof=0.34 in cold trials). Moreover, the percent of food wasted was negatively correlated with food consumption in the cold trials (males: r=-.36, females: r=-.20) and with total litter mass at weaning (the litters into which they were born; r=-.24), two traits that may affect Darwinian fitness. We conclude that food wastage should not be ignored without justification in calculations of food consumption. In addition, "table manners" can convey reliable information about family origin of an individual and its quality, and therefore could potentially play a role in establishment of social status.
Collapse
Affiliation(s)
- Paweł Koteja
- Institute of Environmental Sciences, Jagiellonian University, ul. Ingardena 6, 30-060 Krakow, Poland.
| | | | | | | |
Collapse
|
1510
|
Affiliation(s)
- Kazuhiro Takahashi
- Department of Molecular Biology and Applied Physiology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan.
| |
Collapse
|
1511
|
Nan YS, Feng GG, Hotta Y, Nishiwaki K, Shimada Y, Ishikawa A, Kurimoto N, Shigei T, Ishikawa N. Neuropeptide Y enhances permeability across a rat aortic endothelial cell monolayer. Am J Physiol Heart Circ Physiol 2003; 286:H1027-33. [PMID: 14576078 DOI: 10.1152/ajpheart.00630.2003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previously, in vivo studies showed that neuropeptide Y (NPY) elevates vascular permeability in isolated lung perfusion preparations, possibly through binding to the NPY Y(3) receptor. The present study used monolayers in a double-chamber culture method under conditions of normoxia (5% CO(2)-20% O(2)-75% N(2)) or hypoxia (5% CO(2)-5% O(2)-90% N(2)) to test the hypothesis that NPY directly affects rat aortic endothelial cells (RAECs). RAECs were cultured on the base of the upper chamber, into which FITC-labeled albumin was introduced, and permeation into the lower chamber was measured. The RAEC monolayer was treated with 10(-8)-3 x 10(-7) M NPY for 2 h in normoxia or hypoxia. In hypoxia, NPY concentration dependently increased the permeability of the RAEC monolayer, whereas in normoxia no significant change was observed. Peptide YY, NPY Y(1), and NPY Y(2) receptor agonists and NPY Y(1) receptor antagonist exerted no significant effects under hypoxic conditions. NPY-(18-36), an NPY Y(3) receptor antagonist, elicited an inhibitory action on the NPY-induced increase in monolayer permeability. Furthermore, neither N-monomethyl-l-arginine, a nitric oxide synthase inhibitor, the bradykinin B(2) receptor antagonist FK-3657, nor the vascular endothelial growth factor receptor-coupled tyrosine kinase inhibitor tyrphostin SU-1498, injected into the medium of the upper chamber, affected the NPY-induced permeability changes under hypoxic conditions. The results suggest that the NPY-induced increase in permeability across the RAEC monolayer is closely related to low O(2) tension, possibly mediated by direct action on the NPY Y(3) receptor expressed on the endothelial cell membrane. Furthermore, this NPY-induced increase is not likely due to nitric oxide, bradykinin, or vascular endothelial growth factor.
Collapse
Affiliation(s)
- Yong-Shan Nan
- Dept. of Pharmacology, Aichi Medical Univ., School of Medicine, Nagakute, Aichi 480-1195, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
1512
|
Ishii Y, Blundell JE, Halford JCG, Rodgers RJ. Palatability, food intake and the behavioural satiety sequence in male rats. Physiol Behav 2003; 80:37-47. [PMID: 14568306 DOI: 10.1016/s0031-9384(03)00207-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Food intake is influenced not only by nutritional status but also by diverse environmental factors. Indeed, a unique quality of food reward is its strong modulation by palatability cues, such as taste, with animals generally preferring diets that are sweet and avoiding those that are either bitter or sour. As appetite suppressants (including those currently in development) could alter food intake by modifying taste sensitivity and/or palatability, the aim of the present study was to characterise the influence of taste adulteration on the normal structure of feeding behaviour, i.e., the behavioural satiety sequence (BSS). Adult male rats were initially habituated both to the basic test diet (mash) and the test arena. Following stabilisation of basal intake, a continuous monitoring technique was used to profile behaviour in weekly 1-h sessions during which the animals were presented, in counterbalanced order, with the basic diet (control) or one of four taste-adulterated variants (0.015% quinine, 0.04% quinine, 0.2% saccharin, 0.3% saccharin). Food intake was strongly suppressed by the higher quinine concentration but was not significantly altered by any of the other additives. Behavioural analysis revealed that this anorectic-like response to 0.04% quinine-adulterated food was associated with a significant reduction in the peak feeding response, highly atypical intermittent food sampling/digging and the virtual absence of resting behaviour. Importantly, this pattern of behavioural change is readily distinguishable from those seen in response to other manipulations that reduce intake, including selective anorectics, sedatives and psychostimulants. Despite the lack of significant effect on food intake or the duration of feeding behaviour, dietary adulteration with 0.015% quinine (and, to a lesser degree, 0.3% saccharin) produced some effects on behavioural structure/time course consistent with a mild aversive response, i.e., bouts of midsession food sampling and a delay in the transition from eating to resting. Data are discussed in relation to the specific behavioural signature to quinine-induced anorexia and its potential utility in identifying appetite suppressants that may modify intake via changes in taste sensitivity and/or palatability.
Collapse
Affiliation(s)
- Y Ishii
- Behavioural Pharmacology Laboratory, School of Psychology, University of Leeds, LS2 9JT, Leeds, UK
| | | | | | | |
Collapse
|
1513
|
Picó C, Oliver P, Sánchez J, Palou A. Gastric leptin: a putative role in the short-term regulation of food intake. Br J Nutr 2003; 90:735-41. [PMID: 13129441 DOI: 10.1079/bjn2003945] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The discovery of the production of leptin by the stomach, in addition to its production by adipose tissue, has initiated new investigation into the possible role of this protein in the digestive physiology, in particular in the short-term control of energy balance. Leptin has been identified in the lower half of the stomach glands both in the pepsinogen granules of chief cells and in the granules of a specific endocrine cell type, suggesting that leptin action is exerted by both exocrine and endocrine pathways. Gastric leptin is sensitive to the nutritional state, being rapidly mobilized in response to food intake following fasting, or after the administration of satiety factors; this suggests a role for this protein in the short-term regulation of feeding, acting in collaboration with satiety peptides such as cholecystokinin. Leptin, produced by gastric cells and by adipocytes, could act on both acute and chronic regulation of feeding behaviour respectively, giving information to the brain on the availability of external (food) and internal (fat depots) energy resources, thus participating in short- and long-term satiation.
Collapse
Affiliation(s)
- Catalina Picó
- Department of Fundamental Biology and Health Sciences, University of the Balearic Islands, Palma de Mallorca, Spain
| | | | | | | |
Collapse
|
1514
|
Barroso I, Luan J, Middelberg RPS, Harding AH, Franks PW, Jakes RW, Clayton D, Schafer AJ, O'Rahilly S, Wareham NJ. Candidate gene association study in type 2 diabetes indicates a role for genes involved in beta-cell function as well as insulin action. PLoS Biol 2003; 1:E20. [PMID: 14551916 PMCID: PMC212698 DOI: 10.1371/journal.pbio.0000020] [Citation(s) in RCA: 226] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2003] [Accepted: 08/08/2003] [Indexed: 01/24/2023] Open
Abstract
Type 2 diabetes is an increasingly common, serious metabolic disorder with a substantial inherited component. It is characterised by defects in both insulin secretion and action. Progress in identification of specific genetic variants predisposing to the disease has been limited. To complement ongoing positional cloning efforts, we have undertaken a large-scale candidate gene association study. We examined 152 SNPs in 71 candidate genes for association with diabetes status and related phenotypes in 2,134 Caucasians in a case-control study and an independent quantitative trait (QT) cohort in the United Kingdom. Polymorphisms in five of 15 genes (33%) encoding molecules known to primarily influence pancreatic beta-cell function-ABCC8 (sulphonylurea receptor), KCNJ11 (KIR6.2), SLC2A2 (GLUT2), HNF4A (HNF4alpha), and INS (insulin)-significantly altered disease risk, and in three genes, the risk allele, haplotype, or both had a biologically consistent effect on a relevant physiological trait in the QT study. We examined 35 genes predicted to have their major influence on insulin action, and three (9%)-INSR, PIK3R1, and SOS1-showed significant associations with diabetes. These results confirm the genetic complexity of Type 2 diabetes and provide evidence that common variants in genes influencing pancreatic beta-cell function may make a significant contribution to the inherited component of this disease. This study additionally demonstrates that the systematic examination of panels of biological candidate genes in large, well-characterised populations can be an effective complement to positional cloning approaches. The absence of large single-gene effects and the detection of multiple small effects accentuate the need for the study of larger populations in order to reliably identify the size of effect we now expect for complex diseases.
