101
|
Functional characterisation of the bovine neuropeptide Y gene promoter and evaluation of the transcriptional activities of promoter haplotypes. Mol Biol Rep 2011; 39:919-28. [PMID: 21562764 DOI: 10.1007/s11033-011-0817-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 05/03/2011] [Indexed: 12/11/2022]
Abstract
Neuropeptide Y (NPY) is a potent orexigenic agent. The molecular mechanisms underlying the regulation of bovine NPY gene expression by its promoter region is currently unknown. The objectives of this research were to: (i) identify the SNPs in the promoter region of the bovine NPY gene, (ii) investigate the effects of these SNPs by measuring promoter transcriptional activities of different bovine NPY promoter haplotypes and; (iii) identify the minimal promoter region (MPR) required for basal activity of the NPY gene in vitro. Seventeen SNPs were identified in the promoter region. Of these, 14 affected putative transcription factors binding motifs including a TATA binding protein factor at -20, GC-Box factors SP1 at -170 and GATA binding motifs at -120 and -347. The SNPs were assigned to five major haplotypes (BtNPY_H1-5), of which BtNPY_H5 had maximum transcriptional activity. The region extending to -134 nt was identified as the MPR. This MPR was confirmed by the identification of a putative TATA box (-29 nt) and two SP1/GC binding sites (-94 and -118 nt), within this region. However, promoter expression was significantly enhanced when the construct contained the -614 to -1019 nt region. In conclusion, a number of SNPs characterised in the bovine NPY promoter especially those affecting the transcription factor binding sites, enhancer and repressor regions have the potential to affect NPY gene expression. Natural variation exists in the promoter region of the bovine NPY gene, which should be further explored for selection of energetic efficiency in cattle.
Collapse
|
102
|
García AP, Palou M, Sánchez J, Priego T, Palou A, Picó C. Moderate caloric restriction during gestation in rats alters adipose tissue sympathetic innervation and later adiposity in offspring. PLoS One 2011; 6:e17313. [PMID: 21364997 PMCID: PMC3041800 DOI: 10.1371/journal.pone.0017313] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 01/28/2011] [Indexed: 11/19/2022] Open
Abstract
Maternal prenatal undernutrition predisposes offspring to higher adiposity in adulthood. Mechanisms involved in these programming effects, apart from those described in central nervous system development, have not been established. Here we aimed to evaluate whether moderate caloric restriction during early pregnancy in rats affects white adipose tissue (WAT) sympathetic innervation in the offspring, and its relationship with adiposity development. For this purpose, inguinal and retroperitoneal WAT (iWAT and rpWAT, respectively) were analyzed in male and female offspring of control and 20% caloric-restricted (from 1-12 d of pregnancy) (CR) dams. Body weight (BW), the weight, DNA-content, morphological features and the immunoreactive tyrosine hydroxylase and Neuropeptide Y area (TH+ and NPY+ respectively, performed by immunohistochemistry) of both fat depots, were studied at 25 d and 6 m of age, the latter after 2 m exposure to high fat diet. At 6 m of life, CR males but not females, exhibited greater BW, and greater weight and total DNA-content in iWAT, without changes in adipocytes size, suggesting the development of hyperplasia in this depot. However, in rpWAT, CR males but not females, showed larger adipocyte diameter, with no changes in DNA-content, suggesting the development of hypertrophy. These parameters were not different between control and CR animals at the age of 25 d. In iWAT, both at 25 d and 6 m, CR males but not females, showed lower TH(+) and NPY(+), suggesting lower sympathetic innervation in CR males compared to control males. In rpWAT, at 6 m but not at 25 d, CR males but not females, showed lower TH(+) and NPY(+). Thus, the effects of caloric restriction during gestation on later adiposity and on the differences in the adult phenotype between internal and subcutaneous fat depots in the male offspring may be associated in part with specific alterations in sympathetic innervation, which may impact on WAT architecture.
Collapse
Affiliation(s)
- Ana Paula García
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics), University of Balearic Islands and CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Palma de Mallorca, Spain
| | - Mariona Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics), University of Balearic Islands and CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Palma de Mallorca, Spain
| | - Juana Sánchez
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics), University of Balearic Islands and CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Palma de Mallorca, Spain
| | - Teresa Priego
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics), University of Balearic Islands and CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Palma de Mallorca, Spain
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics), University of Balearic Islands and CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Palma de Mallorca, Spain
- * E-mail:
| | - Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics), University of Balearic Islands and CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Palma de Mallorca, Spain
| |
Collapse
|
103
|
SEDLÁČKOVÁ D, KOPEČKOVÁ J, PAPEŽOVÁ H, VYBÍRAL S, KVASNIČKOVÁ H, HILL M, NEDVÍDKOVÁ J. Changes of Plasma Obestatin, Ghrelin and NPY in Anorexia and Bulimia Nervosa Patients Before and After a High-Carbohydrate Breakfast. Physiol Res 2011; 60:165-73. [DOI: 10.33549/physiolres.931952] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Peptides ghrelin, obestatin and neuropeptide Y (NPY) play an important role in regulation of energy homeostasis, the imbalance of which is associated with eating disorders anorexia (AN) and bulimia nervosa (BN). The changes in ghrelin, obestatin and NPY plasma levels were investigated in AN and BN patients after administration of a high-carbohydrate breakfast (1604 kJ). Eight AN women (aged 25.4±1.9; BMI: 15.8±0.5), thirteen BN women (aged 22.0±1.05; BMI: 20.1±0.41) and eleven healthy women (aged 25.1±1.16; BMI: 20.9±0.40) were recruited for the study. We demonstrated increased fasting ghrelin in AN, but not in BN patients, while fasting obestatin and NPY were increased in both AN and BN patients compared to the controls. Administration of high-carbohydrate breakfast induced a similar relative decrease in ghrelin and obestatin plasma levels in all groups, while NPY remained increased in postprandial period in both patient groups. Ghrelin/obestatin ratio was lower in AN and BN compared to the controls. In conclusions, increased plasma levels of fasting NPY and its unchanged levels after breakfast indicate that NPY is an important marker of eating disorders AN and BN. Different fasting ghrelin and obestatin levels in AN and BN could demonstrate their diverse functions in appetite and eating suppression.
