151
|
Qu H, Li J, Chen W, Li Y, Jiang Q, Jiang H, Huo J, Zhao Z, Liu B, Zhang Q. Differential expression of the melanocortin-4 receptor in male and female C57BL/6J mice. Mol Biol Rep 2014; 41:3245-56. [DOI: 10.1007/s11033-014-3187-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 01/20/2014] [Indexed: 01/09/2023]
|
152
|
Iwen KA, Scherer T, Heni M, Sayk F, Wellnitz T, Machleidt F, Preissl H, Häring HU, Fritsche A, Lehnert H, Buettner C, Hallschmid M. Intranasal insulin suppresses systemic but not subcutaneous lipolysis in healthy humans. J Clin Endocrinol Metab 2014; 99:E246-51. [PMID: 24423295 PMCID: PMC3913807 DOI: 10.1210/jc.2013-3169] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CONTEXT Insulin infused into the central nervous system of rats suppresses lipolysis in white adipose tissue, indicating a role of brain insulin in regulating systemic lipid metabolism. OBJECTIVE We investigated whether central nervous insulin delivery suppresses lipolysis in healthy humans. DESIGN Placebo-controlled, balanced within-subject comparisons were performed in both a main and an independent corroborative experiment. SETTING/PARTICIPANTS/INTERVENTION: Two groups of healthy volunteers were examined at the German University Clinics of Lübeck and Tübingen, respectively, with molecular analyses taking place at Mt Sinai School of Medicine (New York, New York). The 14 healthy male subjects of the main study and the 22 women and 5 men of the corroborative study each received 160 IU of human insulin intranasally. MAIN OUTCOME MEASURES In the main study, we measured systemic levels of free fatty acids (FFAs), triglycerides, and glycerol and the rate of appearance of deuterated glycerol as an estimate of lipolysis before and after intranasal insulin administration. We also analyzed the expression of key lipolytic enzymes in sc fat biopsies and measured blood glucose and glucoregulatory hormones. In the corroborative study, FFA concentrations were assessed before and after intranasal insulin administration. RESULTS In the main experiment, intranasal insulin suppressed circulating FFA concentrations and lipolysis (rate of appearance of deuterated glycerol) in the absence of significant changes in circulating insulin levels. Lipolytic protein expression in sc adipose tissue was not affected. The corroborative study confirmed that intranasal insulin lowers systemic FFA concentrations. CONCLUSIONS Our findings indicate that brain insulin controls systemic lipolysis in healthy humans by predominantly acting on non-sc adipose tissue.
Collapse
|
153
|
Romero-Zerbo SY, Bermúdez-Silva FJ. Cannabinoids, eating behaviour, and energy homeostasis. Drug Test Anal 2013; 6:52-8. [DOI: 10.1002/dta.1594] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Revised: 11/14/2013] [Accepted: 11/20/2013] [Indexed: 12/14/2022]
Affiliation(s)
- Silvana Y. Romero-Zerbo
- Laboratorio de Investigación; Hospital Regional de Málaga-IBIMA, Plaza del Hospital Civil s/n; 29009 Málaga Spain
- Unidad de Gestion Clínica de Endocrinología y Nutrición, Hospital Civil, Pabellón 1, sótano; 29009 Málaga Spain
| | - Francisco J. Bermúdez-Silva
- Laboratorio de Investigación; Hospital Regional de Málaga-IBIMA, Plaza del Hospital Civil s/n; 29009 Málaga Spain
- Unidad de Gestion Clínica de Endocrinología y Nutrición, Hospital Civil, Pabellón 1, sótano; 29009 Málaga Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM); Málaga Spain
| |
Collapse
|
154
|
Filippi BM, Abraham MA, Yue JTY, Lam TKT. Insulin and glucagon signaling in the central nervous system. Rev Endocr Metab Disord 2013; 14:365-75. [PMID: 23959343 DOI: 10.1007/s11154-013-9258-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The prevalence of the obesity and diabetes epidemic has triggered tremendous research investigating the role of the central nervous system (CNS) in the regulation of food intake, body weight gain and glucose homeostasis. This invited review focuses on the role of two pancreatic hormones--insulin and glucagon--that trigger signaling pathways in the brain to regulate energy and glucose homeostasis. Unlike in the periphery, insulin and glucagon signaling in the CNS does not seem to have opposing metabolic effects, as both hormones exert a suppressive effect on food intake and weight gain. They signal through different pathways and alter different neuronal populations suggesting a complementary action of the two hormones in regulating feeding behavior. Similar to its systemic effect, insulin signaling in the brain lowers glucose production. However, the ability of glucagon signaling in the brain to regulate glucose production remains unknown. Future studies that aim to dissect insulin and glucagon signaling in the CNS that regulate energy and glucose homeostasis could unveil novel signaling molecules to lower body weight and glucose levels in obesity and diabetes.
Collapse
|
155
|
Endurance training increases leptin expression in the retroperitoneal adipose tissue of rats fed with a high-sugar diet. Lipids 2013; 49:85-96. [PMID: 24243000 PMCID: PMC3889676 DOI: 10.1007/s11745-013-3854-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 10/10/2013] [Indexed: 12/16/2022]
Abstract
The presence of leptin receptors in white adipose tissue (WAT) suggests a type of peripheral control during the development of obesity and other metabolic disorders. Both diet composition and exercise influence serum leptin; however, the effect of their combination on long-term WAT leptin metabolism is unknown. In this study, rats fed with standard or high-sugar diets (HSD) were simultaneously subjected to running training for 4- and 8-week periods, and the retroperitoneal WAT (rWAT) was evaluated for adipocyte cell size, lipid and catecholamine content, Lep, OB-Rb and Ucp2 mRNA transcription levels, and circulating leptin and non-esterified fatty acids (NEFA). The HSD groups displayed a higher adiposity index and rWAT weight, Lep mRNA and protein upregulation, and a period-dependent effect on OB-Rb mRNA expression. Exercise decreased serum leptin and upregulated the OB-Rb mRNA levels. However, in rats fed with an HSD, the increase in OB-Rb mRNA and reduction in catecholamine levels resulted in a high level of adiposity and hyperleptinemia. The combination of training and an HSD decreases the NEFA levels and upregulating the Ucp2 mRNA expression in the 4-week period, while downregulating the Ucp2 mRNA expression in the 8-week period without changing the NEFA levels. Our results suggest that an HSD induces an increase in leptin expression in rWAT, while reducing adipocytes via leptin-mediated lipolysis after an 8-week period. In exercised rats fed an HSD, TAG synthesis and storage overlaps with lipolysis, promoting fat store development and Lep mRNA and plasma protein upregulation in adult rats.
Collapse
|
156
|
Schneider JE, Wise JD, Benton NA, Brozek JM, Keen-Rhinehart E. When do we eat? Ingestive behavior, survival, and reproductive success. Horm Behav 2013; 64:702-28. [PMID: 23911282 DOI: 10.1016/j.yhbeh.2013.07.005] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 07/21/2013] [Accepted: 07/22/2013] [Indexed: 12/13/2022]
Abstract
The neuroendocrinology of ingestive behavior is a topic central to human health, particularly in light of the prevalence of obesity, eating disorders, and diabetes. The study of food intake in laboratory rats and mice has yielded some useful hypotheses, but there are still many gaps in our knowledge. Ingestive behavior is more complex than the consummatory act of eating, and decisions about when and how much to eat usually take place in the context of potential mating partners, competitors, predators, and environmental fluctuations that are not present in the laboratory. We emphasize appetitive behaviors, actions that bring animals in contact with a goal object, precede consummatory behaviors, and provide a window into motivation. Appetitive ingestive behaviors are under the control of neural circuits and neuropeptide systems that control appetitive sex behaviors and differ from those that control consummatory ingestive behaviors. Decreases in the availability of oxidizable metabolic fuels enhance the stimulatory effects of peripheral hormones on appetitive ingestive behavior and the inhibitory effects on appetitive sex behavior, putting a new twist on the notion of leptin, insulin, and ghrelin "resistance." The ratio of hormone concentrations to the availability of oxidizable metabolic fuels may generate a critical signal that schedules conflicting behaviors, e.g., mate searching vs. foraging, food hoarding vs. courtship, and fat accumulation vs. parental care. In species representing every vertebrate taxa and even in some invertebrates, many putative "satiety" or "hunger" hormones function to schedule ingestive behavior in order to optimize reproductive success in environments where energy availability fluctuates.
Collapse
Affiliation(s)
- Jill E Schneider
- Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem, PA 18015, USA
| | | | | | | | | |
Collapse
|
157
|
Garcia JM, Scherer T, Chen JA, Guillory B, Nassif A, Papusha V, Smiechowska J, Asnicar M, Buettner C, Smith RG. Inhibition of cisplatin-induced lipid catabolism and weight loss by ghrelin in male mice. Endocrinology 2013; 154:3118-29. [PMID: 23832960 PMCID: PMC3749475 DOI: 10.1210/en.2013-1179] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cachexia, defined as an involuntary weight loss ≥ 5%, is a serious and dose-limiting side effect of chemotherapy that decreases survival in cancer patients. Alterations in lipid metabolism are thought to cause the lipodystrophy commonly associated with cachexia. Ghrelin has been proposed to ameliorate the alterations in lipid metabolism due to its orexigenic and anabolic properties. However, the mechanisms of action through which ghrelin could potentially ameliorate chemotherapy-associated cachexia have not been elucidated. The objectives of this study were to identify mechanisms by which the chemotherapeutic agent cisplatin alters lipid metabolism and to establish the role of ghrelin in reversing cachexia. Cisplatin-induced weight and fat loss were prevented by ghrelin. Cisplatin increased markers of lipolysis in white adipose tissue (WAT) and of β-oxidation in liver and WAT and suppressed lipogenesis in liver, WAT, and muscle. Ghrelin prevented the imbalance between lipolysis, β-oxidation, and lipogenesis in WAT and muscle. Pair-feeding experiments demonstrated that the effects of cisplatin and ghrelin on lipogenesis, but not on lipolysis and β-oxidation, were due to a reduction in food intake. Thus, ghrelin prevents cisplatin-induced weight and fat loss by restoring adipose tissue functionality. An increase in caloric intake further enhances the anabolic effects of ghrelin.