Collapse
|
1515
|
Batterham RL, Cohen MA, Ellis SM, Le Roux CW, Withers DJ, Frost GS, Ghatei MA, Bloom SR. Inhibition of food intake in obese subjects by peptide YY3-36. N Engl J Med 2003; 349:941-8. [PMID: 12954742 DOI: 10.1056/nejmoa030204] [Citation(s) in RCA: 1062] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND The gut hormone fragment peptide YY3-36 (PYY) reduces appetite and food intake when infused into subjects of normal weight. In common with the adipocyte hormone leptin, PYY reduces food intake by modulating appetite circuits in the hypothalamus. However, in obesity there is a marked resistance to the action of leptin, which greatly limits its therapeutic effectiveness. We investigated whether obese subjects were also resistant to the anorectic effects of PYY. METHODS We compared the effects of PYY infusion on appetite and food intake in 12 obese and 12 lean subjects in a double-blind, placebo-controlled, crossover study. The plasma levels of PYY, ghrelin, leptin, and insulin were also determined. RESULTS Caloric intake during a buffet lunch offered two hours after the infusion of PYY was decreased by 30 percent in the obese subjects (P<0.001) and 31 percent in the lean subjects (P<0.001). PYY infusion also caused a significant decrease in the cumulative 24-hour caloric intake in both obese and lean subjects. PYY infusion reduced plasma levels of the appetite-stimulatory hormone ghrelin. Endogenous fasting and postprandial levels of PYY were significantly lower in obese subjects (the mean [+/-SE] fasting PYY levels were 10.2+/-0.7 pmol per liter in the obese group and 16.9+/-0.8 pmol per liter in the lean group, P<0.001). Furthermore, the fasting PYY levels correlated negatively with the body-mass index (r = -0.84, P<0.001). CONCLUSIONS We found that obese subjects were not resistant to the anorectic effects of PYY. Endogenous PYY levels were low in the obese subjects, suggesting that PYY deficiency may contribute to the pathogenesis of obesity.
Collapse
Affiliation(s)
- Rachel L Batterham
- Department of Metabolic Medicine, Imperial College Faculty of Medicine at Hammersmith Campus, Du Cane Rd., London W12 0NN, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
1516
|
Abstract
Prospective studies in humans and monkeys have informed our understanding of the mechanism of exercise-associated menstrual disorders (EAMD). These studies have provided convincing evidence that a key causal factor in the development of EAMD is an imbalance between energy intake and energy expenditure. This imbalance is created by the increased energy cost of exercise in the face of inadequate supplementation of caloric intake. Although one prospective study in humans documents the impact of weight loss, studies in nonhuman primates (Macaca fasicularis) reveal that EAMD can occur with unobtrusive compensatory mechanisms indicative of energy conservation. The onset of EAMD is variable between individuals, but is abrupt, and with little forewarning with respect to recognizable symptoms. Future studies aimed at mechanisms should build upon the finding that key metabolic signals such as T3 are correlated with both the onset and reversal of EAMD, perhaps by focusing on concomitant metabolic changes that directly influence GnRH neurons. Translational studies examining the energetics of the reversal of EAMD by manipulating food intake and or exercise should build on the findings in the monkey model. Lastly, because EAMD is often associated with disordered eating, future prospective studies in humans should incorporate the potential interaction of disordered eating and psychosocial stress on EAMD.
Collapse
Affiliation(s)
- Nancy I Williams
- Department of Kinesiology, Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
1517
|
Abstract
Over the past decade, there has been a tremendous increase in the understanding of the molecular and neural mechanisms that control food intake and body weight. Yet eating disorders and cachexia are still common, and obesity cases are rising at alarming rates. Thus, despite recent progress, an increased understanding of the molecular and neural substrates that control body weight homeostasis is a major public health goal. In this review, we discuss the mechanisms by which metabolic signals interact with key behavioral, neuroendocrine, and autonomic regulatory regions of the central nervous system. Additionally, we offer a model in which hormones such as leptin and ghrelin interact with similar central nervous system circuits and engage them in such a way as to maintain an appropriate and tight regulation of body weight and food intake. Our model predicts that overstimulation or understimulation of these central pathways can result in obesity, anorexia, or cachexia.
Collapse
Affiliation(s)
- Jeffrey M Zigman
- Department of Medicine and Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | |
Collapse
|
1518
|
Nonaka N, Shioda S, Niehoff ML, Banks WA. Characterization of blood-brain barrier permeability to PYY3-36 in the mouse. J Pharmacol Exp Ther 2003; 306:948-53. [PMID: 12750431 DOI: 10.1124/jpet.103.051821] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Peptide YY3-36 (PYY) has emerged as an important signal in the gut-brain axis, with peripherally administered PYY affecting feeding and brain function. For these effects to be direct, PYY would have to cross the blood-brain barrier (BBB). Here, we determined the permeability of the BBB to PYY radioactively labeled with 131I (I-PYY). Multiple-time regression analysis showed the unidirectional influx rate (Ki) from blood-to-brain for I-PYY to be 0.49 +/- 0.19 microl/g-min, a rate similar to that previously measured for leptin. Influx was not inhibited by 1 microg/mouse of unlabeled PYY, suggesting PYY crosses the BBB by transmembrane diffusion. About 0.176% of the i.v.-injected dose of I-PYY was taken up by brain, an amount similar to that for other peptides important in gut-brain communication. Capillary depletion showed that 69% of I-PYY crossed the BBB to enter the parenchymal space of the brain, and high-performance liquid chromatography demonstrated that the radioactivity in this space represented intact I-PYY. After intracerebroventricular injection, I-PYY crossed from brain to blood by the mechanism of bulk flow. We conclude that PYY crosses in both the blood-to-brain and brain-to-blood directions by nonsaturable mechanisms. Passage across the BBB provides a mechanism by which blood-borne PYY can affect appetite and brain function.