Collapse
Affiliation(s)
| | | | | | | | | | | | - J. NEDVÍDKOVÁ
- Institute of Endocrinology, Laboratory of Clinical and Experimental Neuroendocrinology, Prague, Czech Republic
| |
Collapse
|
104
|
Avraham Y, Saidian M, Burston JJ, Mevorach R, Vorobiev L, Magen I, Kunkes E, Borges B, Lichtman AH, Berry EM. Fish oil promotes survival and protects against cognitive decline in severely undernourished mice by normalizing satiety signals. J Nutr Biochem 2010; 22:766-76. [PMID: 21109417 DOI: 10.1016/j.jnutbio.2010.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 05/24/2010] [Accepted: 07/22/2010] [Indexed: 12/15/2022]
Abstract
Severe malnutrition resulting from anorexia nervosa or involuntary starvation leads to low weight, cognitive deficits and increased mortality rates. In the present study, we examined whether fish oil supplementation, compared with that of canola oil, would ameliorate the morbidity and mortality associated with these conditions by normalizing endocannabinoid and monoaminergic systems as well as other systems involved in satiety and cognitive function within the hypothalamus and hippocampus. Female Sabra mice restricted to 40% of their daily food intake exhibited decreased body weight, were sickly in appearance, displayed cognitive deficits and had increased mortality rates. Strikingly, fish oil supplementation that contains high omega-3 fatty acids levels decreased mortality and morbidity, and normalized the expression of genes and neurotransmitters in the hippocampus and hypothalamus. Fish oil supplementation, but not canola oil, increased survival rates, improved general appearance and prevented cognitive decline, despite the facts that both diets contained an equivalent number of calories and that there were no differences in weight between mice maintained on the two diets in 100% but decrease in the 40%. In the hypothalamus, the beneficial effects of fish oil supplementation were related to normalization of the endocannabinoid 2-arachidonylglycerol, serotonin (5-HT) (P<.056), dopamine, neuropeptide Y (NPY) and Ca(2+)/calmodulin (CaM)-dependent protein kinase (Camkk2). In the hippocampus, fish oil supplementation normalized 5-HT, Camkk2, silent mating type information regulation 1 and brain-derived neurotrophic factor. In conclusion, dietary supplements of fish oil, as source of omega-3 fatty acids, may alleviate cognitive impairments associated with severe diet restriction and prolong survival independently of weight gain by normalizing neurochemical systems.
Collapse
Affiliation(s)
- Yosefa Avraham
- Department of Human Nutrition and Metabolism and School of Public Health, Hebrew University Hadassah Medical School, Jerusalem 91120, Israel.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
105
|
Abe K, Kuo L, Zukowska Z. Neuropeptide Y is a mediator of chronic vascular and metabolic maladaptations to stress and hypernutrition. Exp Biol Med (Maywood) 2010; 235:1179-84. [PMID: 20881322 DOI: 10.1258/ebm.2010.009136] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Neuropeptide Y (NPY) is a central neuromodulator and peripheral sympathetic neurotransmitter that also has important regulatory roles in cardiovascular, neuroendocrine, immune and metabolic functions during stress. Focusing on the peripheral actions of the peptide in rodent models, we summarize recent studies from our laboratory demonstrating that stress-induced release of NPY mediates accelerated atherosclerosis/restenosis, obesity and metabolic-like syndrome, particularly when combined with a high fat, high sugar diet. In this review, we propose mechanisms of NPY's actions, its receptors and cellular substrates that increase the risk for cardiovascular and metabolic diseases when chronic stress is associated with pre-existing vascular injury and/or states of hypernutrition.
Collapse
Affiliation(s)
- Ken Abe
- Department of Physiology and Biophysics, Georgetown University Medical Center, 3900 Reservoir Rd NW, Washington, DC 20057, USA
| | | | | |
Collapse
|
106
|
Zhang T, Guan H, Yang K. Keratinocyte growth factor promotes preadipocyte proliferation via an autocrine mechanism. J Cell Biochem 2010; 109:737-46. [PMID: 20069574 DOI: 10.1002/jcb.22452] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Keratinocyte growth factor (KGF; also known as FGF-7) is a well-characterized paracrine growth factor for tissue growth and regeneration. However, its role in adipose tissue, which is known to undergo tremendous expansion in obesity, is virtually unknown. Given that we previously identified KGF as one of the up-regulated growth factors in adipose tissue of an early-life programmed rat model of visceral obesity, the present study was undertaken to examine the hypothesis that KGF promotes adipogenesis. Using 3T3-L1 and rat primary preadipocytes as in vitro model systems, we demonstrated that (1) KGF stimulated preadipocyte proliferation in a concentration-dependent manner with a maximal effect at 2.5 ng/ml (approximately 2-fold increase); (2) KGF mRNA was highly expressed in rat adipocytes and preadipocytes as well as 3T3-L1 cells; (3) treatment of preadipocytes with a neutralizing antibody against KGF and siRNA-mediated knockdown of KGF led to a 50% reduction in their proliferative capacity; (4) KGF activated the protein kinase Akt, and the PI3 kinase inhibitor LY294002 blocked KGF stimulation of preadipocyte proliferation; and (5) KGF did not promote differentiation of preadipocytes to mature adipocytes. Together, these results reveal adipocytes and their precursor cells as novel sites of KGF production. Importantly, they also demonstrate that KGF promotes preadipocyte proliferation by an autocrine mechanism that involves activation of the PI3K/Akt signaling pathway. Aberrant KGF expression may have consequences not only for normal adipose tissue growth but also for the pathogenesis of obesity.