Collapse
Affiliation(s)
- Jose M Garcia
- Department of Medicine and Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
158
|
Kola B, Wittman G, Bodnár I, Amin F, Lim CT, Oláh M, Christ-Crain M, Lolli F, van Thuijl H, Leontiou CA, Füzesi T, Dalino P, Isidori AM, Harvey-White J, Kunos G, Nagy GM, Grossman AB, Fekete C, Korbonits M. The CB1 receptor mediates the peripheral effects of ghrelin on AMPK activity but not on growth hormone release. FASEB J 2013; 27:5112-21. [PMID: 23982145 DOI: 10.1096/fj.13-232918] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This study aimed to investigate whether the growth hormone release and metabolic effects of ghrelin on AMPK activity of peripheral tissues are mediated by cannabinoid receptor type 1 (CB1) and the central nervous system. CB1-knockout (KO) and/or wild-type mice were injected peripherally or intracerebroventricularly with ghrelin and CB1 antagonist rimonabant to study tissue AMPK activity and gene expression (transcription factors SREBP1c, transmembrane protein FAS, enzyme PEPCK, and protein HSL). Growth hormone levels were studied both in vivo and in vitro. Peripherally administered ghrelin in liver, heart, and adipose tissue AMPK activity cannot be observed in CB1-KO or CB1 antagonist-treated mice. Intracerebroventricular ghrelin treatment can influence peripheral AMPK activity. This effect is abolished in CB1-KO mice and by intracerebroventricular rimonabant treatment, suggesting that central CB1 receptors also participate in the signaling pathway that mediates the effects of ghrelin on peripheral tissues. Interestingly, in vivo or in vitro growth hormone release is intact in response to ghrelin in CB1-KO animals. Our data suggest that the metabolic effects of ghrelin on AMPK in peripheral tissues are abolished by the lack of functional CB1 receptor via direct peripheral effect and partially through the central nervous system, thus supporting the existence of a possible ghrelin-cannabinoid-CB1-AMPK pathway.
Collapse
Affiliation(s)
- Blerina Kola
- 1Márta Korbonits, Dept. of Endocrinology, Barts and the London School of Medicine and Dentistry, Charterhouse Sq., London EC1M 6BQ, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
159
|
Towards a 'systems'-level understanding of the nervous system and its disorders. Trends Neurosci 2013; 36:674-84. [PMID: 23988221 DOI: 10.1016/j.tins.2013.07.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 07/17/2013] [Accepted: 07/24/2013] [Indexed: 12/26/2022]
Abstract
It is becoming clear that nervous system development and adult functioning are highly coupled with other physiological systems. Accordingly, neurological and psychiatric disorders are increasingly being associated with a range of systemic comorbidities including, most prominently, impairments in immunological and bioenergetic parameters as well as in the gut microbiome. Here, we discuss various aspects of the dynamic crosstalk between these systems that underlies nervous system development, homeostasis, and plasticity. We believe a better definition of this underappreciated systems physiology will yield important insights into how nervous system diseases with systemic comorbidities arise and potentially identify novel diagnostic and therapeutic strategies.
Collapse
|
160
|
Marcelin G, Liu SM, Schwartz GJ, Chua SC. Identification of a loss-of-function mutation in Ube2l6 associated with obesity resistance. Diabetes 2013; 62:2784-95. [PMID: 23557705 PMCID: PMC3717837 DOI: 10.2337/db12-1054] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We previously mapped a locus on BALB/c chromosome 2 associated with protection from leptin-deficiency-induced obesity. Here, we generated the corresponding congenic mouse strain by introgression of a segment of C57BL/6J chromosome 2 to the BALB/c background to confirm the genotype-phenotype associations. We found that the BALB/c alleles decreased fat mass expansion by limiting adipocyte hyperplasia and adipocyte hypertrophy. This was concomitant to an increase in adipocyte triglyceride lipase (ATGL)-mediated triglyceride breakdown and prolongation of ATGL half-life in adipose tissue. In addition, BALB/c alleles on chromosome 2 exerted a cell-autonomous role in restraining the adipogenic potential of preadipocytes. Within a 9.8-Mb critical interval, we identified a nonsynonymous coding single nucleotide polymorphism in the gene coding for the ubiquitin-conjugating enzyme E2L6 (Ube2l6, also known as Ubch8) and showed that the BALB/c allele of Ube2l6 is a hypomorph leading to the lack of UBE2L6 protein expression. Ube2l6 knockdown in 3T3-L1 adipocytes repressed adipogenesis. Thus, altered adipogenic potential caused by Ube2l6 knockdown is likely critically involved in BALB/c obesity resistance by inhibiting adipogenesis and reducing adipocyte numbers. Overall, we have identified a loss-of-function mutation in Ube2l6 that contributes to the chromosome 2 obesity quantitative trait locus.
Collapse
Affiliation(s)
- Genevieve Marcelin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Shun-Mei Liu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Gary J. Schwartz
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| | - Streamson C. Chua
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
- Corresponding author: Streamson C. Chua, Jr.,
| |
Collapse
|
161
|
Harris RBS. Evidence that leptin-induced weight loss requires activation of both forebrain and hindbrain receptors. Physiol Behav 2013; 120:83-92. [PMID: 23911693 DOI: 10.1016/j.physbeh.2013.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/25/2013] [Accepted: 07/22/2013] [Indexed: 01/03/2023]
Abstract
Previous studies with chronic decerebrate rats and rats infused with leptin into the 4th ventricle suggest that hindbrain leptin receptors attenuate the catabolic effect of forebrain leptin receptor activation. To test this further, rats were fitted with both 3rd and 4th ventricle cannulae. They were infused for 12 days with different combinations of saline, low dose leptin or leptin receptor antagonist (leptin mutein protein). Infusion of 0.1 μg leptin/day into the 3rd ventricle or 0.6 μg leptin/day into the 4th ventricle had no significant effect on food intake, energy expenditure or body composition. Infusion of 2 μg mutein/day into either ventricle caused a small, but significant weight gain. When mutein was infused into one ventricle and leptin into the other, the rats lost weight irrespective of which combination was applied. Surprisingly, rats that received leptin infusions into both ventricles showed an initial hypophagia, no change in energy expenditure, but a 75% loss of carcass fat after 12 days. These data suggest that neuronal pathways activated by leptin receptors in either the forebrain or hindbrain modulate each other's effects. In normal conditions hindbrain leptin may attenuate the catabolic effect of forebrain leptin, but if activity in one area is blocked with mutein, then the catabolic response to leptin in the other ventricle is exaggerated. When receptors in both areas are activated there is an integration of response to produce negative energy balance. This may ensure that leptin causes a loss of fat only when leptin is elevated in both the CSF and periphery.
Collapse
Affiliation(s)
- Ruth B S Harris
- Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 15th Street, CA 1020, Augusta, GA 30912, United States.
| |
Collapse
|
162
|
Nasrallah MP, Ziyadeh FN. Overview of the physiology and pathophysiology of leptin with special emphasis on its role in the kidney. Semin Nephrol 2013; 33:54-65. [PMID: 23374894 DOI: 10.1016/j.semnephrol.2012.12.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The adipocyte product leptin is a pleiotropic adipokine and hormone, with a role extending beyond appetite suppression and increased energy expenditure. This review summarizes the biology of the leptin system and the roles of its different receptors in a multitude of cellular functions in different organs, with special emphasis on the kidney. Leptin's physiological functions as well as deleterious effects in states of leptin deficiency or hyperleptinemia are emphasized. Chronic hyperleptinemia can increase blood pressure through the sympathetic nervous system and renal salt retention. The concept of selective leptin resistance in obesity is emerging, whereby leptin's effect on appetite and energy expenditure is blunted, with a concomitant increase in leptin's other effects as a result of the accompanying hyperleptinemia. The divergence in response likely is explained by different receptors and post-receptor activating mechanisms. Chronic kidney disease is a known cause of hyperleptinemia. There is an emerging view that the effect of hyperleptinemia on the kidney can contribute to the development and/or progression of chronic kidney disease in selective resistance states such as in obesity or type 2 diabetes mellitus. The mechanisms of renal injury are likely the result of exaggerated and undesirable hemodynamic influences as well as profibrotic effects.
Collapse
Affiliation(s)
- Mona P Nasrallah
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon.
| | | |
Collapse
|
163
|
Murphy KT, Schwartz GJ, Nguyen NLT, Mendez JM, Ryu V, Bartness TJ. Leptin-sensitive sensory nerves innervate white fat. Am J Physiol Endocrinol Metab 2013; 304:E1338-47. [PMID: 23612999 PMCID: PMC3680695 DOI: 10.1152/ajpendo.00021.2013] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Leptin, the primary white adipose tissue (WAT) adipokine, is thought to convey lipid reserve information to the brain via the circulation. Because WAT responds to environmental/internal signals in a fat pad-specific (FPS) manner, systemic signals such as leptin would fail to communicate such distinctive information. Saturation of brain leptin transport systems also would fail to convey increased lipid levels beyond that point. WAT possesses sensory innervation exemplified by proven sensory-associated peptides in nerves within the tissue and by viral sensory nerve-specific transneuronal tract tracer, H129 strain of herpes simplex virus 1 labeling of dorsal root ganglia (DRG) pseudounipolar neurons, spinal cord and central sensory circuits. Leptin as a paracrine factor activating WAT sensory innervation could supply the brain with FPS information. Therefore, we tested for and found the presence of the long form of the leptin receptor (Ob-Rb) on DRG pseudounipolar neurons immunohistochemically labeled after injections of Fluorogold, a retrograde tract tracer, into inguinal WAT (IWAT). Intra-IWAT leptin injections (300 ng) significantly elevated IWAT nerve spike rate within 5 min and persisted for at least 30 min. Intra-IWAT leptin injections also induced significant c-Fos immunoreactivity (ir), indicating neural activation across DRG pseudounipolar sensory neurons labeled with Fluorogold IWAT injections. Intraperitoneal leptin injection did not increase c-Fos-ir in DRG or the arcuate nucleus, nor did it increase arcuate signal transducer and activator of transcription 3 phosphorylation-ir. Collectively, these results strongly suggest that endogenous leptin secreted from white adipocytes functions as a paracrine factor to activate spinal sensory nerves innervating the tissue.