Collapse
Affiliation(s)
- Naoko Nonaka
- Oral Anatomy, School of Dentistry, Showa University, Tokyo, Japan
| | | | | | | |
Collapse
|
1519
|
Chu DQ, Cox HM, Costa SKP, Herzog H, Brain SD. The ability of neuropeptide Y to mediate responses in the murine cutaneous microvasculature: an analysis of the contribution of Y1 and Y2 receptors. Br J Pharmacol 2003; 140:422-30. [PMID: 12970079 PMCID: PMC1574044 DOI: 10.1038/sj.bjp.0705452] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. The ability of neuropeptide Y (NPY) to modulate skin blood flow, oedema formation and neutrophil accumulation was investigated. Experiments were designed to examine the possible contribution of the Y2 receptor, in addition to the Y1 receptor, through use of Y2 receptor knockout mice (Y2-/-) and selective receptor antagonists. 2. The development of a 99mTc clearance technique for the measurement of microvascular blood flow changes in mouse dorsal skin revealed a dose-dependent ability of picomole amounts of NPY, and also of the Y1-preferred agonist Pro34NPY and the Y2-preferred agonist PYY(3-36) to decrease blood flow. 3. The Y1 receptor antagonist BIBO3304 blocked responses to the Y1 agonist at the lower doses, but only partially inhibited at the higher doses tested in Y2+/+. In Y2-/- receptor mice, the responses to the Y2 agonist were abolished at the lower doses and partially reduced at the highest dose tested, while those to the Y1 agonist were similar in both Y2+/+ and Y2-/-receptor mice. 4. In Y2+/+ receptor mice, the simultaneous injection of the Y2 antagonist BIIE0246 with BIBO3304 abolished Y2 agonist-induced decreases in blood flow over the dose range used (10-100 pmol). When the Y2 receptor antagonist BIIE0246 was given alone, it was not able to significantly affect the PYY(3-36)-induced response, whereas the Y1 receptor antagonist BIBO3304 partially (P<0.001) inhibited the decrease in blood flow evoked by PYY(3-36) at the highest dose. 5. NPY did not mediate either oedema formation, even when investigated in the presence of the vasodilator calcitonin gene-related peptide (CGRP), or neutrophil accumulation in murine skin. 6. We conclude that the major vasoactive activity of NPY in the cutaneous microvasculature is to act in a potent manner to decrease blood flow via Y1 receptors, with evidence for the additional involvement of postjunctional Y2 receptors. Our results do not provide evidence for a potent proinflammatory activity of NPY in the cutaneous microvasculature.
Collapse
Affiliation(s)
- Duc Quyen Chu
- Centre for Cardiovascular Biology & Medicine, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL
| | - Helen M Cox
- Centre for Neuroscience Research, King's College London, Hodgkin Building, Guy's Campus, London SE1 1UL
| | - Soraia K P Costa
- Centre for Cardiovascular Biology & Medicine, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL
| | - Herbert Herzog
- Neurobiology Program, Garvan Institute of Medical Research, St Vincent's Hospital, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Susan D Brain
- Centre for Cardiovascular Biology & Medicine, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL
- Author for correspondence:
| |
Collapse
|
1520
|
Lecklin A, Lundell I, Salmela S, Männistö PT, Beck-Sickinger AG, Larhammar D. Agonists for neuropeptide Y receptors Y1 and Y5 stimulate different phases of feeding in guinea pigs. Br J Pharmacol 2003; 139:1433-40. [PMID: 12922930 PMCID: PMC1573983 DOI: 10.1038/sj.bjp.0705389] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
1. The stimulatory effect of neuropeptide Y (NPY) on food intake is well established but the roles of the receptor subtypes Y(1) and Y(5) have been difficult to define. We have studied the effects of two novel Y(1)-preferring and two Y(5)-preferring agonists on feeding in guinea pigs. 2. The Y(1)-preferring receptor agonists [Arg(6),Pro(34)]pNPY and [Phe(7),Pro(34)]pNPY had high affinity for the Y(1) receptor (K(i) values 0.07 and 0.04 nM, respectively) and nanomolar affinity for the Y(5) receptor. Administration of either compound into the third brain ventricle increased food intake equally to NPY. 3. The Y(5) agonist [Ala(31),Aib(32)]pNPY displayed a moderate affinity for the Y(5) receptor (K(i) 7.42 nM) and a low affinity for Y(1) (K(i) 1.7 micro M). This compound had only a modest effect on feeding. 4. The other Y(5)-preferring peptide [cPP(1-7),NPY(19-23),Ala(31),Aib(32),Gln(34)]hPP had a higher affinity at the Y(5) receptor (K(i) 1.32 nM) and also at the Y(1) receptor (K(i) 85 nM). It potently stimulated feeding: the food consumption after administration of this peptide was two-fold compared to NPY. 5. Our results support the view that both the receptor subtypes Y(1) and Y(5) are involved in the stimulation of feeding. As the action profiles of the Y(1) and Y(5) agonists on feeding parameters were different, it seems that they influence different phases of eating.
Collapse
Affiliation(s)
- Anne Lecklin
- Department of Neuroscience, Unit of Pharmacology, Uppsala University, Box 593, S-75124 Uppsala, Sweden.
| | | | | | | | | | | |
Collapse
|
1521
|
Parker SL, Parker MS, Buschauer A, Balasubramaniam A. Ligand internalization by cloned neuropeptide Y Y5 receptors excludes Y2 and Y4 receptor-selective peptides. Eur J Pharmacol 2003; 474:31-42. [PMID: 12909193 DOI: 10.1016/s0014-2999(03)02039-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In human embryonic kidney-293 (HEK-293) cells, the cloned human neuropeptide Y Y5 receptor saturably internalized agonists, with the rank order of neuropeptide Y-(19-23)-[Gly1,Ser3,Gln4,Thr6,Ala31,Aib32,Gln34]human pancreatic polypeptide (neuropeptide Y-Aib-pancreatic polypeptide)>human neuropeptide Y>porcine peptide YY>[Pro34]human peptide YY>[Leu31,Pro34]human peptide YY>>human peptide YY-(3-36). Human pancreatic polypeptide competed [125I]neuropeptide Y binding and internalization in neuropeptide Y Y5 receptor-expressing cells, but itself showed no internalization. The internalization was strongly dependent on temperature. The surface binding, and especially the internalization, of human neuropeptide Y were highly sensitive to the clathrin network inhibitor phenylarsine oxide, and to the cholesterol-complexing antibiotic filipin III. The internalized ligands were present in particles corresponding to secondary endosomes in Percoll gradients, but especially in particles banding with the acid hexosaminidase lysosomal marker. At any temperature tested, internalization of the neuropeptide Y Y5 receptor driven by human neuropeptide Y in HEK-293 cells was much slower than the internalization of the neuropeptide Y Y1 receptor reported in the same cells, or in Chinese hamster ovary (CHO) cells. The neuropeptide Y Y5 receptor subtype could be the metabotropic receptor responding to protracted challenges by neuropeptide Y-like peptides, and its density could be little sensitive to concentration of extracellular agonists.
Collapse
Affiliation(s)
- Steven L Parker
- Department of Pharmacology, University of Tennessee College of Medicine, 874 Union Avenue, Memphis, TN 38163, USA.
| | | | | | | |
Collapse
|
1522
|
Abstract
The role of peptides as signalling molecules in the nervous system has been studied for more than 30 years. Neuropeptides and their G-protein-coupled receptors are widely distributed throughout the body and they commonly occur with, and are complementary to, classic neurotransmitters. The functions of neuropeptides range from neurotransmitter to growth factor. They are present in glial cells, are hormones in the endocrine system, and are messengers in the immune system. Much evidence indicates that neuropeptides are of particular importance when the nervous system is challenged (eg, by stress, injury, or drug abuse). These features and the large number of neuropeptides and neuropeptide receptors provide many opportunities for the discovery of new drug targets for the treatment of nervous-system disorders. In fact, receptor-subtype-selective antagonists and agonists have been developed, and recently a substance P receptor (neurokinin 1) antagonist has been shown to have clinical efficacy in the treatment of major depression and chemotherapy-induced emesis. Several other neuropeptide receptor ligands are in clinical trials for various indications.