Collapse
Affiliation(s)
- Ting Zhang
- Children's Health Research Institute & Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada
| | | | | |
Collapse
|
107
|
Lee NJ, Doyle KL, Sainsbury A, Enriquez RF, Hort YJ, Riepler SJ, Baldock PA, Herzog H. Critical role for Y1 receptors in mesenchymal progenitor cell differentiation and osteoblast activity. J Bone Miner Res 2010; 25:1736-47. [PMID: 20200977 DOI: 10.1002/jbmr.61] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The neuropeptide Y (NPY) system has been implicated in the regulation of bone homeostasis and osteoblast activity, but the mechanism behind this is unclear. Here we show that Y1 receptor signaling is directly involved in the differentiation of mesenchymal progenitor cells isolated from bone tissue, as well as the activity of mature osteoblasts. Importantly, the mRNA levels of two key osteogenic transcription factors, runx2 and osterix, as well as the adipogenic transcription factor PPAR-gamma, were increased in long bones of Y1(-/-) mice compared with wild-type mice. In vitro, bone marrow stromal cells (BMSCs) isolated from Y1(-/-) mice formed a greater number of mineralized nodules under osteogenic conditions and a greater number of adipocytes under adipogenic conditions than controls. In addition, both the number and size of fibroblast colony-forming units formed in vitro by purified osteoprogenitor cells were increased in the absence of the Y1 receptors, suggestive of enhanced proliferation and osteogenesis. Furthermore, the ability of two specific populations of mesenchymal progenitor cells isolated from bone tissue, an immature mesenchymal stem cell population and a more committed osteoprogenitor cell population, to differentiate into osteoblasts and adipocytes in vitro was enhanced in the absence of Y1 receptor signaling. Finally, Y1 receptor deletion also enhanced the mineral-producing ability of mature osteoblasts, as shown by increased in vitro mineralization by BMSCs isolated from osteoblast-specific Y1(-/-) mice. Together these data demonstrate that the NPY system, via the Y1 receptor, directly inhibits the differentiation of mesenchymal progenitor cells as well as the activity of mature osteoblasts, constituting a likely mechanism for the high-bone-mass phenotype evident in Y1(-/-) mice.
Collapse
Affiliation(s)
- Nicola J Lee
- Neuroscience Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | |
Collapse
|
108
|
Zengin A, Zhang L, Herzog H, Baldock PA, Sainsbury A. Neuropeptide Y and sex hormone interactions in humoral and neuronal regulation of bone and fat. Trends Endocrinol Metab 2010; 21:411-8. [PMID: 20202858 DOI: 10.1016/j.tem.2010.02.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Revised: 02/09/2010] [Accepted: 02/09/2010] [Indexed: 02/03/2023]
Abstract
The hypothalamus regulates the skeleton and adipose tissue via endocrine mechanisms. Changes in sex steroid levels in menopause and aging are central to the associated changes in bone mass and adiposity. Whereas many of these effects occur via direct actions on osteoblasts or adipocytes, sex hormones can also mediate effects on bone and adipose tissue via interaction with neuronal pathways. A key hypothalamic regulator of bone and adipose tissue is neuropeptide Y (NPY), which coordinately influences these tissues via effects on neuroendocrine and sympathetic nervous output. Better understanding of the interaction between NPY and sex steroids in regulating skeletal and energy homeostasis could lead to more effective treatments for osteoporosis and obesity.
Collapse
Affiliation(s)
- Ayse Zengin
- Osteoporosis and Bone Biology Program, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst, Sydney, New South Wales (NSW), Australia
| | | | | | | | | |
Collapse
|
109
|
Delahaye F, Lukaszewski MA, Wattez JS, Cisse O, Dutriez-Casteloot I, Fajardy I, Montel V, Dickes-Coopman A, Laborie C, Lesage J, Breton C, Vieau D. Maternal perinatal undernutrition programs a “brown-like” phenotype of gonadal white fat in male rat at weaning. Am J Physiol Regul Integr Comp Physiol 2010; 299:R101-10. [DOI: 10.1152/ajpregu.00604.2009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Several studies indicate that maternal undernutrition sensitizes the offspring to the development of metabolic disorders, such as obesity. Using a model of perinatal maternal 50% food-restricted diet (FR50), we recently reported that rat neonates from undernourished mothers exhibit decreased leptin plasma levels associated with alterations of hypothalamic proopiomelanocortin system. The present study aimed at examining the consequences of FR50 on the brain-adipose axis in male rat neonates. Using quantitative RT-PCR array containing 84 obesity-related genes, we demonstrated that most of the genes involved in energy metabolism regulation are expressed in rat gonadal white adipose tissue (WAT) and are sensitive to maternal perinatal undernutrition (MPU). In contrast, hypothalamic gene expression was not substantially affected by MPU. Gene expression of uncoupling protein 1 (UCP1), a marker of brown adipocytes, showed an almost 400-fold stimulation in postnatal day 21 (PND21) FR50 animals, suggesting that their gonadal WAT possesses a brown-like phenotype. This was confirmed by histological and immunoshistochemical procedures, which demonstrated that PND21 FR50 gonadal adipocytes are multilocular, resembling those present in interscapular brown adipose tissue, and exhibit an overexpression of UCP1 and neuropeptide Y (NPY) at the protein level. Control animals contained almost exclusively “classical” unilocular white adipocytes that did not show high UCP1 and NPY labeling. After weaning, FR50 animals exhibited a transient hyperphagia that was associated with the disappearance of brown-like fat pads in PND30 WAT. Our results demonstrate that MPU delays the maturation of gonadal WAT during critical developmental time windows, suggesting that it could have long-term consequences on body weight regulation in the offspring.