Collapse
Affiliation(s)
- Keegan T Murphy
- Department of Biology, Obesity Reversal Center, Georgia State University, Atlanta, Georgia; and
| | | | | | | | | | | |
Collapse
|
164
|
Gogga P, Karbowska J, Kochan Z, Meissner W. Circulating leptin levels do not reflect the amount of body fat in the dunlin Calidris alpina during migration. Gen Comp Endocrinol 2013; 187:74-8. [PMID: 23583518 DOI: 10.1016/j.ygcen.2013.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 03/25/2013] [Accepted: 04/03/2013] [Indexed: 10/27/2022]
Abstract
Leptin is a peptide hormone that plays an important role in the regulation of energy homeostasis. Studies in mammals have shown that circulating leptin levels reflect adiposity and that this adipocyte-derived cytokine acts as an afferent satiety signal to the brain, decreasing food intake and increasing energy expenditure. Since leptin has been found in the liver and adipose tissue of migratory birds that are able to accumulate fat reserves as endogenous fuel for flight, we hypothesized that individuals with higher fat score would have higher plasma leptin levels, as it had been found previously in mammals. The aim of this study was to determine if circulating leptin levels correlate with the amount of body fat in a migratory bird, the dunlin Calidris alpina. Adult dunlins were caught during autumn migration on the Baltic coast, and their fat score was determined. Blood samples from 150 birds were used to assess the levels of circulating leptin. We did not find any statistical differences between dunlins with various fat scores. In fact, plasma leptin levels tended to be lower in fat birds than in lean individuals. Our data indicate that in wild birds in migration mode leptin does not reflect the amount of accumulated fat. It suggests that leptin in birds during migration is neither involved in the regulation of energy homeostasis nor acts as a signal to control the amount of body fat.
Collapse
Affiliation(s)
- Patrycja Gogga
- Avian Ecophysiology Unit, Department of Vertebrate Ecology and Zoology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland.
| | | | | | | |
Collapse
|
165
|
Nohara K, Waraich RS, Liu S, Ferron M, Waget A, Meyers MS, Karsenty G, Burcelin R, Mauvais-Jarvis F. Developmental androgen excess programs sympathetic tone and adipose tissue dysfunction and predisposes to a cardiometabolic syndrome in female mice. Am J Physiol Endocrinol Metab 2013; 304:E1321-30. [PMID: 23612996 PMCID: PMC3680697 DOI: 10.1152/ajpendo.00620.2012] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Among women, the polycystic ovarian syndrome (PCOS) is considered a form of metabolic syndrome with reproductive abnormalities. Women with PCOS show increased sympathetic tone, visceral adiposity with enlarged adipocytes, hypoadiponectinemia, insulin resistance, glucose intolerance, increased inactive osteocalcin, and hypertension. Excess fetal exposure to androgens has been hypothesized to play a role in the pathogenesis of PCOS. Previously, we showed that neonatal exposure to the androgen testosterone (NT) programs leptin resistance in adult female mice. Here, we studied the impact of NT on lean and adipose tissues, sympathetic tone in cardiometabolic tissues, and the development of metabolic dysfunction in mice. Neonatally androgenized adult female mice (NTF) displayed masculinization of lean tissues with increased cardiac and skeletal muscle as well as kidney masses. NTF mice showed increased and dysfunctional white adipose tissue with increased sympathetic tone in both visceral and subcutaneous fat as well as increased number of enlarged and insulin-resistant adipocytes that displayed altered expression of developmental genes and hypoadiponectinemia. NTF exhibited dysfunctional brown adipose tissue with increased mass and decreased energy expenditure. They also displayed decreased undercarboxylated and active osteocalcin and were predisposed to obesity during chronic androgen excess. NTF showed increased renal sympathetic tone associated with increased blood pressure, and they developed glucose intolerance and insulin resistance. Thus, developmental exposure to testosterone in female mice programs features of cardiometabolic dysfunction, as can be observed in women with PCOS, including increased sympathetic tone, visceral adiposity, insulin resistance, prediabetes, and hypertension.
Collapse
Affiliation(s)
- Kazunari Nohara
- Division of Endocrinology, Metabolism, and Molecular Medicine, and
| | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Telles MM, da Silva TG, Watanabe RLH, de Andrade IS, Estadella D, Nascimento CMO, Oyama LM, Ribeiro EB. Lateral hypothalamic serotonin is not stimulated during central leptin hypophagia. REGULATORY PEPTIDES 2013; 184:75-80. [PMID: 23518461 DOI: 10.1016/j.regpep.2013.03.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 09/12/2012] [Accepted: 03/03/2013] [Indexed: 10/27/2022]
Abstract
Whether leptin targets the hypothalamic serotonergic system to inhibit food intake is not established. We examined the effect of a short-term i.c.v. leptin treatment on serotonin microdialysate levels in rat lateral hypothalamus. Adipose tissue gene expression was also evaluated. Male rats received four daily injections of leptin (5 μg) or vehicle (with pair-feeding to leptin-induced intake) and a fifth injection during collection of LH microdialysates. We found that serotonin and 5-HIAA levels were not affected by the leptin pre-treatment, as basal levels were similar between the leptin and the pair-fed group. These levels remained unaltered after the acute leptin injection. For gene expression studies, rats were pre-treated with five daily injections of either leptin (5 μg) or vehicle (with either pair-feeding or ad libitum intake). mRNA levels of resistin, adiponectin, lipoprotein lipase, and PPAR-gamma were unaltered by either leptin or pair-feeding. Leptin gene expression was significantly reduced by leptin but not by pair-feeding, in both the retroperitoneal (-74%) and the epididymal (-99%) depots while no differences were observed in the subcutaneous depot. The observations confirmed the absence of an acute stimulatory effect of central leptin on serotonin release in the lateral hypothalamus and showed that the pre-treatment with leptin failed to modify this pattern. This indicates that components of the serotonergic system are probably not directly affected by leptin. Additionally, the central effect of leptin was able to downregulate its own adipose tissue gene expression in a depot-specific manner while other adipokine genes were not affected.
Collapse
Affiliation(s)
- Mônica Marques Telles
- Departamento de Fisiologia, Disciplina de Fisiologia da Nutrição, Universidade Federal de São Paulo (UNIFESP), São Paulo/SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
167
|
Moon HS, Dalamaga M, Kim SY, Polyzos SA, Hamnvik OP, Magkos F, Paruthi J, Mantzoros CS. Leptin's role in lipodystrophic and nonlipodystrophic insulin-resistant and diabetic individuals. Endocr Rev 2013; 34:377-412. [PMID: 23475416 PMCID: PMC3660716 DOI: 10.1210/er.2012-1053] [Citation(s) in RCA: 192] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Leptin is an adipocyte-secreted hormone that has been proposed to regulate energy homeostasis as well as metabolic, reproductive, neuroendocrine, and immune functions. In the context of open-label uncontrolled studies, leptin administration has demonstrated insulin-sensitizing effects in patients with congenital lipodystrophy associated with relative leptin deficiency. Leptin administration has also been shown to decrease central fat mass and improve insulin sensitivity and fasting insulin and glucose levels in HIV-infected patients with highly active antiretroviral therapy (HAART)-induced lipodystrophy, insulin resistance, and leptin deficiency. On the contrary, the effects of leptin treatment in leptin-replete or hyperleptinemic obese individuals with glucose intolerance and diabetes mellitus have been minimal or null, presumably due to leptin tolerance or resistance that impairs leptin action. Similarly, experimental evidence suggests a null or a possibly adverse role of leptin treatment in nonlipodystrophic patients with nonalcoholic fatty liver disease. In this review, we present a description of leptin biology and signaling; we summarize leptin's contribution to glucose metabolism in animals and humans in vitro, ex vivo, and in vivo; and we provide insights into the emerging clinical applications and therapeutic uses of leptin in humans with lipodystrophy and/or diabetes.
Collapse
Affiliation(s)
- Hyun-Seuk Moon
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | |
Collapse
|
168
|
López M, Alvarez CV, Nogueiras R, Diéguez C. Energy balance regulation by thyroid hormones at central level. Trends Mol Med 2013; 19:418-27. [PMID: 23707189 DOI: 10.1016/j.molmed.2013.04.004] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 04/23/2013] [Accepted: 04/25/2013] [Indexed: 12/21/2022]
Abstract
Classically, medical textbooks taught that most effects of thyroid hormones (THs) on energy homeostasis are directly exerted in peripheral tissues. However, current evidence is changing (and challenging) our perspective about the role of THs from a 'peripheral' to a 'central' vision, implying that they affect food intake, energy expenditure, and metabolism by acting, to a large extent, at the central level. Interestingly, effects of THs are interrelated with global energy sensors in the central nervous system (CNS), such as uncoupling protein 2 (UCP2), AMP-activated protein kinase (AMPK; the 'AMPK-BAT axis'), and mechanistic target of rapamycin (mTOR). Here, we review what is currently known about THs and their regulation of energy balance and metabolism in both peripheral and central tissues.