Collapse
Affiliation(s)
- Tomas Hökfelt
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden.
| | | | | |
Collapse
|
1523
|
Sainsbury A, Baldock PA, Schwarzer C, Ueno N, Enriquez RF, Couzens M, Inui A, Herzog H, Gardiner EM. Synergistic effects of Y2 and Y4 receptors on adiposity and bone mass revealed in double knockout mice. Mol Cell Biol 2003; 23:5225-33. [PMID: 12861009 PMCID: PMC165708 DOI: 10.1128/mcb.23.15.5225-5233.2003] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neuropeptide Y regulates numerous physiological processes via at least five different Y receptors, but the specific roles of each receptor are still unclear. We previously demonstrated that Y2 receptor knockout results in a lean phenotype, increased cancellous bone volume, and an increase in plasma pancreatic polypeptide (PP), a ligand for Y4 receptors. PP-overexpressing mice are also known to have a lean phenotype. Deletion of the Y4 receptor also produced a lean phenotype and increased plasma PP levels. We therefore hypothesized that part of the Y2 phenotype results from increased PP action on Y4 receptors and tested this in PP transgenic Y4(-/-) and Y2(-/-) Y4(-/-) double knockout mice. Bone mass was not altered in Y4 knockout mice. Surprisingly, despite significant hyperphagia, Y2(-/-) Y4(-/-) mice retained a markedly lean phenotype, with reduced body weight, white adipose tissue mass, leptinemia, and insulinemia. Furthermore, bone volume was also increased threefold in Y2(-/-) Y4(-/-) mice, and this was associated with enhanced osteoblastic activity. These changes were more pronounced than those observed in Y2(-/-) mice, suggesting synergy between Y2 and Y4 receptor pathways. The lack of bone changes in PP transgenic mice suggests that PP alone is not responsible for the bone mass increases but might play a major role in the lean phenotype. However, a synergistic interaction between Y2 and Y4 pathways seems to regulate bone volume and adiposity and could have important implications for possible interventions in obesity and for anabolic treatment of osteoporotic bone loss.
Collapse
Affiliation(s)
- Amanda Sainsbury
- Neurobiology Program, Garvan Institute of Medical Research, St Vincent's Hospital, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
1524
|
Rodrigues AM, Suplicy HL, Radominski RB. Controle neuroendócrino do peso corporal: implicações na gênese da obesidade. ACTA ACUST UNITED AC 2003. [DOI: 10.1590/s0004-27302003000400012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
O peso corporal é regulado por uma interação complexa entre hormônios e neuropeptídeos, sob o controle principal de núcleos hipotalâmicos. Mutações nos genes de hormônios e neuropeptídeos, de seus receptores ou de elementos regulatórios, têm sido descritas na espécie humana, mas são tidas como raras, não explicando as formas mais comuns de obesidade. No entanto, o estudo destas mutações tem propiciado um grande avanço nos conhecimentos sobre a base genética e a fisiopatologia da obesidade, possibilitando o estudo e abrindo perspectivas para o desenvolvimento de novas modalidades terapêuticas. Recentemente, demonstrou-se que mutações no receptor 4 da melanocortina podiam ser encontradas em até 5% dos casos de obesidade severa, representando até o presente momento a forma mais prevalente de obesidade monogênica na espécie humana. Nesta revisão, são discutidas as diversas mutações descritas nos seres humanos de elementos da rede neuroendócrina de controle do peso corporal, bem como as implicações dos mesmos na gênese da obesidade.
Collapse
|
1525
|
Fetissov SO, Xu ZQ, Byrne LC, Hassani H, Ernfors P, Hökfelt T. Neuropeptide y targets in the hypothalamus: nitric oxide synthesizing neurones express Y1 receptor. J Neuroendocrinol 2003; 15:754-60. [PMID: 12834436 DOI: 10.1046/j.1365-2826.2003.01051.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neuropeptide Y (NPY)-expressing neurones in the arcuate nucleus densely innervate many hypothalamic nuclei. To determine the neurochemical phenotype of target neurones for NPY, we studied the immunohistochemical localization of the NPY Y1 receptor (Y1R) in discrete subpopulations of neurones in the rat hypothalamus. Among several tested populations, including hypocretin/orexin-, melanin-concentrating hormone (MCH)- and nitric oxide synthase (NOS)-positive neurones, only the latter were found to coexpress the Y1R. Numerous Y1R/NOS-positive neurones were found as a densely packaged group of cells located ventrolateral to the ventromedial nucleus, forming a band ascending towards the fornix. Lower numbers of Y1R/NOS-positive neurones were found in the perifornical area and in the peri- and paraventricular nuclei. Expression of the Y1R gene was found in the same locations in the mouse by colocalizing beta-galactosidase, a Y1R gene reporter, with NOS in a Y1R knockout mouse. To explore possible downstream targets of NO in the rat hypothalamus, the NO-regulated molecule cGMP was analysed immunohistochemically after incubation of brain slices with sodium nitroprusside, an NO donor. We observed several cGMP-positive cell bodies in the arcuate nucleus, cGMP-positive blood vessels and a cGMP-positive network of thin fibres, some of which colocalized with choline acetyltransferase.
Collapse
Affiliation(s)
- S O Fetissov
- Department of Neuroscience B3:4, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
1526
|
Loktionov A. Common gene polymorphisms and nutrition: emerging links with pathogenesis of multifactorial chronic diseases (review). J Nutr Biochem 2003; 14:426-51. [PMID: 12948874 DOI: 10.1016/s0955-2863(03)00032-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Rapid progress in human genome decoding has accelerated search for the role of gene polymorphisms in the pathogenesis of complex multifactorial diseases. This review summarizes the results of recent studies on the associations of common gene variants with multifactorial chronic conditions strongly affected by nutritional factors. Three main individual sections discuss genes related to energy homeostasis regulation and obesity, cardiovascular disease (CVD), and cancer. It is evident that several major chronic diseases are closely related (often through obesity) to deregulation of energy homeostasis. Multiple polymorphic genes encoding central and peripheral determinants of energy intake and expenditure have been revealed over the past decade. Food intake control may be affected by polymorphisms in the genes encoding taste receptors and a number of peripheral signaling peptides such as insulin, leptin, ghrelin, cholecystokinin, and corresponding receptors. Polymorphic central regulators of energy intake include hypothalamic neuropeptide Y, agouti-related protein, melanocortin pathway factors, CART (cocaine- and amphetamine-regulated transcript), some other neuropeptides, and receptors for these molecules. Potentially important polymorphisms in the genes encoding energy expenditure modulators (alpha- and beta- adrenoceptors, uncoupling proteins, and regulators of adipocyte growth and differentiation) are also discussed. CVD-related gene polymorphisms comprising those involved in the pathogenesis of atherosclerosis, blood pressure regulation, hemostasis control, and homocysteine metabolism are considered in a separate section with emphasis on multiple polymorphisms affecting lipid transport and metabolism and their interactions with diet. Cancer-associated polymorphisms are discussed for groups of genes encoding enzymes of xenobiotic metabolism, DNA repair enzymes, factors involved in the cell cycle control, hormonal regulation-associated proteins, enzymes related to DNA methylation through folate metabolism, and angiogenesis-related factors. There is an apparent progress in the field with hundreds of new gene polymorphisms discovered and characterized, however firm evidence consistently linking them with pathogenesis of complex chronic diseases is still limited. Ways of improving the efficiency of candidate gene approach-based studies are discussed in a short separate section. Successful unraveling of interaction between dietary factors, polymorphisms, and pathogenesis of several multifactorial diseases is exemplified by studies of folate metabolism in relation to CVD and cancer. It appears that several new directions emerge as targets of research on the role of genetic variation in relation to diet and complex chronic diseases. Regulation of energy homeostasis is a fundamental problem insufficiently investigated in this context so far. Impacts of genetic variation on systems controlling angiogenesis, inflammatory reactions, and cell growth and differentiation (comprising regulation of the cell cycle, DNA repair, and DNA methylation) are also largely unknown and need thorough analysis. These goals can be achieved by complex simultaneous analysis of multiple polymorphic genes controlling carefully defined and selected elements of relevant metabolic and regulatory pathways in meticulously designed large-scale studies.