Collapse
Affiliation(s)
- Fabien Delahaye
- Unité Environnement Périnatal et Croissance EA 4489, Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille 1, Villeneuve d'Ascq, France
| | - Marie-Amélie Lukaszewski
- Unité Environnement Périnatal et Croissance EA 4489, Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille 1, Villeneuve d'Ascq, France
| | - Jean-Sébastien Wattez
- Unité Environnement Périnatal et Croissance EA 4489, Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille 1, Villeneuve d'Ascq, France
| | - Ouma Cisse
- Unité Environnement Périnatal et Croissance EA 4489, Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille 1, Villeneuve d'Ascq, France
| | - Isabelle Dutriez-Casteloot
- Unité Environnement Périnatal et Croissance EA 4489, Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille 1, Villeneuve d'Ascq, France
| | - Isabelle Fajardy
- Unité Environnement Périnatal et Croissance EA 4489, Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille 1, Villeneuve d'Ascq, France
| | - Valérie Montel
- Unité Environnement Périnatal et Croissance EA 4489, Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille 1, Villeneuve d'Ascq, France
| | - Anne Dickes-Coopman
- Unité Environnement Périnatal et Croissance EA 4489, Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille 1, Villeneuve d'Ascq, France
| | - Christine Laborie
- Unité Environnement Périnatal et Croissance EA 4489, Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille 1, Villeneuve d'Ascq, France
| | - Jean Lesage
- Unité Environnement Périnatal et Croissance EA 4489, Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille 1, Villeneuve d'Ascq, France
| | - Christophe Breton
- Unité Environnement Périnatal et Croissance EA 4489, Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille 1, Villeneuve d'Ascq, France
| | - Didier Vieau
- Unité Environnement Périnatal et Croissance EA 4489, Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille 1, Villeneuve d'Ascq, France
| |
Collapse
|
110
|
Mulumba M, Jossart C, Granata R, Gallo D, Escher E, Ghigo E, Servant MJ, Marleau S, Ong H. GPR103b functions in the peripheral regulation of adipogenesis. Mol Endocrinol 2010; 24:1615-25. [PMID: 20534693 DOI: 10.1210/me.2010-0010] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The activation of G protein-coupled receptor 103 (GPR103) by its endogenous peptidic ligands, QRFPs, is involved in the central regulation of feeding by increasing food intake, body weight, and fat mass after intracerebroventricular injection in mice. However, the role of GPR103 in regulating peripheral metabolic pathways has not yet been explored. The present study aimed to investigate the role of GPR103 in adipogenesis and lipid metabolism using 3T3-L1 adipocyte cells. Our results show that differentiated 3T3-L1 cells expressed the GPR103b subtype mRNA and protein, as well as QRFP mRNA. QRFP-43 and -26 induced an increase in triglyceride accumulation of 50 and 41%, respectively, and elicited a dose-dependent increase in fatty acid uptake, by up to approximately 60% at the highest concentration, in 3T3-L1-differentiated cells. QRFP-43 and -26 inhibited isoproterenol (ISO)-induced lipolysis in a dose-dependent manner, with IC(50)s of 2.3 +/- 1.2 and 1.1 +/- 1.0 nm, respectively. The expression of genes involved in lipid uptake (FATP1, CD36, LPL, ACSL1, PPAR-gamma, and C/EBP-alpha), was increased by 2- to 3-fold after treatment with QRFP. The effects of QRFP on ISO-induced lipolysis and fatty acid uptake were abolished when GPR103b was silenced. In a mouse model of diet-induced obesity, the expression of GPR103b in epididymal fat pads was elevated by 16-fold whereas that of QRFP was reduced by 46% compared to lean mice. Furthermore, QRFP was bioactive in omental adipocytes from obese individuals, inhibiting ISO-induced lipolysis in these cells. Our results suggest that GPR103b and QRFP work in an autocrine/paracrine manner to regulate adipogenesis.
Collapse
Affiliation(s)
- Mukandila Mulumba
- Faculty of Pharmacy, Université de Montréal Case Postale 6128, Succursale Centre-Ville, Montréal, Québec, Canada H3C 3J7.
| | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Stewart A, Guan H, Yang K. BMP-3 promotes mesenchymal stem cell proliferation through the TGF-beta/activin signaling pathway. J Cell Physiol 2010; 223:658-66. [PMID: 20143330 DOI: 10.1002/jcp.22064] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Adipogenesis plays a key role in the pathogenesis of obesity. It begins with the commitment of mesenchymal stem cells (MSCs) to the adipocyte lineage, followed by terminal differentiation of preadipocytes to mature adipocytes. A critical, but poorly understood, component of adipogenesis involves proliferation of MSCs and preadipocytes. The present study was undertaken to examine the hypothesis that bone morphogenetic protein-3 (BMP-3) promotes adipogenesis using C3H10T1/2 MSCs and 3T3-L1 preadipocytes as in vitro model systems. We demonstrated that although it did not promote the commitment of MSCs to the adipocyte lineage or the differentiation of preadipocytes to adipocytes, BMP-3-stimulated proliferation by threefold in both cell types. Owing to a lack of information on MSC proliferation, we then delineated the molecular mechanisms underlying BMP-3-stimulated MSC proliferation. We showed that BMP-3 activated the transforming growth factor-beta (TGF-beta)/activin but not ERK1/2, p38 MAPK, or JNK signaling pathways in C3H10T1/2 cells. Furthermore, the TGF-beta/activin receptor kinase inhibitor SB-431542 blocked BMP-3-stimulated proliferation. Importantly, siRNA-mediated knockdown of the key TGF-beta/activin signaling pathway components, ActRIIB, ALK4, or Smad2, abrogated the mitogenic effects of BMP-3 on MSCs. Together, these results demonstrate that BMP-3 stimulates MSC proliferation via the TGF-beta/activin signaling pathway, thus revealing a novel role for this divergent and poorly understood member of the TGF-beta superfamily in regulating MSC proliferation.