Collapse
Affiliation(s)
- Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain.
| | | | | | | |
Collapse
|
169
|
Harris RBS. Direct and indirect effects of leptin on adipocyte metabolism. Biochim Biophys Acta Mol Basis Dis 2013; 1842:414-23. [PMID: 23685313 DOI: 10.1016/j.bbadis.2013.05.009] [Citation(s) in RCA: 201] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 04/18/2013] [Accepted: 05/06/2013] [Indexed: 12/22/2022]
Abstract
Leptin is hypothesized to function as a negative feedback signal in the regulation of energy balance. It is produced primarily by adipose tissue and circulating concentrations correlate with the size of body fat stores. Administration of exogenous leptin to normal weight, leptin responsive animals inhibits food intake and reduces the size of body fat stores whereas mice that are deficient in either leptin or functional leptin receptors are hyperphagic and obese, consistent with a role for leptin in the control of body weight. This review discusses the effect of leptin on adipocyte metabolism. Because adipocytes express leptin receptors there is the potential for leptin to influence adipocyte metabolism directly. Adipocytes also are insulin responsive and receive sympathetic innervation, therefore leptin can also modify adipocyte metabolism indirectly. Studies published to date suggest that direct activation of adipocyte leptin receptors has little effect on cell metabolism in vivo, but that leptin modifies adipocyte sensitivity to insulin to inhibit lipid accumulation. In vivo administration of leptin leads to a suppression of lipogenesis, an increase in triglyceride hydrolysis and an increase in fatty acid and glucose oxidation. Activation of central leptin receptors also contributes to the development of a catabolic state in adipocytes, but this may vary between different fat depots. Leptin reduces the size of white fat depots by inhibiting cell proliferation both through induction of inhibitory circulating factors and by contributing to sympathetic tone which suppresses adipocyte proliferation. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.
Collapse
Affiliation(s)
- Ruth B S Harris
- Department of Physiology, Medical College of Georgia, Georgia Regents University, USA.
| |
Collapse
|
170
|
Abstract
Endocannabinoids and cannabinoid CB1 receptors are known to play a generalized role in energy homeostasis. However, clinical trials with the first generation of CB1 blockers, now discontinued due to psychiatric side effects, were originally designed to reduce food intake and body weight rather than the metabolic risk factors associated with obesity. In this review, we discuss how, in addition to promoting energy intake, endocannabinoids control lipid and glucose metabolism in several peripheral organs, particularly the liver and adipose tissue. Direct actions in skeletal muscle and pancreas are also emerging. This knowledge may help in the design of future therapies for the metabolic syndrome.
Collapse
|
171
|
Mul JD, O’Duibhir E, Shrestha YB, Koppen A, Vargoviç P, Toonen PW, Zarebidaki E, Kvetnansky R, Kalkhoven E, Cuppen E, Bartness TJ. Pmch-deficiency in rats is associated with normal adipocyte differentiation and lower sympathetic adipose drive. PLoS One 2013; 8:e60214. [PMID: 23555928 PMCID: PMC3608591 DOI: 10.1371/journal.pone.0060214] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 02/22/2013] [Indexed: 02/01/2023] Open
Abstract
The orexigenic neuropeptide melanin-concentrating hormone (MCH), a product of Pmch, is an important mediator of energy homeostasis. Pmch-deficient rodents are lean and smaller, characterized by lower food intake, body-, and fat mass. Pmch is expressed in hypothalamic neurons that ultimately are components in the sympathetic nervous system (SNS) drive to white and interscapular brown adipose tissue (WAT, iBAT, respectively). MCH binds to MCH receptor 1 (MCH1R), which is present on adipocytes. Currently it is unknown if Pmch-ablation changes adipocyte differentiation or sympathetic adipose drive. Using Pmch-deficient and wild-type rats on a standard low-fat diet, we analyzed dorsal subcutaneous and perirenal WAT mass and adipocyte morphology (size and number) throughout development, and indices of sympathetic activation in WAT and iBAT during adulthood. Moreover, using an in vitro approach we investigated the ability of MCH to modulate 3T3-L1 adipocyte differentiation. Pmch-deficiency decreased dorsal subcutaneous and perirenal WAT mass by reducing adipocyte size, but not number. In line with this, in vitro 3T3-L1 adipocyte differentiation was unaffected by MCH. Finally, adult Pmch-deficient rats had lower norepinephrine turnover (an index of sympathetic adipose drive) in WAT and iBAT than wild-type rats. Collectively, our data indicate that MCH/MCH1R-pathway does not modify adipocyte differentiation, whereas Pmch-deficiency in laboratory rats lowers adiposity throughout development and sympathetic adipose drive during adulthood.
Collapse
Affiliation(s)
- Joram D. Mul
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Eoghan O’Duibhir
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Yogendra B. Shrestha
- Department of Biology, Neurobiology and Behavior Program, and Exploring and Testing Strategies for Obesity Reversal Center, Georgia State University, Atlanta, Georgia, United States of America
| | - Arjen Koppen
- Department of Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Peter Vargoviç
- Laboratory for Stress Research, Institute of Experimental Endocrinology, Bratislava, Slovakia
| | - Pim W. Toonen
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Eleen Zarebidaki
- Department of Biology, Neurobiology and Behavior Program, and Exploring and Testing Strategies for Obesity Reversal Center, Georgia State University, Atlanta, Georgia, United States of America
| | - Richard Kvetnansky
- Laboratory for Stress Research, Institute of Experimental Endocrinology, Bratislava, Slovakia
| | - Eric Kalkhoven
- Department of Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Edwin Cuppen
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Timothy J. Bartness
- Department of Biology, Neurobiology and Behavior Program, and Exploring and Testing Strategies for Obesity Reversal Center, Georgia State University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
172
|
Neonatal leptin exposure specifies innervation of presympathetic hypothalamic neurons and improves the metabolic status of leptin-deficient mice. J Neurosci 2013; 33:840-51. [PMID: 23303959 DOI: 10.1523/jneurosci.3215-12.2013] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The paraventricular nucleus of the hypothalamus (PVH) consists of distinct functional compartments regulating neuroendocrine, behavioral, and autonomic activities that are involved in the homeostatic control of energy balance. These compartments receive synaptic inputs from neurons of the arcuate nucleus of the hypothalamus (ARH) that contains orexigenic agouti-related peptide (AgRP) and anorexigenic pro-opiomelanocortin (POMC) neuropeptides. The axon outgrowth from the ARH to PVH occurs during a critical postnatal period and is influenced by the adipocyte-derived hormone leptin, which promotes its development. However, little is known about leptin's role in specifying patterns of cellular connectivity in the different compartments of the PVH. To address this question, we used retrograde and immunohistochemical labeling to evaluate neuronal inputs onto sympathetic preautonomic and neuroendocrine neurons in PVH of leptin-deficient mice (Lep(ob)/Lep(ob)) exposed to a postnatal leptin treatment. In adult Lep(ob)/Lep(ob) mice, densities of AgRP- and α-melanocortin stimulating hormone (αMSH)-immunoreactive fibers were significantly reduced in neuroendocrine compartments of the PVH, but only AgRP were reduced in all regions containing preautonomic neurons. Moreover, postnatal leptin treatment significantly increased the density of AgRP-containing fibers and peptidergic inputs onto identified preautonomic, but not onto neuroendocrine cells. Neonatal leptin treatment neither rescued αMSH inputs onto neuroendocrine neurons, nor altered cellular ratios of inhibitory and excitatory inputs. These effects were associated with attenuated body weight gain, food intake and improved physiological response to sympathetic stimuli. Together, these results provide evidence that leptin directs cell type-specific patterns of ARH peptidergic inputs onto preautonomic neurons in the PVH, which contribute to normal energy balance regulation.
Collapse
|
173
|
Scherer T, Lehnert H, Hallschmid M. Brain insulin and leptin signaling in metabolic control: from animal research to clinical application. Endocrinol Metab Clin North Am 2013; 42:109-25. [PMID: 23391243 DOI: 10.1016/j.ecl.2012.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Besides the well-characterized effects of brain insulin and leptin in regulating food intake, insulin and leptin signaling to the central nervous system modulates a variety of metabolic processes, such as glucose and lipid homeostasis, as well as energy expenditure. This review summarizes the current literature on the contribution of central nervous insulin and leptin action to metabolic control in animals and humans. Potential therapeutic options based on the direct delivery of these peptides to the brain by, for example, intranasal administration, are discussed.
Collapse
Affiliation(s)
- Thomas Scherer
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna 1090, Austria.
| | | | | |
Collapse
|
174
|
Imbernon M, Beiroa D, Vázquez MJ, Morgan DA, Veyrat–Durebex C, Porteiro B, Díaz–Arteaga A, Senra A, Busquets S, Velásquez DA, Al–Massadi O, Varela L, Gándara M, López–Soriano F, Gallego R, Seoane LM, Argiles JM, López M, Davis RJ, Sabio G, Rohner–Jeanrenaud F, Rahmouni K, Dieguez C, Nogueiras R. Central melanin-concentrating hormone influences liver and adipose metabolism via specific hypothalamic nuclei and efferent autonomic/JNK1 pathways. Gastroenterology 2013; 144:636-649.e6. [PMID: 23142626 PMCID: PMC3663042 DOI: 10.1053/j.gastro.2012.10.051] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 10/10/2012] [Accepted: 10/31/2012] [Indexed: 01/18/2023]
Abstract
BACKGROUND & AIMS Specific neuronal circuits modulate autonomic outflow to liver and white adipose tissue. Melanin-concentrating hormone (MCH)-deficient mice are hypophagic, lean, and do not develop hepatosteatosis when fed a high-fat diet. Herein, we sought to investigate the role of MCH, an orexigenic neuropeptide specifically expressed in the lateral hypothalamic area, on hepatic and adipocyte metabolism. METHODS Chronic central administration of MCH and adenoviral vectors increasing MCH signaling were performed in rats and mice. Vagal denervation was performed to assess its effect on liver metabolism. The peripheral effects on lipid metabolism were assessed by real-time polymerase chain reaction and Western blot. RESULTS We showed that the activation of MCH receptors promotes nonalcoholic fatty liver disease through the parasympathetic nervous system, whereas it increases fat deposition in white adipose tissue via the suppression of sympathetic traffic. These metabolic actions are independent of parallel changes in food intake and energy expenditure. In the liver, MCH triggers lipid accumulation and lipid uptake, with c-Jun N-terminal kinase being an essential player, whereas in adipocytes MCH induces metabolic pathways that promote lipid storage and decreases lipid mobilization. Genetic activation of MCH receptors or infusion of MCH specifically in the lateral hypothalamic area modulated hepatic lipid metabolism, whereas the specific activation of this receptor in the arcuate nucleus affected adipocyte metabolism. CONCLUSIONS Our findings show that central MCH directly controls hepatic and adipocyte metabolism through different pathways.