Collapse
|
1527
|
Huang XF, Han M, Storlien LH. The level of NPY receptor mRNA expression in diet-induced obese and resistant mice. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2003; 115:21-8. [PMID: 12824051 DOI: 10.1016/s0169-328x(03)00174-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Some mice become obese whereas others remain lean when raised on a high-energy diet. This study examined the levels of neuropeptide Y (NPY), and of Y1, Y2, Y5 and leptin receptor mRNA expression in the hypothalamic arcuate nucleus (Arc) of chronic high-energy diet-induced obese (DIO) and resistant (DR) mice. Forty mice were divided into two groups and fed either a high-fat (HF: 40% of calories from fat, 20% of calories from saturated fat; n=34) or low-fat (LF: 10% of calories from fat, 1% from saturated fat; n=6) diet. After 22 weeks of feeding, visceral fat accumulation was 69% higher in DIO mice compared with DR mice, and the former showed a moderate level of glucose intolerance. In DIO mice, the levels of NPY and leptin receptor mRNA expressions were significantly higher than in LF mice (+32 and +14%, P<0.001 and 0.05 respectively), indicating central leptin resistance, whereas the DR and LF groups did not differ. The level of Y2 receptor mRNA expression was similar between the DIO and LF groups but, importantly, was reduced approximately 20% in DR mice (P<0.005). The level of Y5 receptor mRNA was 36% lower in DR mice than DIO mice (P<0.05). The differences between DIO and DR mice identified by this study may assist in a better understanding of genetic predisposition to an increased fat deposition induced by a chronic high-fat diet. A low level of Y2 and Y5 receptor mRNA expression may contribute to the prevention of chronic high-energy diet-induced obesity in DR mice.
Collapse
Affiliation(s)
- Xu-Feng Huang
- Molecular Neurobiology Laboratory, Department of Biomedical Science, University of Wollongong, Northfield Avenue, Wollongong NSW2522, Australia.
| | | | | |
Collapse
|
1528
|
Ishii Y, Blundell JE, Halford JCG, Rodgers RJ. Effects of systematic variation in presatiation and fasting on the behavioural satiety sequence in male rats. Physiol Behav 2003; 79:227-38. [PMID: 12834794 DOI: 10.1016/s0031-9384(03)00066-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Preclinical research on the neurobiology of appetite regulation is increasingly employing detailed behavioural assessment in addition to measures of food intake. One of the most widely used approaches examines treatment effects on the behavioural satiety sequence (BSS), a concept describing the predictable pattern of behavioural change (feeding-->grooming-->resting) associated with the process of satiation in rats, mice and other mammals. Surprisingly, however, comparatively little published detail is available concerning the impact of more natural appetite modulators on the BSS. In two separate experiments, a continuous monitoring technique was used to calibrate the effects of prefeeding (3, 6 and 9 min) and prior fasting (3, 6 and 12 h) on the microstructure of rat behaviour during a 1 h test with palatable mash. Prefeeding significantly increased eating latencies and reduced both food intake and total duration (but not frequency) of feeding behaviour. The reduction in time spent eating was most evident during the first 15 min of the test when feeding is normally at peak levels. Although behavioural structure was fully preserved in all test conditions, the two larger preloads resulted in shifts to the left (i.e., an acceleration) in the BSS. In contrast, fasting for 6 and 12 h (but not 3 h) increased food intake and duration (but not frequency) of feeding behaviour in the early part of the test. All fasting conditions (including 3 h) produced considerable shifts to the right (i.e., a delay) in the BSS, confirming the greater sensitivity of this measure relative to food intake per se. The potential utility of these reference profiles is discussed in relation to drug-induced changes in food intake and the BSS.
Collapse
Affiliation(s)
- Y Ishii
- Behavioural Pharmacology Laboratory, School of Psychology, University of Leeds, Leeds LS2 9JT, UK
| | | | | | | |
Collapse
|
1529
|
Xu B, Goulding EH, Zang K, Cepoi D, Cone RD, Jones KR, Tecott LH, Reichardt LF. Brain-derived neurotrophic factor regulates energy balance downstream of melanocortin-4 receptor. Nat Neurosci 2003; 6:736-42. [PMID: 12796784 PMCID: PMC2710100 DOI: 10.1038/nn1073] [Citation(s) in RCA: 652] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2003] [Accepted: 05/07/2003] [Indexed: 01/19/2023]
Abstract
The melanocortin-4 receptor (MC4R) is critically involved in regulating energy balance, and obesity has been observed in mice with mutations in the gene for brain-derived neurotrophic factor (BDNF). Here we report that BDNF is expressed at high levels in the ventromedial hypothalamus (VMH) where its expression is regulated by nutritional state and by MC4R signaling. In addition, similar to MC4R mutants, mouse mutants that expresses the BDNF receptor TrkB at a quarter of the normal amount showed hyperphagia and excessive weight gain on higher-fat diets. Furthermore, BDNF infusion into the brain suppressed the hyperphagia and excessive weight gain observed on higher-fat diets in mice with deficient MC4R signaling. These results show that MC4R signaling controls BDNF expression in the VMH and support the hypothesis that BDNF is an important effector through which MC4R signaling controls energy balance.
Collapse
Affiliation(s)
- Baoji Xu
- Howard Hughes Medical Institute, University of California, San Francisco, California 94143, USA.
| | | | | | | | | | | | | | | |
Collapse
|
1530
|
Segal-Lieberman G, Trombly DJ, Juthani V, Wang X, Maratos-Flier E. NPY ablation in C57BL/6 mice leads to mild obesity and to an impaired refeeding response to fasting. Am J Physiol Endocrinol Metab 2003; 284:E1131-9. [PMID: 12582011 DOI: 10.1152/ajpendo.00491.2002] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neuropeptide Y (NPY) is an orexigenic (appetite-stimulating) peptide that plays an important role in regulating energy balance. When administered directly into the central nervous system, animals exhibit an immediate increase in feeding behavior, and repetitive injections or chronic infusions lead to obesity. Surprisingly, initial studies of Npy(-/-) mice on a mixed genetic background did not reveal deficits in energy balance, with the exception of an attenuation in obesity seen in ob/ob mice in which the NPY gene was also deleted. Here, we show that, on a C57BL/6 background, NPY ablation is associated with an increase in body weight and adiposity and a significant defect in refeeding after a fast. This impaired refeeding response in Npy(-/-) mice resulted in a deficit in weight gain in these animals after 24 h of refeeding. These data indicate that genetic background must be taken into account when the biological role of NPY is evaluated. When examined on a C57BL/6 background, NPY is important for the normal refeeding response after starvation, and its absence promotes mild obesity.