Collapse
Affiliation(s)
- Aaron Stewart
- Department of Obstetrics and Gynaecology, The University of Western Ontario, Children's Health Research Institute-Lawson Health Research Institute, London, Ontario, Canada
| | | | | |
Collapse
|
112
|
Abstract
PURPOSE OF REVIEW Regulation of body weight, food intake and appetite is complex and the gastrointestinal tract represents a central organ participating in the regulation of energy homeostasis by signaling to other tissues relevant in this context. This update will provide information regarding recent advances in the understanding of the interaction of gastrointestinal peptides with adipocytes in fat tissue and which biological effects they may exert. RECENT FINDINGS Several gastrointestinal peptides signal to their functional cognate receptors on adipocytes in white adipose tissue (WAT) thereby regulating glucose homeostasis, lipogenesis, lipolysis, free fatty acid release and may also participate in adipocyte differentiation. SUMMARY Gastrointestinal peptides emanate from enteroendocrine cells in the luminal digestive tract and are critical regulators of energy homeostasis, food intake and appetite. Recent studies have identified that gastrointestinal peptides communicate with WAT and exert their biological effects on fat cells. Fundamental understanding of gastrointestinal peptides and their interaction with adipocytes will provide future insights for the development of pharmacological targets in the treatment of obesity and insulin resistant states.
Collapse
Affiliation(s)
- Ishita D Majumdar
- Section of Gastroenterology, Boston University School of Medicine, Boston, Massachusetts 02118-2518, USA
| | | |
Collapse
|
113
|
Lee NJ, Herzog H. NPY regulation of bone remodelling. Neuropeptides 2009; 43:457-63. [PMID: 19748118 DOI: 10.1016/j.npep.2009.08.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Revised: 08/13/2009] [Accepted: 08/21/2009] [Indexed: 01/06/2023]
Abstract
Neuropeptide Y (NPY), a classic neuronal regulator of energy homeostasis, is now also known to be involved in the control of bone homeostasis. Of the five known Y receptors through which the NPY family of ligands signals, the Y1 and Y2 receptors have so far been implicated in the control of osteoblast activity and thus bone formation. Analysis of brain specific NPY overexpressing and Y receptor knockout models has revealed a powerful anabolic pathway likely involving hypothalamic Y2 receptors and osteoblastic Y1 receptors. Furthering our understanding of the mechanisms underlying the involvement of the NPY system in the control of bone could lead to the development of therapies to improve bone mass in patients with diseases such as osteoporosis.
Collapse
Affiliation(s)
- Nicola J Lee
- Neuroscience Research Program, Garvan Institute of Medical Research, St. Vincent's Hospital, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
| | | |
Collapse
|
114
|
Igwe JC, Jiang X, Paic F, Ma L, Adams DJ, Baldock PA, Pilbeam CC, Kalajzic I. Neuropeptide Y is expressed by osteocytes and can inhibit osteoblastic activity. J Cell Biochem 2009; 108:621-30. [PMID: 19670271 DOI: 10.1002/jcb.22294] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Osteocytes are the most abundant osteoblast lineage cells within the bone matrix. They respond to mechanical stimulation and can participate in the release of regulatory proteins that can modulate the activity of other bone cells. We hypothesize that neuropeptide Y (NPY), a neurotransmitter with regulatory functions in bone formation, is produced by osteocytes and can affect osteoblast activity. To study the expression of NPY by the osteoblast lineage cells, we utilized transgenic mouse models in which we can identify and isolate populations of osteoblasts and osteocytes. The Col2.3GFP transgene is active in osteoblasts and osteocytes, while the DMP1 promoter drives green fluorescent protein (GFP) expression in osteocytes. Real-time PCR analysis of RNA from the isolated populations of cells derived from neonatal calvaria showed higher NPY mRNA in the preosteocytes/osteocytes fraction compared to osteoblasts. NPY immunostaining confirmed the strong expression of NPY in osteocytes (DMP1GFP(+)), and lower levels in osteoblasts. In addition, the presence of NPY receptor Y1 mRNA was detected in cavaria and long bone, as well as in primary calvarial osteoblast cultures, whereas Y2 mRNA was restricted to the brain. Furthermore, NPY expression was reduced by 30-40% in primary calvarial cultures when subjected to fluid shear stress. In addition, treatment of mouse calvarial osteoblasts with exogenous NPY showed a reduction in the levels of intracellular cAMP and markers of osteoblast differentiation (osteocalcin, BSP, and DMP1). These results highlight the potential regulation of osteoblast lineage differentiation by local NPY signaling.