Collapse
Affiliation(s)
- Monica Imbernon
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, S. Francisco s/n, Santiago de Compostela (A Coruña), Spain,Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Daniel Beiroa
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, S. Francisco s/n, Santiago de Compostela (A Coruña), Spain,Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - María J. Vázquez
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, S. Francisco s/n, Santiago de Compostela (A Coruña), Spain,Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Donald A. Morgan
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Christelle Veyrat–Durebex
- Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Begoña Porteiro
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, S. Francisco s/n, Santiago de Compostela (A Coruña), Spain,Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Adenis Díaz–Arteaga
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, S. Francisco s/n, Santiago de Compostela (A Coruña), Spain,Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Ana Senra
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, S. Francisco s/n, Santiago de Compostela (A Coruña), Spain,Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Silvia Busquets
- Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain
| | - Douglas A. Velásquez
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, S. Francisco s/n, Santiago de Compostela (A Coruña), Spain,Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Omar Al–Massadi
- Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain,Grupo Fisiopatología Endocrina, Complejo Hospitalario Universitario de Santiago-Instituto de Investigación Sanitaria (IDIS/SERGAS) Santiago de Compostela, Spain
| | - Luis Varela
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, S. Francisco s/n, Santiago de Compostela (A Coruña), Spain,Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Marina Gándara
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, S. Francisco s/n, Santiago de Compostela (A Coruña), Spain
| | | | - Rosalía Gallego
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, S. Francisco s/n, Santiago de Compostela (A Coruña), Spain
| | - Luisa M. Seoane
- Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain,Grupo Fisiopatología Endocrina, Complejo Hospitalario Universitario de Santiago-Instituto de Investigación Sanitaria (IDIS/SERGAS) Santiago de Compostela, Spain
| | - Josep M. Argiles
- Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain
| | - Miguel López
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, S. Francisco s/n, Santiago de Compostela (A Coruña), Spain,Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Roger J. Davis
- Howard Hughes Medical Institute, Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Guadalupe Sabio
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Françoise Rohner–Jeanrenaud
- Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Carlos Dieguez
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, S. Francisco s/n, Santiago de Compostela (A Coruña), Spain,Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Ruben Nogueiras
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, S. Francisco s/n, Santiago de Compostela (A Coruña), Spain,Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| |
Collapse
|
175
|
Abstract
We previously reported that peripheral leptin infusions in chronically decrebrate rats, in which the forebrain is neurally isolated from the hindbrain, increased body fat and decreased energy expenditure. Any central leptin response in decerebrate rats would depend upon the hindbrain. Here, we tested whether selective activation of hindbrain leptin receptors increased body fat. Fourth ventricle infusion of 0.6 μg leptin/day for 12 days increased body fat by 13% with no increase in food intake. Third ventricle leptin infusions decreased food intake, body fat, and lean tissue with a maximal response at 0.3 μg leptin/day. To test whether hindbrain receptors opposed activity of hypothalamic receptors, rats received peripheral infusions of 40 μg leptin/day and increasing 4th ventricle doses of the leptin receptor antagonist mutein protein. Mutein (3.0 μg/day) reduced body fat in PBS-infused rats to the same level as leptin-infused rats and reduced lean tissue in all rats. Leptin, but not mutein, inhibited food intake. By contrast, 3.0 μg/day mutein in the 3rd ventricle increased food intake and body fat in both PBS- and leptin-infused rats. In basal conditions, hindbrain leptin receptors may antagonize activity of forebrain receptors to protect lean and fat tissue, but there is no evidence for an anabolic role for hindbrain receptors when leptin is elevated. In a dietary study, rats increased energy intake when offered lard and 30% sucrose solution in addition to chow. Peripheral leptin infusion exaggerated the gain in body fat without altering energy intake confirming the potential for leptin to increase adiposity.
Collapse
Affiliation(s)
- Ruth B S Harris
- Department of Physiology, Georgia Health Sciences University, Augusta, GA 30912, USA.
| |
Collapse
|
176
|
Alén F, Ramírez-López MT, Gómez de Heras R, Rodríguez de Fonseca F, Orio L. Cannabinoid Receptors and Cholecystokinin in Feeding Inhibition. ANOREXIA 2013; 92:165-96. [DOI: 10.1016/b978-0-12-410473-0.00007-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
177
|
Jorgensen SB, O'Neill HM, Sylow L, Honeyman J, Hewitt KA, Palanivel R, Fullerton MD, Öberg L, Balendran A, Galic S, van der Poel C, Trounce IA, Lynch GS, Schertzer JD, Steinberg GR. Deletion of skeletal muscle SOCS3 prevents insulin resistance in obesity. Diabetes 2013; 62:56-64. [PMID: 22961088 PMCID: PMC3526029 DOI: 10.2337/db12-0443] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Obesity is associated with chronic low-grade inflammation that contributes to defects in energy metabolism and insulin resistance. Suppressor of cytokine signaling (SOCS)-3 expression is increased in skeletal muscle of obese humans. SOCS3 inhibits leptin signaling in the hypothalamus and insulin signal transduction in adipose tissue and the liver. Skeletal muscle is an important tissue for controlling energy expenditure and whole-body insulin sensitivity; however, the physiological importance of SOCS3 in this tissue has not been examined. Therefore, we generated mice that had SOCS3 specifically deleted in skeletal muscle (SOCS MKO). The SOCS3 MKO mice had normal muscle development, body mass, adiposity, appetite, and energy expenditure compared with wild-type (WT) littermates. Despite similar degrees of obesity when fed a high-fat diet, SOCS3 MKO mice were protected against the development of hyperinsulinemia and insulin resistance because of enhanced skeletal muscle insulin receptor substrate 1 (IRS1) and Akt phosphorylation that resulted in increased skeletal muscle glucose uptake. These data indicate that skeletal muscle SOCS3 does not play a critical role in regulating muscle development or energy expenditure, but it is an important contributing factor for inhibiting insulin sensitivity in obesity. Therapies aimed at inhibiting SOCS3 in skeletal muscle may be effective in reversing obesity-related glucose intolerance and insulin resistance.
Collapse
Affiliation(s)
- Sebastian Beck Jorgensen
- St. Vincent’s Institute of Medical Research and Department of Medicine, University of Melbourne, Fitzroy, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
178
|
Borges R, Dominguez N, Smith CB, Bandyopadhyay GK, O'Connor DT, Mahata SK, Bartolomucci A. Granins and catecholamines: functional interaction in chromaffin cells and adipose tissue. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2013; 68:93-113. [PMID: 24054141 DOI: 10.1016/b978-0-12-411512-5.00005-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Catecholamines (CAs) and granin peptides are costored in dense-core vesicles within the chromaffin cells of the adrenal medulla and in other endocrine organs and neurons. Granins play a major functional and structural role in chromaffin cells but are ubiquitous proteins, which are present also in secretory cells of the nervous, endocrine, and immune systems, where they regulate a number of cellular functions. Furthermore, recent studies also demonstrate that granin-derived peptides can functionally interact with CA to modulate key physiological functions such as lipolysis and blood pressure. In this chapter, we will provide a brief update on the interaction between CA and granins at the cellular and organ levels. We will first discuss recent data on the regulation of exocytosis of CA and peptides from the chromaffin cells by the sympathetic nervous system with a specific reference to the prominent role played by splanchnic nerve-derived pituitary adenylate cyclase-activating peptide (PACAP). Secondly, we will discuss the role of granins in the storage and regulation of exocytosis in large dense-core vesicles. Finally, we will provide an up-to-date review of the roles played by two granin-derived peptides, the chromogranin A-derived peptide catestatin and the VGF-derived peptide TLQP-21, on lipolysis and obesity. In conclusion, the knowledge gathered from recent findings on the role played by proteins/peptides in the sympathetic/target cell synapses, discussed in this chapter, would contribute to and provide novel mechanistic support for an increased appreciation of the physiological role of CA in human pathophysiology.
Collapse
Affiliation(s)
- Ricardo Borges
- Pharmacology Unit, Medical School, University of La Laguna, Tenerife, Spain
| | | | | | | | | | | | | |
Collapse
|
179
|
McDonald M, Higham DJ, Vass JK. Spectral algorithms for heterogeneous biological networks. Brief Funct Genomics 2012; 11:457-68. [PMID: 23117863 DOI: 10.1093/bfgp/els040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Spectral methods, which use information relating to eigenvectors, singular vectors and generalized singular vectors, help us to visualize and summarize sets of pairwise interactions. In this work, we motivate and discuss the use of spectral methods by taking a matrix computation view and applying concepts from applied linear algebra. We show that this unified approach is sufficiently flexible to allow multiple sources of network information to be combined. We illustrate the methods on microarray data arising from a large population-based study in human adipose tissue, combined with related information concerning metabolic pathways.
Collapse
Affiliation(s)
- Martin McDonald
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow G1 1XH, UK
| | | | | |
Collapse
|
180
|
Burgos-Ramos E, Sackmann-Sala L, Baquedano E, Cruz-Topete D, Barrios V, Argente J, Kopchick JJ. Central leptin and insulin administration modulates serum cytokine- and lipoprotein-related markers. Metabolism 2012; 61:1646-57. [PMID: 22658937 DOI: 10.1016/j.metabol.2012.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 04/12/2012] [Accepted: 05/01/2012] [Indexed: 10/28/2022]
Abstract
UNLABELLED In most obese patients there is an inflammatory state characterized by lipid abnormalities, hyperleptinemia and hyperinsulinemia. OBJECTIVE The objective was to identify mechanisms involved in leptin's role in the attenuation of the response to insulin using a proteomic approach. MATERIAL/METHODS We studied the serum proteomic profile of rats treated by central leptin infusion followed by an injection of insulin. We analyzed the relationship between these proteins and serum cytokine and apolipoprotein levels. RESULTS Out of 81 protein spots, intensity differences were found in 11, corresponding to 5 proteins: three isoforms of α1 macroglobulin; three of haptoglobin and serum amyloid P component-precursor. All of these are acute-phase proteins involved in inflammation and are correlated with cytokine levels. Additionally, two apolipoprotein E and two apolipoprotein A1 isoforms were identified and were found to correlate with LDL and HDL. CONCLUSIONS Our results indicate that increased leptin and insulin levels change these circulating proteins, thus promoting systemic inflammation and changing lipid metabolism.