Collapse
|
1531
|
Hyland NP, Sjöberg F, Tough IR, Herzog H, Cox HM. Functional consequences of neuropeptide Y Y 2 receptor knockout and Y2 antagonism in mouse and human colonic tissues. Br J Pharmacol 2003; 139:863-71. [PMID: 12813010 PMCID: PMC1573894 DOI: 10.1038/sj.bjp.0705298] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
1 Neuropeptide Y (NPY), peptide YY (PYY) and pancreatic polypeptide (PP) differentially activate three Y receptors (Y(1), Y(2) and Y(4)) in mouse and human isolated colon. 2 The aim of this study was to characterise Y(2) receptor-mediated responses in colon mucosa and longitudinal smooth muscle preparations from wild type (Y(2)+/+) and knockout (Y(2)-/-) mice and to compare the former with human mucosal Y agonist responses. Inhibition of mucosal short-circuit current and increases in muscle tone were monitored in colonic tissues from Y(2)+/+ and Y(2)-/- mice+/-Y(1) ((R)-N-[[4-(aminocarbonylaminomethyl)phenyl)methyl]-N(2)-(diphenylacetyl)-argininamide-trifluoroacetate (BIBO3304) or Y(2) (S)-N(2)-[[1-[2-[4-[(R,S)-5,11-dihydro-6(6H)-oxodibenz[b,e]azepin-11-yl]-1-piperazinyl]-2-oxoethyl]cyclopentyl]acetyl]-N-[2-[1,2-dihydro-3,5(4H)-dioxo-1,2-diphenyl-3H-1,2,4-triazol-4-yl]ethyl]-argininamide (BIIE0246) antagonists. 3 Predictably, Y(2)-/- tissues were insensitive to Y(2)-preferred agonist PYY(3-36) (</=100 nM), but unexpectedly Y(4)-preferred PP responses were right-shifted probably as a consequence of elevated circulating PP levels, particularly in male Y(2)-/- mice (Sainsbury et al., 2002). 4 BIBO3304 and BIIE0246 elevated mucosal ion transport, indicating blockade of inhibitory mucosal tone in Y(2)+/+ tissue. While BIBO3304 effects were unchanged, those to BIIE0246 were absent in Y(2)-/- mucosae. Neither antagonist altered muscle tone; however, BIIE0246 blocked NPY and PYY(3-36) increases in Y(2)+/+ basal tone. BIBO3304 abolished residual Y(1)-mediated NPY responses in Y(2)-/- smooth muscle. 5 Tetrodotoxin significantly reduced BIIE0246 and PYY(3-36) effects in Y(2)+/+ mouse and human mucosae, but had no effect upon Y-agonist contractile responses, indicating that Y(2) receptors are located on submucosal, but not myenteric neurones. 6 Tonic activation of submucosal Y(2) receptors by endogenous NPY, PYY or PYY(3-36) could indirectly reduce mucosal ion transport in murine and human colon, while direct activation of Y(2) receptors on longitudinal muscle results in contraction.
Collapse
Affiliation(s)
- Niall P Hyland
- Centre for Neuroscience Research, King's College London, GKT School of Biomedical Sciences, Guy's Campus, London SE1 1UL
| | - Frida Sjöberg
- Centre for Neuroscience Research, King's College London, GKT School of Biomedical Sciences, Guy's Campus, London SE1 1UL
| | - Iain R Tough
- Centre for Neuroscience Research, King's College London, GKT School of Biomedical Sciences, Guy's Campus, London SE1 1UL
| | - Herbert Herzog
- Neurobiology Program, Garvan Institute of Medical Research, 384, Victoria Street, NSW 2010, Sydney, Australia
| | - Helen M Cox
- Centre for Neuroscience Research, King's College London, GKT School of Biomedical Sciences, Guy's Campus, London SE1 1UL
- Author for correspondence:
| |
Collapse
|
1532
|
Abstract
Early onset obesity and type II diabetes is rapidly becoming an epidemic, especially within the United States. This dramatic increase is likely due to many factors including both prenatal and postnatal environmental cues. The purpose of this review is to highlight some of the recent advances in our knowledge of the development of the hypothalamic circuits involved in the regulation of energy balance, with a focus on the neuropeptide Y (NPY) system. Unlike the adult rat, during the postnatal period NPY is transiently expressed in several hypothalamic regions, along with the expected expression within the arcuate nucleus (ARH). These transient populations of NPY neurons during the postnatal period may provide local NPY production to sustain the necessary energy intake during this critical growth phase. This may be physiologically important since ARH-NPY projections do not fully develop until the 3rd postnatal week. The significance of this ontogeny is that many peripheral metabolic signals have little effect of feeding prior to the development of the ARH projections. The essential questions now are whether prenatal and/or postnatal exposure to high levels of insulin or leptin during development can cause permanent changes in the function of hypothalamic circuits. It is vital to understand not only the natural development of the hypothalamic circuits that regulate energy homeostasis, but also their abnormal development caused by maternal and postnatal environmental cues. This will be pivotal for designing intervention and therapeutics to treat early onset obesity/type II diabetes, which may very well need to be different from those designed to prevent/treat adult onset obesity/type II diabetes.
Collapse
Affiliation(s)
- Kevin L Grove
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 Northwest 185th Ave, Beaverton, OR 97006, USA.
| | | |
Collapse
|
1533
|
Cowley MA, Cone RD, Enriori P, Louiselle I, Williams SM, Evans AE. Electrophysiological actions of peripheral hormones on melanocortin neurons. Ann N Y Acad Sci 2003; 994:175-86. [PMID: 12851314 DOI: 10.1111/j.1749-6632.2003.tb03178.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Neurons of the arcuate nucleus of the hypothalamus (ARH) appear to be sites of convergence of central and peripheral signals of energy stores, and profoundly modulate the activity of the melanocortin circuits, providing a strong rationale for pursuing these circuits as therapeutic targets for disorders of energy homeostasis. Recently, tremendous advances have been made in identifying genes and pathways important to regulating energy homeostasis, particularly the hormone leptin and its receptor. This hormone/receptor pair is expressed at high levels in the so-called satiety centers in the hypothalamus, and at lower levels elsewhere in the body. Recent studies in our lab and those of our collaborators have shown that leptin modulates different populations of hypothalamic cells in different ways, rapidly activating POMC neurons and inhibiting NPY/AgRP neurons. In this report, we outline an integrated model of leptin's action in the arcuate nucleus of the hypothalamus, derived from our electrophysiological studies of brain slice preparations taken from transgenic mice that have been bred to express a variety of fluorescent proteins in specific cell types. We also discuss the recently withdrawn obesity drug fenfluramine, which appears to act on POMC neurons via the serotonin 2C receptor. Nutrient-sensing serotonin neurons may project from the raphe nuclei in the brainstem to the hypothalamus; within the arcuate nucleus, serotonin signals are integrated with others such as leptin, ghrelin, and peptide YY(3-36) from the gut, to produce a coordinated response to nutrient state. Finally, we review the current inquiries into the ability of the hormone ghrelin to stimulate appetite by its action of NPY neurons and inhibition of POMC neurons.
Collapse
Affiliation(s)
- Michael A Cowley
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, Oregon 97006, USA.
| | | | | | | | | | | |
Collapse
|
1534
|
Abstract
The gut hormone peptide YY (PYY) belongs to the pancreatic polypeptide (PP) family along with PP and neuropeptide Y (NPY). These peptides mediate their effects through the NPY receptors of which there are several subtypes (Y1, Y2, Y4, and Y5). The L cells of the gastrointestinal tract are the major source of PYY, which exists in two endogenous forms: PYY(1-36) and PYY(3-36). The latter is produced by the action of the enzyme dipeptidyl peptidase-IV (DPP-IV). PYY(1-36) binds to and activates at least three Y receptor subtypes (Y1, Y2, and Y5), whereas PYY(3-36) is more selective for Y2 receptor (Y2R). The hypothalamic arcuate nucleus, a key brain area regulating appetite, has access to nutrients and hormones within the peripheral circulation. NPY neurons within the arcuate nucleus express the Y2R. In response to food ingestion plasma PYY(3-36) concentrations rise within 15 min and plateau by approximately 90 min. The peak PYY(3-36) level achieved is proportional to the calories ingested, suggesting that PYY(3-36) may signal food ingestion from the gut to appetite-regulating circuits within the brain. We found that peripheral administration of PYY(3-36) inhibited food intake in rodents and increased C-Fos immunoreactivity in the arcuate nucleus. Moreover, direct intra-arcuate administration of PYY(3-36) inhibited food intake. We have shown that Y2R null mice are resistant to the anorectic effects of peripherally administered PYY(3-36), suggesting that PYY(3-36) inhibits food intake through the Y2R. In humans, peripheral infusion of PYY(3-36), at a dose which produced normal postprandial concentrations, significantly decreased appetite and reduced food intake by 33% over 24 h. These findings suggest that PYY(3-36) released in response to a meal acts via the Y2R in the arcuate nucleus to physiologically regulate food intake.