Collapse
Affiliation(s)
- John C Igwe
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut 06032, USA
| | | | | | | | | | | | | | | |
Collapse
|
115
|
Watson E, Fargali S, Okamoto H, Sadahiro M, Gordon RE, Chakraborty T, Sleeman MW, Salton SR. Analysis of knockout mice suggests a role for VGF in the control of fat storage and energy expenditure. BMC PHYSIOLOGY 2009; 9:19. [PMID: 19863797 PMCID: PMC2774661 DOI: 10.1186/1472-6793-9-19] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Accepted: 10/28/2009] [Indexed: 11/29/2022]
Abstract
Background Previous studies of mixed background mice have demonstrated that targeted deletion of Vgf produces a lean, hypermetabolic mouse that is resistant to diet-, lesion-, and genetically-induced obesity. To investigate potential mechanism(s) and site(s) of action of VGF, a neuronal and endocrine secreted protein and neuropeptide precursor, we further analyzed the metabolic phenotypes of two independent VGF knockout lines on C57Bl6 backgrounds. Results Unlike hyperactive VGF knockout mice on a mixed C57Bl6-129/SvJ background, homozygous mutant mice on a C57Bl6 background were hypermetabolic with similar locomotor activity levels to Vgf+/Vgf+ mice, during day and night cycles, indicating that mechanism(s) other than hyperactivity were responsible for their increased energy expenditure. In Vgf-/Vgf- knockout mice, morphological analysis of brown and white adipose tissues (BAT and WAT) indicated decreased fat storage in both tissues, and decreased adipocyte perimeter and area in WAT. Changes in gene expression measured by real-time RT-PCR were consistent with increased fatty acid oxidation and uptake in BAT, and increased lipolysis, decreased lipogenesis, and brown adipocyte differentiation in WAT, suggesting that increased sympathetic nervous system activity in Vgf-/Vgf- mice may be associated with or responsible for alterations in energy expenditure and fat storage. In addition, uncoupling protein 1 (UCP1) and UCP2 protein levels, mitochondrial number, and mitochondrial cristae density were upregulated in Vgf-/Vgf- BAT. Using immunohistochemical and histochemical techniques, we detected VGF in nerve fibers innervating BAT and Vgf promoter-driven reporter expression in cervical and thoracic spinal ganglia that project to and innervate the chest wall and tissues including BAT. Moreover, VGF peptide levels were quantified by radioimmunoassay in BAT, and were found to be down-regulated by a high fat diet. Lastly, despite being hypermetabolic, VGF knockout mice were cold intolerant. Conclusion We propose that VGF and/or VGF-derived peptides modulate sympathetic outflow pathways to regulate fat storage and energy expenditure.
Collapse
Affiliation(s)
- Elizabeth Watson
- Department of Neuroscience, Mount Sinai School of Medicine, New York, NY, USA.
| | | | | | | | | | | | | | | |
Collapse
|
116
|
Karalis KP, Giannogonas P, Kodela E, Koutmani Y, Zoumakis M, Teli T. Mechanisms of obesity and related pathology: linking immune responses to metabolic stress. FEBS J 2009; 276:5747-54. [PMID: 19754872 DOI: 10.1111/j.1742-4658.2009.07304.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
There is a tightly regulated interaction, which is well-conserved in evolution, between the metabolic and immune systems that is deranged in states of over- or under-nutrition. Obesity, an energy-rich condition, is characterized by the activation of an inflammatory process in metabolically active sites such as adipose tissue, liver and immune cells. The consequence of this response is a sharp increase in circulating levels of proinflammatory cytokines, adipokines and other inflammatory markers. Activation of the immune response in obesity is mediated by specific signaling pathways, with Jun N-terminal kinase and IkappaB kinase beta/nuclear factor kappa-light-chain-enhancer of activated B cells being the most well studied. It is known that the above events modify insulin signaling and result in the development of insulin resistance. The nutrient overload characterizing obesity is a metabolic stressor associated with intracellular organelle (e.g. the endoplasmic reticulum) stress. The exact characterization of the series of events and the mechanisms that integrate the inflammatory response with metabolic homeostasis at the cellular and systemic level is a very active research field. In this minireview, we discuss the signaling pathways and molecules associated with the development of obesity-induced inflammation, as well as the evidence that supports a critical role for the stress response in this process.
Collapse
Affiliation(s)
- Katia P Karalis
- Biomedical Research Foundation of the Academy of Athens (BRFAA), Greece.
| | | | | | | | | | | |
Collapse
|
117
|
Sato N, Ogino Y, Mashiko S, Ando M. Modulation of neuropeptide Y receptors for the treatment of obesity. Expert Opin Ther Pat 2009; 19:1401-15. [DOI: 10.1517/13543770903251722] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
118
|
Sharma A, Guan H, Yang K. The p38 mitogen-activated protein kinase regulates 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2) expression in human trophoblast cells through modulation of 11beta-HSD2 messenger ribonucleic acid stability. Endocrinology 2009; 150:4278-86. [PMID: 19497972 DOI: 10.1210/en.2009-0479] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The placental 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2; encoded by the HSD11B2 gene) has emerged as a key player in controlling fetal development, but its regulation is incompletely understood. Here we identified p38 MAPK as an important regulator of placental 11beta-HSD2. We showed that inhibition of p38 MAPK with the pharmacological inhibitor SB202190 led to an approximately 50% reduction in 11beta-HSD2 activity, protein, and mRNA in primary human placental trophoblast cells. Furthermore, the effect of SB202190 was confirmed by the use of two additional p38 inhibitors, SB203580 and SB220025. In addition, SB202190 decreased the half-life of 11beta-HSD2 mRNA without altering the HSD11B2 promoter activity, indicating that p38 MAPK regulates placental 11beta-HSD2 expression through modulation of 11beta-HSD2 mRNA stability. Importantly, small interfering RNA-mediated knockdown of p38alpha caused a 50% reduction in 11beta-HSD2 activity, suggesting that p38alpha is the primary p38 isoform involved. Taken together, these findings suggest a novel pathway controlling placental 11beta-HSD2 expression resulting from the activation of p38 MAPK. Given that p38alpha is abundantly expressed in the human placenta in which its function is largely unknown, our present study also reveals 11beta-HSD2 as an important target through which p38alpha may regulate human placental function and consequently fetal growth and development.