Collapse
Affiliation(s)
- Emma Burgos-Ramos
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto Investigación Sanitaria Princesa, Madrid, E-28009 Spain
| | | | | | | | | | | | | |
Collapse
|
181
|
Scherer T, Lindtner C, Zielinski E, O'Hare J, Filatova N, Buettner C. Short term voluntary overfeeding disrupts brain insulin control of adipose tissue lipolysis. J Biol Chem 2012; 287:33061-9. [PMID: 22810223 PMCID: PMC3463338 DOI: 10.1074/jbc.m111.307348] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 07/06/2012] [Indexed: 12/20/2022] Open
Abstract
Insulin controls fatty acid (FA) release from white adipose tissue (WAT) through direct effects on adipocytes and indirectly through hypothalamic signaling by reducing sympathetic nervous system outflow to WAT. Uncontrolled FA release from WAT promotes lipotoxicity, which is characterized by inflammation and insulin resistance that leads to and worsens type 2 diabetes. Here we tested whether early diet-induced insulin resistance impairs the ability of hypothalamic insulin to regulate WAT lipolysis and thus contributes to adipose tissue dysfunction. To this end we fed male Sprague-Dawley rats a 10% lard diet (high fat diet (HFD)) for 3 consecutive days, which is known to induce systemic insulin resistance. Rats were studied by euglycemic pancreatic clamps and concomitant infusion of either insulin or vehicle into the mediobasal hypothalamus. Short term HFD feeding led to a 37% increase in caloric intake and elevated base-line free FAs and insulin levels compared with rats fed regular chow. Overfeeding did not impair insulin signaling in WAT, but it abolished the ability of mediobasal hypothalamus insulin to suppress WAT lipolysis and hepatic glucose production as assessed by glycerol and glucose flux. HFD feeding also increased hypothalamic levels of the endocannabinoid 2-arachidonoylglycerol after only 3 days. In summary, overfeeding impairs hypothalamic insulin action, which may contribute to unrestrained lipolysis seen in human obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Thomas Scherer
- From the Department of Medicine and Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029
| | - Claudia Lindtner
- From the Department of Medicine and Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029
| | - Elizabeth Zielinski
- From the Department of Medicine and Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029
| | - James O'Hare
- From the Department of Medicine and Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029
| | - Nika Filatova
- From the Department of Medicine and Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029
| | - Christoph Buettner
- From the Department of Medicine and Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029
| |
Collapse
|
182
|
Coppari R, Bjørbæk C. Leptin revisited: its mechanism of action and potential for treating diabetes. Nat Rev Drug Discov 2012; 11:692-708. [PMID: 22935803 PMCID: PMC4019022 DOI: 10.1038/nrd3757] [Citation(s) in RCA: 214] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since the discovery of leptin in 1994, we now have a better understanding of the cellular and molecular mechanisms underlying its biological effects. In addition to its established anti-obesity effects, leptin exerts antidiabetic actions that are independent of its regulation of body weight and food intake. In particular, leptin can correct diabetes in animal models of type 1 and type 2 diabetes. In addition, long-term leptin replacement therapy improves glycaemic control, insulin sensitivity and plasma triglycerides in patients with severe insulin resistance due to lipodystrophy. These results have spurred enthusiasm for the use of leptin therapy to treat diabetes. Here, we review the current understanding of the glucoregulatory functions of leptin, emphasizing its central mechanisms of action and lessons learned from clinical studies, and discuss possible therapeutic applications of leptin in the treatment of type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Roberto Coppari
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas TX, 75390, USA
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- The Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA, 92697, USA
| | - Christian Bjørbæk
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston MA, 02215, USA
| |
Collapse
|
183
|
Abstract
The function of MEX3C, the mammalian homolog of Caenorhabditis elegans RNA-binding protein muscle excess 3 (MEX-3), was unknown until our recent report that MEX3C is necessary for normal postnatal growth and enhances the expression of local bone Igf1 expression. Here we report the pivotal role of Mex3c in energy balance regulation. Mex3c mutation caused leanness in both heterozygous and homozygous transgenic mice, as well as a more beneficial blood glucose and lipid profile in homozygous transgenic mice, in both sexes. Although transgenic mice showed normal food intake and fecal lipid excretion, they had increased energy expenditure independent of physical activity. Mutant mice had normal body temperature, Ucp1 expression in brown adipose tissue, and muscle and liver fatty acid oxidation. Mex3c is expressed in neurons and is detectable in the arcuate nucleus, the ventromedial nucleus, and the dorsomedial nucleus of the hypothalamus. Mex3c was not detected in NPY or POMC neurons but was detected in leptin-responsive neurons in the ventromedial nucleus. Mex3c and Leptin double mutant mice were growth retarded and obese and had blood profiles similar to those of ob/ob mice but showed none of the steatosis observed in ob/ob mice. Our data show that Mex3c is involved in energy balance regulation.
Collapse
|
184
|
Abstract
Bariatric procedures are now known to have an effect on hunger as well as on metabolism. The role of central nervous pathways in causing these effects after bariatric surgery is now being elucidated. A brief overview of these pathways has been presented for the sake of bariatric surgeons. A PubMed search was made using various search phrases to retrieve all original articles concerning the effect of bariatric surgery on the neural pathways. The mechanisms regulating the food intake and energy expenditure can be broadly divided into homeostatic and hedonic systems. The effect of bariatric surgery on the homeostatic system in animal models is not clear. A decrease in preference for sweet taste and high calorie foods has been demonstrated in animal models. The effect of bariatric surgery on the hedonic system in humans has been consistent with decreased activation of the hedonic system being demonstrated by functional MRI and decreased preference for intake of high energy foods also being observed post-surgery. The effect of bariatric surgery on dopamine signaling, which is involved in the hedonic system, is however not clear. Functional MRI studies have also demonstrated increased activation of the hypothalamus after surgery. Various studies utilizing questionnaires have demonstrated increased satiety and decreased hunger after bariatric surgery.
Collapse
|
185
|
Yonezawa R, Wada T, Matsumoto N, Morita M, Sawakawa K, Ishii Y, Sasahara M, Tsuneki H, Saito S, Sasaoka T. Central versus peripheral impact of estradiol on the impaired glucose metabolism in ovariectomized mice on a high-fat diet. Am J Physiol Endocrinol Metab 2012; 303:E445-56. [PMID: 22550066 DOI: 10.1152/ajpendo.00638.2011] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Age-related loss of ovarian function promotes adiposity and insulin resistance in women. Estrogen (E(2)) directly enhances insulin sensitivity and suppresses lipogenesis in peripheral tissues. Recently, the central actions of E(2) in the regulation of energy homeostasis are becoming clearer; however, the functional relevance and degree of contribution of the central vs. peripheral actions of E(2) are currently unknown. Therefore, we prepared and analyzed four groups of mice. 1) CONTROL: sham-operated mice fed a regular diet, 2) OVX-HF: ovariectomized (OVX) mice fed a 60% high-fat diet (HF), 3) E2-SC: OVX-HF mice subcutaneously treated with E(2), and 4) E2-ICV: OVX-HF mice treated with E(2) intracerebroventricularly. OVX-HF mice showed increased body weight with both visceral and subcutaneous fat volume enlargement, glucose intolerance, and insulin resistance. Both E2-SC and E2-ICV equally ameliorated these abnormalities. Although the size of adipocytes and number of CD11c-positive macrophages in perigonadal fat in OVX-HF were reduced by both E(2) treatments, peripherally administered E(2) decreased the expression of TNFα, lipoprotein lipase, and fatty acid synthase in the white adipose tissue (WAT) of OVX-HF. In contrast, centrally administered E(2) increased hormone-sensitive lipase in WAT, decreased the hepatic expression of gluconeogenic enzymes, and elevated core body temperature and energy expenditure with marked upregulation of uncoupling proteins in the brown adipose tissue. These results suggest that central and peripheral actions of E(2) regulate insulin sensitivity and glucose metabolism via different mechanisms, and their coordinated effects may be important to prevent the development of obesity and insulin resistance in postmenopausal women.
Collapse
Affiliation(s)
- Rika Yonezawa
- Department of Obstetrics and Gynecology, University of Toyama, Toyama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
186
|
Is leptin the parabiotic "satiety" factor? Past and present interpretations. Appetite 2012; 61:111-8. [PMID: 22889986 DOI: 10.1016/j.appet.2012.08.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 08/01/2012] [Indexed: 01/15/2023]
Abstract
In 1959 Hervey hypothesized that a circulating feedback signal informed the hypothalamus of the size of fat stores and initiated appropriate corrections to energy balance. The hypothesis resulted from a parabiosis study in which one animal became obese following lesioning of the ventromedial hypothalamus. The partner of the lesioned rat was hypophagic and lost a large amount of body fat. Similar results came from parabiosis studies with obese Zucker rats and rats that overate due to stimulation of the lateral hypothalamus. In studies in which one parabiont was made obese by overfeeding the non-overfed partners lost substantial amounts of fat with a minimal reduction in food intake and no loss of lean tissue. The loss of fat was due to inhibition of adipose lipogenesis and other metabolic adjustments typical of food restriction. Parabiosis with genetically obese mice implied that ob/ob mice did not produce the feedback signal and subsequently the mutant ob protein, leptin, was identified. This paper provides a review and interpretation of parabiosis work that preceded the discovery of leptin, an evaluation of leptin in relation to its function as the circulating feedback signal and evidence for additional circulating factors involved in the control of adipose tissue mass.