Collapse
Affiliation(s)
- Rachell L Batterham
- Imperial College Faculty of Medicine, Hammersmith Campus, London, W12 0NN, United Kingdom.
| | | |
Collapse
|
1535
|
Jégou S, Mounien L, Boutelet I, Vaudry H. [The YY3-36 peptide, a new therapeutic weapon against obesity?]. Med Sci (Paris) 2003; 19:537-9. [PMID: 12836385 DOI: 10.1051/medsci/2003195537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
1536
|
|
1537
|
Asakawa A, Inui A, Yuzuriha H, Ueno N, Katsuura G, Fujimiya M, Fujino MA, Niijima A, Meguid MM, Kasuga M. Characterization of the effects of pancreatic polypeptide in the regulation of energy balance. Gastroenterology 2003; 124:1325-36. [PMID: 12730873 DOI: 10.1016/s0016-5085(03)00216-6] [Citation(s) in RCA: 227] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND & AIMS Pancreatic polypeptide (PP) belongs to a family of peptides including neuropeptide Y and peptide YY. We examined the role of PP in the regulation of body weight as well as the therapeutic potential of PP. METHODS We measured food intake, gastric emptying, oxygen consumption, and gene expression of hypothalamic neuropeptides, gastric ghrelin, and adipocytokines in mice after administering PP intraperitoneally. Peptide gene expression was also examined in PP-overexpressing mice. Vagal and sympathetic nerve activities were recorded after intravenous administration in rats. Effects of repeated administrations of PP on energy balance and on glucose and lipid metabolism were examined in both ob/ob obese mice and fatty liver Shionogi (FLS)-ob/ob obese mice. RESULTS Peripherally administered PP induced negative energy balance by decreasing food intake and gastric emptying while increasing energy expenditure. The mechanism involved modification of expression of feeding-regulatory peptides (decrease in orexigenic neuropeptide Y, orexin, and ghrelin along with an increase in anorexigenic urocortin) and activity of the vagovagal or vagosympathetic reflex arc. PP reduced leptin in white adipose tissue and corticotropin-releasing factor gene expression. The expression of gastric ghrelin and hypothalamic orexin was decreased in PP-overexpressing mice. Repeated administrations of PP decreased body weight gain and ameliorated insulin resistance and hyperlipidemia in both ob/ob obese mice and FLS-ob/ob obese mice. Liver enzyme abnormalities in FLS-ob/ob obese mice were also ameliorated by PP. CONCLUSIONS These observations indicate that PP may influence food intake, energy metabolism, and the expression of hypothalamic peptides and gastric ghrelin.
Collapse
Affiliation(s)
- Akihiro Asakawa
- Department of Clinical Molecular Medicine, Kobe University Graduate School of Meidicine, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1538
|
Gribble FM, Williams L, Simpson AK, Reimann F. A novel glucose-sensing mechanism contributing to glucagon-like peptide-1 secretion from the GLUTag cell line. Diabetes 2003; 52:1147-54. [PMID: 12716745 DOI: 10.2337/diabetes.52.5.1147] [Citation(s) in RCA: 299] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Glucagon-like peptide 1 (GLP-1) secretion from intestinal L-cells is triggered by luminal nutrients. We reported previously that glucose-triggered GLP-1 release from the L-cell model GLUTag involves closure of ATP-sensitive K+ (K(ATP)) channels. We show here that GLP-1 secretion and electrical activity of GLUTag cells is triggered not only by metabolizable sugars (glucose or fructose) but also by the nonmetabolizable monosaccharide methyl-alpha-glucopyranoside. Responses to glucose and methyl-alpha-glucopyranoside were impaired by the sodium-glucose cotransporter (SGLT) inhibitor phloridzin. SLGT1 and 3 were detected in GLUTag cells by RT-PCR. Whereas fructose closed K(ATP) channels, methyl-alpha-glucopyranoside increased the membrane conductance and generated an inward current. Low concentrations of glucose and methyl-alpha-glucopyranoside also triggered small inward currents and enhanced the action potential frequency. We conclude that whereas low concentrations of metabolizable sugars trigger GLP-1 secretion via K(ATP) channel closure, SGLT substrates generate small inward currents as a result of the electrogenic action of the transporter. This transporter-associated current can trigger electrical activity and secretion when the concentration of substrate is high or when outward currents are reduced by metabolic closure of the K(ATP) channels. Electrogenic sugar entry via SGLTs provides a novel mechanism for glucose sensing by neuroendocrine cells.
Collapse
Affiliation(s)
- Fiona M Gribble
- Department of Clinical Biochemistry, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 2QR, United Kingdom
| | | | | | | |
Collapse
|
1539
|
Korner J, Aronne LJ. The emerging science of body weight regulation and its impact on obesity treatment. J Clin Invest 2003. [PMID: 12618507 DOI: 10.1172/jci200317953] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Judith Korner
- Columbia University College of Physicians and Surgeons, New York, New York 10032, USA.
| | | |
Collapse
|
1540
|
Dhillo WS, Small CJ, Jethwa PH, Russell SH, Gardiner JV, Bewick GA, Seth A, Murphy KG, Ghatei MA, Bloom SR. Paraventricular nucleus administration of calcitonin gene-related peptide inhibits food intake and stimulates the hypothalamo-pituitary-adrenal axis. Endocrinology 2003; 144:1420-5. [PMID: 12639925 DOI: 10.1210/en.2002-220902] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Calcitonin gene-related protein (CGRP) inhibits food intake and stimulates the hypothalamo-pituitary-adrenal (HPA) axis after intracerebroventricular injection in rats. However, the hypothalamic site and mechanism of action are unknown. We investigated the effects of intraparaventricular nucleus administration (iPVN) of CGRP on food intake and the HPA axis in rats and the effect of CGRP on the release of hypothalamic neuropeptides in vitro. In addition, we investigated the effects of food deprivation on hypothalamic CGRP expression. CGRP dose-dependently reduced food intake in the first hour after iPVN injection in fasted male rats (saline, 5.1 +/- 0.8 g; 0.3 nmol CGRP, 1.1 +/- 0.5 g; P < 0.001 vs. saline). iPVN injection of CGRP(8-37) (a CGRP(1) receptor antagonist) alone had no effect on food intake. However, the reduction in food intake by iPVN CGRP was attenuated by prior administration of CGRP(8-37) [CGRP(8-37) (10 nmol)/CGRP (0.3 nmol), 3.0 +/- 0.8 g; P < 0.05 vs. 0.3 nmol CGRP]. CGRP (100 nM) stimulated the release of alpha-melanocyte stimulating hormone, cocaine- and amphetamine-related transcript, corticotropin-releasing hormone, and arginine vasopressin from hypothalamic explants to 127 +/- 19%, 148 +/- 10%, 158 +/- 17%, and 198 +/- 21% of basal levels, respectively (P < 0.05 vs. basal), but did not alter the release of either neuropeptide Y or agouti-related protein. Hypothalamic CGRP mRNA levels in 24-h fasted rats were increased to 130 +/- 8% of control levels [CGRP mRNA (arbitrary units), 4.75 +/- 0.4; controls, 3.65 +/- 0.34; P < 0.05]. Our data suggest that CGRP administered to the PVN inhibits food intake and stimulates the HPA axis.