Collapse
Affiliation(s)
- Anju Sharma
- Department of Obstetrics and Gynaecology, Children's Health Research Institute and Lawson Health Research Institute, The University of Western Ontario, London, Ontario, Canada
| | | | | |
Collapse
|
119
|
Shiraev T, Chen H, Morris MJ. Differential effects of restricted versus unlimited high-fat feeding in rats on fat mass, plasma hormones and brain appetite regulators. J Neuroendocrinol 2009; 21:602-9. [PMID: 19490368 DOI: 10.1111/j.1365-2826.2009.01877.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The rapid rise in obesity has been linked to altered food consumption patterns. There is increasing evidence that, in addition to total energy intake, the macronutrient composition of the diet may influence the development of obesity. The present study aimed to examine the impact of high dietary fat content, under both isocaloric and hypercaloric conditions, compared with a low fat diet, on adiposity, glucose and lipid metabolism, and brain appetite regulators in rats. Male Sprague-Dawley rats were exposed to one of three diets: control (14% fat), ad lib high-fat palatable (HFD, 35% fat) or high-fat palatable restricted (HFD-R, matched to the energy intake of control) and were killed in the fasting state 11 weeks later. Body weight was increased by 28% in unrestricted HFD fed rats, with an almost tripling of caloric intake and fat mass (P < 0.001) and double the plasma triglycerides of controls. Glucose intolerance and increased insulin levels were observed. HFD-R animals calorie matched to control had double their fat mass, plasma insulin and triglycerides (P < 0.05). Only ad lib consumption of the HFD increased the hypothalamic mRNA expression of the appetite-regulating peptides, neuropeptide Y and pro-opiomelanocortin. Although restricted consumption of palatable HFD had no significant impact on hypothalamic appetite regulators or body weight, it increased adiposity and circulating triglycerides, suggesting that the proportion of dietary fat, independent of caloric intake, affects fat deposition and the metabolic profile.
Collapse
Affiliation(s)
- T Shiraev
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | | | | |
Collapse
|
120
|
Synergistic interaction between neuropeptide Y1 and Y5 receptor pathways in regulation of energy homeostasis. Eur J Pharmacol 2009; 615:113-7. [PMID: 19482021 DOI: 10.1016/j.ejphar.2009.05.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Revised: 05/14/2009] [Accepted: 05/19/2009] [Indexed: 11/22/2022]
Abstract
Neuropeptide Y plays a key role in the physiological control of energy homeostasis. Five neuropeptide Y receptor subtypes have been cloned, and multiple neuropeptide Y receptor subtypes are thought to mediate neuropeptide Y activity. However, interactions among neuropeptide Y receptor subtypes have not been elucidated to date. Herein, we examined the interaction between neuropeptide Y(1) and Y(5) receptors in feeding regulation by employing selective neuropeptide Y(1) and Y(5) receptor antagonists in C57BL/6 and neuropeptide Y(1) receptor knockout mice fed a high-fat diet. A single-dose of a neuropeptide Y(1) receptor antagonist (10-30 mg/kg) suppressed spontaneous food intake and reduced body weight in high-fat diet-fed C57BL/6 mice, while treatment with a neuropeptide Y(5) receptor antagonist did not significantly reduce food intake or body weight. Coadministration of a neuropeptide Y(1) receptor antagonist with a neuropeptide Y(5) receptor antagonist further suppressed food intake and reduced body weight. Next, we evaluated the chronic efficacy of a neuropeptide Y(5) receptor antagonist in high-fat diet-fed neuropeptide Y(1) receptor knockout mice in order to mimic chronic combination treatment with neuropeptide Y(1) and Y(5) receptor antagonists. The neuropeptide Y(5) receptor antagonist produced greater body weight reductions in high-fat diet-fed neuropeptide Y(1) receptor knockout mice than in wild-type C57BL/6 mice. These findings confirm an interaction between neuropeptide Y(1) and Y(5) receptors in the regulation of energy homeostasis, as blockade of both the neuropeptide Y(1) and Y(5) receptors produced a greater anti-obesity effect than blocking either receptor alone.
Collapse
|
121
|
Gericke MT, Kosacka J, Koch D, Nowicki M, Schröder T, Ricken AM, Nieber K, Spanel-Borowski K. Receptors for NPY and PACAP differ in expression and activity during adipogenesis in the murine 3T3-L1 fibroblast cell line. Br J Pharmacol 2009; 157:620-32. [PMID: 19422400 DOI: 10.1111/j.1476-5381.2009.00164.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Neuropeptides are involved in the regulation of food intake in the central nervous system, but they might also act on peripheral fat tissue via neuropeptide receptors. EXPERIMENTAL APPROACH We investigated the receptor expression and activity of pituitary adenylate cyclase-activating polypeptide (PACAP) and of neuropeptide Y at the mRNA and protein levels in the 3T3-L1 fibroblast line during differentiation into adipocytes. Intracellular calcium concentration was measured by calcium imaging. KEY RESULTS The PACAP receptors PAC(1) and VPAC(2) as well as the neuropeptide Y(1) receptor were expressed at the mRNA level in fibroblasts, pre-adipocytes and adipocytes. The mRNA profile of the PAC(1) receptor isoforms showed the HOP sequence, whereas the HIP-isoform was present in subconfluent 3T3-L1 fibroblasts only. At the protein level, the mature 3T3-L1 adipocytes produced the PAC(1) and Y(1) receptors; only the PAC(1) receptor showed carbohydrate residues. Both neuropeptides induced an increase of intracellular calcium in mature adipocytes, which was absent in the precursor cells. These changes in calcium were mediated by Y(1) and PAC(1) receptors as demonstrated by the effects of specific receptor agonists and antagonists. CONCLUSIONS AND IMPLICATIONS As the PAC(1)-HOP receptor variant seems to be responsible for PACAP-mediated calcium influx in many cell types, the HOP sequence might also mediate the increase in intracellular calcium in adipocytes. Because a high intracellular calcium level is associated with lipogenesis, peptidergic innervation of adipose tissue might be involved in stress-induced obesity.