Collapse
|
187
|
Tam J, Cinar R, Liu J, Godlewski G, Wesley D, Jourdan T, Szanda G, Mukhopadhyay B, Chedester L, Liow JS, Innis RB, Cheng K, Rice KC, Deschamps JR, Chorvat RJ, McElroy JF, Kunos G. Peripheral cannabinoid-1 receptor inverse agonism reduces obesity by reversing leptin resistance. Cell Metab 2012; 16:167-79. [PMID: 22841573 PMCID: PMC3832894 DOI: 10.1016/j.cmet.2012.07.002] [Citation(s) in RCA: 285] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 04/16/2012] [Accepted: 07/06/2012] [Indexed: 12/13/2022]
Abstract
Obesity-related leptin resistance manifests in loss of leptin's ability to reduce appetite and increase energy expenditure. Obesity is also associated with increased activity of the endocannabinoid system, and CB(1) receptor (CB(1)R) inverse agonists reduce body weight and the associated metabolic complications, although adverse neuropsychiatric effects halted their therapeutic development. Here we show that in mice with diet-induced obesity (DIO), the peripherally restricted CB(1)R inverse agonist JD5037 is equieffective with its brain-penetrant parent compound in reducing appetite, body weight, hepatic steatosis, and insulin resistance, even though it does not occupy central CB(1)R or induce related behaviors. Appetite and weight reduction by JD5037 are mediated by resensitizing DIO mice to endogenous leptin through reversing the hyperleptinemia by decreasing leptin expression and secretion by adipocytes and increasing leptin clearance via the kidney. Thus, inverse agonism at peripheral CB(1)R not only improves cardiometabolic risk in obesity but has antiobesity effects by reversing leptin resistance.
Collapse
Affiliation(s)
- Joseph Tam
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
- Correspondence: (J.T.), (G.K.)
| | - Resat Cinar
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jie Liu
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Grzegorz Godlewski
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Wesley
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tony Jourdan
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gergö Szanda
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bani Mukhopadhyay
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lee Chedester
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert B. Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kejun Cheng
- Chemical Biology Research Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kenner C. Rice
- Chemical Biology Research Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
- Correspondence: (J.T.), (G.K.)
| |
Collapse
|
188
|
Tang Y, Ho G, Li Y, Hall MA, Hills RL, Black SC, Liang Y, Demarest KT. Beneficial metabolic effects of CB1R anti-sense oligonucleotide treatment in diet-induced obese AKR/J mice. PLoS One 2012; 7:e42134. [PMID: 22870290 PMCID: PMC3411690 DOI: 10.1371/journal.pone.0042134] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 07/03/2012] [Indexed: 11/30/2022] Open
Abstract
An increasing amount of evidence supports pleiotropic metabolic roles of the cannibinoid-1 receptor (CB1R) in peripheral tissues such as adipose, liver, skeletal muscle and pancreas. To further understand the metabolic consequences of specific blockade of CB1R function in peripheral tissues, we performed a 10-week-study with an anti-sense oligonucleotide directed against the CB1R in diet-induced obese (DIO) AKR/J mice. DIO AKR/J mice were treated with CB1R ASO Isis-414930 (6.25, 12.5 and 25 mg/kg/week) or control ASO Isis-141923 (25 mg/kg/week) via intraperitoneal injection for 10 weeks. At the end of the treatment, CB1R mRNA from the 25 mg/kg/week CB1R ASO group in the epididymal fat and kidney was decreased by 81% and 63%, respectively. Body weight gain was decreased in a dose-dependent fashion, significantly different in the 25 mg/kg/week CB1R ASO group (46.1±1.0 g vs veh, 51.2±0.9 g, p<0.05). Body fat mass was reduced in parallel with attenuated body weight gain. CB1R ASO treatment led to decreased fed glucose level (at week 8, 25 mg/kg/week group, 145±4 mg/dL vs veh, 195±10 mg/dL, p<0.05). Moreover, CB1R ASO treatment dose-dependently improved glucose excursion during an oral glucose tolerance test, whereas control ASO exerted no effect. Liver steatosis was also decreased upon CB1R ASO treatment. At the end of the study, plasma insulin and leptin levels were significantly reduced by 25 mg/kg/week CB1R ASO treatment. SREBP1 mRNA expression was decreased in both epididymal fat and liver. G6PC and fatty acid translocase/CD36 mRNA levels were also reduced in the liver. In summary, CB1R ASO treatment in DIO AKR/J mice led to improved insulin sensitivity and glucose homeostasis. The beneficial effects of CB1R ASO treatment strongly support the notion that selective inhibition of the peripheral CB1R, without blockade of central CB1R, may serve as an effective approach for treating type II diabetes, obesity and the metabolic syndrome.
Collapse
MESH Headings
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/pathology
- Animals
- CD36 Antigens/genetics
- CD36 Antigens/metabolism
- Diet/adverse effects
- Dose-Response Relationship, Drug
- Fatty Liver/chemically induced
- Fatty Liver/genetics
- Fatty Liver/metabolism
- Fatty Liver/pathology
- Female
- Glucose/genetics
- Glucose/metabolism
- Insulin/genetics
- Insulin/metabolism
- Liver/metabolism
- Liver/pathology
- Male
- Metabolic Syndrome/chemically induced
- Metabolic Syndrome/genetics
- Metabolic Syndrome/metabolism
- Metabolic Syndrome/pathology
- Mice
- Mice, Inbred AKR
- Obesity/chemically induced
- Obesity/drug therapy
- Obesity/genetics
- Obesity/metabolism
- Oligodeoxyribonucleotides, Antisense/pharmacology
- Organ Specificity/drug effects
- Organ Specificity/genetics
- RNA, Messenger/antagonists & inhibitors
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Sterol Regulatory Element Binding Protein 1/genetics
- Sterol Regulatory Element Binding Protein 1/metabolism
Collapse
Affiliation(s)
- Yuting Tang
- Cardiovascular and Metabolism Therapeutic Area, Janssen Pharmaceutical Companies of Johnson and Johnson, Spring House, Pennsylvania, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|
189
|
Elias CF, Purohit D. Leptin signaling and circuits in puberty and fertility. Cell Mol Life Sci 2012; 70:841-62. [PMID: 22851226 PMCID: PMC3568469 DOI: 10.1007/s00018-012-1095-1] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 07/05/2012] [Accepted: 07/09/2012] [Indexed: 12/22/2022]
Abstract
Leptin is an adipocyte-derived hormone involved in a myriad of physiological process, including the control of energy balance and several neuroendocrine axes. Leptin-deficient mice and humans are obese, diabetic, and display a series of neuroendocrine and autonomic abnormalities. These individuals are infertile due to a lack of appropriate pubertal development and inadequate synthesis and secretion of gonadotropins and gonadal steroids. Leptin receptors are expressed in many organs and tissues, including those related to the control of reproductive physiology (e.g., the hypothalamus, pituitary gland, and gonads). In the last decade, it has become clear that leptin receptors located in the brain are major players in most leptin actions, including reproduction. Moreover, the recent development of molecular techniques for brain mapping and the use of genetically modified mouse models have generated crucial new findings for understanding leptin physiology and the metabolic influences on reproductive health. In the present review, we will highlight the new advances in the field, discuss the apparent contradictions, and underline the relevance of this complex physiological system to human health. We will focus our review on the hypothalamic circuitry and potential signaling pathways relevant to leptin’s effects in reproductive control, which have been identified with the use of cutting-edge technologies of molecular mapping and conditional knockouts.
Collapse
Affiliation(s)
- Carol F Elias
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Y6-220B, Dallas, TX, 75390-9077, USA.
| | | |
Collapse
|
190
|
Silvestri C, Di Marzo V. Second generation CB1 receptor blockers and other inhibitors of peripheral endocannabinoid overactivity and the rationale of their use against metabolic disorders. Expert Opin Investig Drugs 2012; 21:1309-22. [DOI: 10.1517/13543784.2012.704019] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Cristoforo Silvestri
- Institute of Biomolecular Chemistry – CNR, Endocannabinoid Research Group,
Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy ;
| | - Vincenzo Di Marzo
- Institute of Biomolecular Chemistry – CNR, Endocannabinoid Research Group,
Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy ;
| |
Collapse
|
191
|
Renal leptin in experimental nephrotic syndrome. J Genet Eng Biotechnol 2012. [DOI: 10.1016/j.jgeb.2011.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
192
|
Kim KW, Donato J, Berglund ED, Choi YH, Kohno D, Elias CF, Depinho RA, Elmquist JK. FOXO1 in the ventromedial hypothalamus regulates energy balance. J Clin Invest 2012; 122:2578-89. [PMID: 22653058 DOI: 10.1172/jci62848] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 04/18/2012] [Indexed: 12/15/2022] Open
Abstract
The transcription factor FOXO1 plays a central role in metabolic homeostasis by regulating leptin and insulin activity in many cell types, including neurons. However, the neurons mediating these effects and the identity of the molecular targets through which FOXO1 regulates metabolism remain to be defined. Here, we show that the ventral medial nucleus of the hypothalamus (VMH) is a key site of FOXO1 action. We found that mice lacking FOXO1 in steroidogenic factor 1 (SF-1) neurons of the VMH are lean due to increased energy expenditure. The mice also failed to appropriately suppress energy expenditure in response to fasting. Furthermore, these mice displayed improved glucose tolerance due to increased insulin sensitivity in skeletal muscle and heart. Gene expression profiling and sequence analysis revealed several pathways regulated by FOXO1. In addition, we identified the nuclear receptor SF-1 as a direct FOXO1 transcriptional target in the VMH. Collectively, our data suggest that the transcriptional networks modulated by FOXO1 in VMH neurons are key components in the regulation of energy balance and glucose homeostasis.