Collapse
Affiliation(s)
- Waljit S Dhillo
- Endocrine Unit, Faculty of Medicine, Imperial College, Hammersmith Hospital, London W12 ONN, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1541
|
Piroli GG. Regulation of food intake: an 'old' actor plays a 'new' role. Mol Psychiatry 2003; 8:364-5. [PMID: 12740590 DOI: 10.1038/sj.mp.4001265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
1542
|
Korner J, Aronne LJ. The emerging science of body weight regulation and its impact on obesity treatment. J Clin Invest 2003; 111:565-70. [PMID: 12618507 PMCID: PMC151906 DOI: 10.1172/jci17953] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Judith Korner
- Columbia University College of Physicians and Surgeons, New York, New York 10032, USA.
| | | |
Collapse
|
1543
|
Cowley MA, Smith RG, Diano S, Tschöp M, Pronchuk N, Grove KL, Strasburger CJ, Bidlingmaier M, Esterman M, Heiman ML, Garcia-Segura LM, Nillni EA, Mendez P, Low MJ, Sotonyi P, Friedman JM, Liu H, Pinto S, Colmers WF, Cone RD, Horvath TL. The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis. Neuron 2003; 37:649-61. [PMID: 12597862 DOI: 10.1016/s0896-6273(03)00063-1] [Citation(s) in RCA: 1187] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The gastrointestinal peptide hormone ghrelin stimulates appetite in rodents and humans via hypothalamic actions. We discovered expression of ghrelin in a previously uncharacterized group of neurons adjacent to the third ventricle between the dorsal, ventral, paraventricular, and arcuate hypothalamic nuclei. These neurons send efferents onto key hypothalamic circuits, including those producing neuropeptide Y (NPY), Agouti-related protein (AGRP), proopiomelanocortin (POMC) products, and corticotropin-releasing hormone (CRH). Within the hypothalamus, ghrelin bound mostly on presynaptic terminals of NPY neurons. Using electrophysiological recordings, we found that ghrelin stimulated the activity of arcuate NPY neurons and mimicked the effect of NPY in the paraventricular nucleus of the hypothalamus (PVH). We propose that at these sites, release of ghrelin may stimulate the release of orexigenic peptides and neurotransmitters, thus representing a novel regulatory circuit controlling energy homeostasis.
Collapse
Affiliation(s)
- Michael A Cowley
- The Vollum Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1544
|
|
1545
|
Chapter 7. Recent developments in neuropeptide Y receptor modulators. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2003. [DOI: 10.1016/s0065-7743(03)38008-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
1546
|
Strand FL. Neuropeptides: general characteristics and neuropharmaceutical potential in treating CNS disorders. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2003; 61:1-37. [PMID: 14674607 DOI: 10.1007/978-3-0348-8049-7_1] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The general characteristics of neuropeptides are discussed as a background for the understanding of their role in regulation of physiological systems. The extent of those systems that are crucially affected by neuropeptides is vast and the complexity of their interactions makes the clinical focus on a specific neuropeptide unsatisfactory. The clinical potential of neuropeptides affecting eating disorders, CNS behavioral disorders and the neuroregenerative and neuroprotective action of neuropeptides is discussed. It is probable that successful neuropeptide therapeutics will depend upon the application of translational and combinational research using various ingenious combinations of neuropeptides, their agonists and antagonists, neuropeptide receptor agonists and antagonists, improved methods of delivery and the development of peptides targeted to the genetic profile of individual patients.
Collapse
Affiliation(s)
- Fleur L Strand
- New York University, 340 East 64th Street, New York, NY 10021, USA.
| |
Collapse
|
1547
|
Mizuno TM, Makimura H, Mobbs CV. The physiological function of the agouti-related peptide gene: the control of weight and metabolic rate. Ann Med 2003; 35:425-33. [PMID: 14572167 DOI: 10.1080/07853890310012076] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The central melanocortin system plays an important role in the regulation of energy homeostasis both in rodents and humans, and melanocortin receptors appear to be the core of this system. Alpha-melanocyte-stimulating hormone (alpha-MSH) inhibits feeding through melanocrtin 3 and 4 receptors (MC3-R and MC4-R) as an endogenous agonist. Although mutations in the agouti gene cause an over-expression of agouti peptide which antagonizes effects of alpha-MSH at MC4-R in the brain and causes obese phenotypes, there was no evidence for the presence of an endogenous antagonist for MC3-R and MC4-R until agouti related protein (AGRP) was identified. AGRP is expressed primarily in the hypothalamic arcuate nucleus and central administration of AGRP stimulates feeding and weight gain, and decreases metabolic rate. Although a complete deletion of the AGRP gene does not produce any significant metabolic phenotypes, reduction in AGRP expression by RNA interference is associated with increased metabolic rate along with reduced weight gain. The currently available data suggest that elevated AGRP mRNA along with reduced proopiomelanocortin (POMC) mRNA is associated with many types of obesity and agents antagonizing the effect of AGRP may be a potential therapeutic target in treating obesity and obesity-associated disorders in which endogenous hypothalamic AGRP is elevated.
Collapse
Affiliation(s)
- Tooru M Mizuno
- Fishberg Center for Neurobiology, Neurobiology of Aging Laboratories, Department of Geriatrics, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | |
Collapse
|
1548
|
Stanner S. The ‘third-helping’ hormone. NUTR BULL 2002. [DOI: 10.1046/j.1467-3010.2002.00275.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
1549
|
Abstract
The growing recognition of the health risks of obesity coupled with the difficulties in treating it successfully by lifestyle modification predicates a need for effective drug treatment. The history of drug treatment in the second half of the 20th century is, however, one of disappointment and concern over drug toxicity. However, the advances in our understanding of the mechanism of weight control, together with improved ways of evaluating anti-obesity drugs, has resulted in two effective compounds, sibutramine and orlistat, becoming available for clinical use. Sibutramine has actions on both energy intake and expenditure and had been shown to enhance weight loss and weight maintenance achieved by diet, in simple obesity as well as when accompanied by complications of diabetes or hypertension. About 50-80% of patients can achieve a >5% loss, significantly more than if patients receive the same lifestyle intervention with placebo. Orlistat, which acts peripherally to block the absorption of dietary fat, has had similar results in clinical trials; a recent study (XENDOS) has just reported results which show that the enhanced, albeit modest, weight loss achieved with orlistat delays the development of diabetes over a 4-year period. A number of other compounds are expected to complete or enter clinical trials over the next decade. There is considerable optimism that we will soon have the pharmacological tools needed to make the treatment of obesity feasible.
Collapse
Affiliation(s)
- N Finer
- Wellcome Trust Clinical Research Facility, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
1550
|
Sainsbury A, Schwarzer C, Couzens M, Herzog H. Y2 receptor deletion attenuates the type 2 diabetic syndrome of ob/ob mice. Diabetes 2002; 51:3420-7. [PMID: 12453895 DOI: 10.2337/diabetes.51.12.3420] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Hypothalamic neuropeptide Y (NPY) is implicated in the regulation of a variety of physiological functions, notably energy homeostasis and reproduction. Chronically elevated NPY levels in the hypothalamus, as in genetically obese ob/ob mice, are associated with obesity, a syndrome of type 2 diabetes, and infertility. However, it is not known which of the five cloned Y receptors mediate these effects. Here, we show that crossing the Y2 receptor knockout mouse (Y2(-/-)) onto the ob/ob background attenuates the increased adiposity, hyperinsulinemia, hyperglycemia, and increased hypothalamo-pituitary-adrenal (HPA) axis activity of ob/ob mice. Compared with lean controls, ob/ob mice had elevated expression of NPY and agouti-related protein (AgRP) mRNA in the arcuate nucleus and decreased expression of proopiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART) mRNA. Y2 deletion in ob/ob mice significantly increased the hypothalamic POMC mRNA expression, with no effect on NPY, AgRP, or CART expression. [Y2(-/-)ob/ob] mice were no different from ob/ob littermates with respect to food intake and body weight, and Y2 receptor deficiency had no beneficial effect on the infertility or the reduced hypothalamo-pituitary-gonadotropic function of ob/ob mice. These data demonstrate that Y2 receptors mediate the obese type 2 diabetes phenotype of ob/ob mice, possibly via alterations in melanocortin tonus in the arcuate nucleus and/or effects on the HPA axis.
Collapse
Affiliation(s)
- Amanda Sainsbury
- Neurobiology Program, Garvan Institute of Medical Research, St. Vincent's Hospital, 384 Victoria Street, Darlinghurst NSW 2010, Sydney, Australia
| | | | | | | |
Collapse
|