Collapse
|
122
|
Abstract
Obesity prevalence is generally higher in women than in men, and there is also a sex difference in body fat distribution. Sex differences in obesity can be explained in part by the influence of gonadal steroids on body composition and appetite; however, behavioural, socio-cultural and chromosomal factors may also play a role. This review, which evolved from the 2008 Stock Conference on sex differences in obesity, summarizes current research and recommendations related to hormonal and neuroendocrine influences on energy balance and fat distribution. A number of important gaps in the research are identified, including a need for more studies on chromosomal sex effects on energy balance, the role of socio-cultural (i.e. gender) factors in obesity and the potential deleterious effects of high-fat diets during pregnancy on the foetus. Furthermore, there is a paucity of clinical trials examining sex-specific approaches and outcomes of obesity treatment (lifestyle-based or pharmacological), and research is urgently needed to determine whether current weight loss programmes, largely developed and tested on women, are appropriate for men. Last, it is important that both animal and clinical research on obesity be designed and analysed in such a way that data can be separately examined in both men and women.
Collapse
Affiliation(s)
- J C Lovejoy
- Free and Clear Inc., 999 Third Avenue, Seattle, WA 98104, USA.
| | | | | |
Collapse
|
123
|
Bahar B, Sweeney T. Mapping of the transcription start site (TSS) and identification of SNPs in the bovine neuropeptide Y (NPY) gene. BMC Genet 2008; 9:91. [PMID: 19105820 PMCID: PMC2657160 DOI: 10.1186/1471-2156-9-91] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Accepted: 12/23/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neuropeptide Y is a key neurotransmitter of the central nervous system which plays a vital role in the feed energy homeostasis in mammals. Mutations in the regulatory and coding regions of the bovine NPY gene can potentially affect the neuronal regulation of appetite and feeding behaviour in cattle. The objectives of this experiment were to: a) fully characterize the bovine NPY gene transcript and b) identify the SNP diversity in both coding and non-coding regions of the NPY gene in a panel of Bos taurus and B. indicus cattle. RESULTS Bovine NPY gene consists of four exons (99, 188, 81 and 195 nucleotides) and three introns. The promoter region of the NPY gene consists of TATA and GC boxes which are separated from the transcription start site (TSS) by 29 and ~100 nt, respectively. Analyses of the tissue specific expression of the bovine NPY gene revealed the presence of highly abundant NPY gene transcripts in the arcuate nucleus, cerebral and cerebellar regions of the bovine brain. We identified a total of 59 SNPs in the 8.4 kb region of the bovine NPY gene. Seven out of nine total SNPs in the promoter region affect binding of putative transcription factors. A high level of nucleotide diversity was evident in the promoter regions (2.84 x 10(-3)) compared to the exonic (1.44 x 10(-3)), intronic (1.30 x 10(-3)) and 3' untranslated (1.26 x 10(-3)) regions. CONCLUSION The SNPs identified in different regions of bovine NPY gene may serve as a basis for understanding the regulation of the expression of the bovine NPY gene under a variety of physiological conditions and identification of genotypes with high feed energy conversion efficiency.
Collapse
Affiliation(s)
- Bojlul Bahar
- Cell and Molecular Biology Lab, School of Agriculture, Food Science & Veterinary Medicine, Veterinary Science Centre, University College Dublin, Belfield, Dublin 4, Ireland.
| | | |
Collapse
|
124
|
Balachandran A, Guan H, Sellan M, van Uum S, Yang K. Insulin and dexamethasone dynamically regulate adipocyte 11beta-hydroxysteroid dehydrogenase type 1. Endocrinology 2008; 149:4069-79. [PMID: 18467433 PMCID: PMC2488250 DOI: 10.1210/en.2008-0088] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The adipocyte enzyme 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) amplifies local glucocorticoid action by generating active glucocorticoids from inactive metabolites and has emerged as a key player in the pathogenesis of central obesity and metabolic syndrome. However, the regulation of adipocyte 11beta-HSD1 is incompletely understood. Therefore, the present study was designed to investigate the effects of insulin and glucocorticoid as well as their underlying molecular mechanisms on 11beta-HSD1 activity and expression in 3T3-L1 adipocytes and determine whether the in vitro findings could be confirmed in vivo. Our main in vitro findings are 1) insulin stimulated whereas dexamethasone inhibited 11beta-HSD1 activity and expression in a time- and concentration-dependent manner; 2) the effect of dexamethasone was mimicked by both cortisol and corticosterone but blocked by the glucocorticoid receptor antagonist RU486; 3) the p38 MAPK inhibitor SB220025, but not the ERK inhibitor U0126 or the phosphatidylinositol 3-kinase inhibitor LY294002, prevented insulin stimulation of 11beta-HSD1 activity; and 4) although dexamethasone did not alter the half-life of 11beta-HSD1 mRNA, insulin doubled it. Taken together, these in vitro results demonstrate that insulin stimulates adipocyte 11beta-HSD1 through a posttranscriptional mechanism that involves activation of the p38 MAPK signaling pathway, whereas dexamethasone exerts an opposite effect by a glucocorticoid receptor-mediated transcriptional mechanism. In contrast, both insulin and dexamethasone augmented 11beta-HSD1 activity and expression in rat white adipose tissue in vivo, thus confirming the role of insulin but revealing a fundamental difference regarding the role of dexamethasone in regulating adipocyte 11beta-HSD1 between the two model systems.
Collapse
Affiliation(s)
- Aran Balachandran
- Children's Health Research Institute and Lawson Health Research Institute, London, Ontario, Canada
| | | | | | | | | |
Collapse
|