Collapse
Affiliation(s)
- Ki Woo Kim
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center(UT Southwestern), Dallas, TX, USA
| | | | | | | | | | | | | | | |
Collapse
|
193
|
Roubos EW, Dahmen M, Kozicz T, Xu L. Leptin and the hypothalamo-pituitary-adrenal stress axis. Gen Comp Endocrinol 2012; 177:28-36. [PMID: 22293575 DOI: 10.1016/j.ygcen.2012.01.009] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 01/10/2012] [Accepted: 01/12/2012] [Indexed: 12/20/2022]
Abstract
Leptin is a 16-kDa protein mainly produced and secreted by white adipose tissue and informing various brain centers via leptin receptor long and short forms about the amount of fat stored in the body. In this way leptin exerts a plethora of regulatory functions especially related to energy intake and metabolism, one of which is controlling the activity of the hypothalamo-pituitary-adrenal (HPA) stress axis. First, this review deals with the basic properties of leptin's structure and signaling at the organ, cell and molecule level, from lower vertebrates to humans but with emphasis on rodents because these have been investigated in most detail. Then, attention is given to the various interactions of adipose leptin with the HPA-axis, at the levels of the hypothalamus (especially the paraventricular nucleus), the anterior lobe of the pituitary gland (action on corticotropes) and the adrenal gland, where it releases corticosteroids needed for adequate stress adaptation. Also, possible local production and autocrine and paracrine actions of leptin at the hypothalamic and pituitary levels of the HPA-axis are being considered. Finally, a schematic model is presented showing the ways peripherally and centrally produced leptin may modulate, via the HPA-axis, stress adaptation in conjunction with the control of energy homeostasis.
Collapse
Affiliation(s)
- Eric W Roubos
- Department of Cellular Animal Physiology, Faculty of Science, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, PO Box 9010, 6500 GL Nijmegen, The Netherlands.
| | | | | | | |
Collapse
|
194
|
Abstract
Osteoporosis is less common in individuals with high fat mass. This putative osteoprotection is likely an adaptive mechanism that allows obese individuals to better carry their increased body mass. Recent studies have focused on hormones that link fat to bone. Adipokines, such as leptin, modulate bone cells through both direct and indirect actions, whereas molecules activating peroxisome proliferator-activated receptor γ drive mesenchymal stem cell differentiation towards adipocytes away from the osteoblastic lineage. There is emerging evidence that bone-derived osteocalcin regulates insulin release and insulin sensitivity and, hence, might indirectly affect fat mass. Despite these molecular connections between fat and bone, animal and human studies call into question a primary role for body fat in determining bone mass. Mice devoid of fat do not have a skeletal phenotype, and in humans, the observed correlations between bone and body mass are not just due to adipose tissue. An improved understanding of the integrative physiology at the fat-bone interface should allow us develop therapies for both osteoporosis and obesity.
Collapse
Affiliation(s)
- Mone Zaidi
- Mount Sinai Bone Program and Department of Medicine, Mount Sinai School of Medicine, New York, New York 10029, USA.
| | | | | | | |
Collapse
|
195
|
Yue JTY, Mighiu PI, Naples M, Adeli K, Lam TKT. Glycine normalizes hepatic triglyceride-rich VLDL secretion by triggering the CNS in high-fat fed rats. Circ Res 2012; 110:1345-54. [PMID: 22474253 DOI: 10.1161/circresaha.112.268276] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE Dysregulation of hepatic triglyceride (TG)-rich very low-density lipoproteins (VLDL-TG) in obesity and type 2 diabetes contributes to the dyslipidemia that leads to cardiovascular morbidity. The central nervous system (CNS), particularly the hypothalamus, regulates hepatic lipid metabolism. Although the underlying neurocircuitry remains elusive, glycine has been documented to enhance CNS N-methyl-d-aspartate (NMDA) receptor-mediated transmission. OBJECTIVE We tested the hypothesis that glycine regulates hepatic VLDL-TG secretion by potentiating NMDA receptor-mediated transmission in the CNS. METHODS AND RESULTS Using 10-hour fasted male Sprague-Dawley rats implanted with stereotaxic cannulae into an extrahypothalamic region termed the dorsal vagal complex (DVC) and vascular catheters to enable direct DVC infusion and blood sampling, respectively, the rate of hepatic VLDL-TG secretion was measured following tyloxapol (an inhibitor of lipoprotein lipase) injection. Direct DVC infusion of glycine lowered VLDL-TG secretion, whereas NMDA receptor blocker MK-801 fully negated glycine's effect. NR1 subunit of NMDA receptor antagonist 7-chlorokynurenic acid, adenoviral injection of NR1 short hairpin RNA (shRNA), and hepatic vagotomy also nullified glycine's effect. Finally, DVC glycine normalized the hypersecretion of VLDL-TG induced by high-fat feeding. CONCLUSIONS Molecular and pharmacological inhibition of the NR1-containing NMDA receptors in the DVC negated the ability of glycine to inhibit hepatic secretion of VLDL-TG in vivo. Importantly, the hypersecretion of VLDL-TG from the liver induced by a model of high-fat feeding was restored by the hepatic lipid control of CNS glycine sensing. These findings collectively suggest that glycine or glycine analogues may have therapeutic benefits in lowering plasma lipid levels in diabetes and obesity by triggering the CNS.
Collapse
Affiliation(s)
- Jessica T Y Yue
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | | | | | | | | |
Collapse
|
196
|
Ko JY, Wu RW, Kuo SJ, Chen MW, Yeh DW, Ke HC, Wu SL, Wang FS. Cannabinoid receptor 1 mediates glucocorticoid-induced bone loss in rats by perturbing bone mineral acquisition and marrow adipogenesis. ACTA ACUST UNITED AC 2012; 64:1204-14. [DOI: 10.1002/art.33457] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
197
|
Pan W, Hsuchou H, Cornelissen-Guillaume GG, Jayaram B, Wang Y, Tu H, Halberg F, Wu X, Chua SC, Kastin AJ. Endothelial leptin receptor mutation provides partial resistance to diet-induced obesity. J Appl Physiol (1985) 2012; 112:1410-8. [PMID: 22323652 DOI: 10.1152/japplphysiol.00590.2011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Leptin, a polypeptide hormone produced mainly by adipocytes, has diverse effects in both the brain and peripheral organs, including suppression of feeding. Other than mediating leptin transport across the blood-brain barrier, the role of the endothelial leptin receptor remains unclear. We recently generated a mutant mouse strain lacking endothelial leptin receptor signaling, and showed that there is an increased uptake of leptin by brain parenchyma after its delivery by in situ brain perfusion. Here, we tested the hypothesis that endothelial leptin receptor mutation confers partial resistance to diet-induced obesity. These ELKO mice had similar body weight and percent fat as their wild-type littermates when fed with rodent chow, but blood concentrations of leptin were significantly elevated. In response to a high-fat diet, wild-type mice had a greater gain of body weight and fat than ELKO mice. As shown by metabolic chamber measurement, the ELKO mice had higher oxygen consumption, carbon dioxide production, and heat dissipation, although food intake was similar to that of the wild-type mice and locomotor activity was even reduced. This indicates that the partial resistance to diet-induced obesity was mediated by higher metabolic activity in the ELKO mice. Since neuronal leptin receptor knockout mice show obesity and diabetes, the results suggest that endothelial leptin signaling shows opposite effects from that of neuronal leptin signaling, with a facilitatory role in diet-induced obesity.
Collapse
Affiliation(s)
- Weihong Pan
- Blood-Brain Barrier Group, Pennington Biomedical Research Center, 6400 Perkins Rd., Baton Rouge, LA 70808, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Schulz C, Paulus K, Jöhren O, Lehnert H. Intranasal leptin reduces appetite and induces weight loss in rats with diet-induced obesity (DIO). Endocrinology 2012; 153:143-53. [PMID: 22128019 DOI: 10.1210/en.2011-1586] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Resistance to brain-mediated effects of leptin is a characteristic feature of obesity, resulting from alterations in leptin receptor signaling in hypothalamic neurons and/or transport across the blood-brain-barrier. We have shown previously, that the latter can be circumvented by intranasal (i.n.) application of leptin in lean rats. This prompted us to test i.n. leptin in animals with diet-induced obesity (DIO) as a basis for future human administration. DIO was induced in male Wistar rats by feeding a cafeteria diet for 25 or 32 wk, respectively. Consecutively, these DIO animals (seven to eight per treatment) and standard diet rats (lean) (14-15 per treatment, matched for age and diet duration) were treated with 0.1, 0.2 mg/kg leptin, or control solution i.n. daily for 4 wk before onset of dark period. Energy intake and body weight were measured daily; blood glucose, serum insulin, and leptin were measured before and after treatment. Expression of hypothalamic neuropeptides was assessed by quantitative real-time PCR. We demonstrate, for the first time, that i.n. leptin reduces appetite and induces weight loss in DIO to the same extent as in lean rats. Our findings are supported accordingly by an altered expression pattern of anorexigenic and orexigenic neuropeptides in the hypothalamus, e.g. proopiomelanocortin, cocaine and amphetamine-related transcript, neuropeptide Y, agouti-related protein. It now appears clear that i.n. leptin is effectively acting in obese animals in the same fashion as in their lean counterparts. These findings now clearly warrant studies in humans and may open new perspectives in the treatment of obesity.
Collapse
Affiliation(s)
- Carla Schulz
- Department of Internal Medicine I, Luebeck University, Ratzeburger Allee 160, 23538 Luebeck, Germany.
| | | | | | | |
Collapse
|
199
|
Bermudez-Silva FJ, Cardinal P, Cota D. The role of the endocannabinoid system in the neuroendocrine regulation of energy balance. J Psychopharmacol 2012; 26:114-24. [PMID: 21824982 DOI: 10.1177/0269881111408458] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Animal and human studies carried out so far have established a role for the endocannabinoid system (ECS) in the regulation of energy balance. Here we critically discuss the role of the endocannabinoid signalling in brain structures, such as the hypothalamus and reward-related areas, and its interaction with neurotransmitter and neuropeptide systems involved in the regulation of food intake and body weight. The ECS has been found to interact with peripheral signals, like leptin, insulin, ghrelin and satiety hormones and the resulting effects on both central and peripheral mechanisms affecting energy balance and adiposity will be described. Furthermore, ECS dysregulation has been associated with the development of dyslipidemia, glucose intolerance and obesity; phenomena that are often accompanied by a plethora of neuroendocrine alterations which might play a causal role in determining ECS dysregulation. Despite the withdrawal of the first generation of cannabinoid type 1 receptor (CB1) antagonists from the pharmaceutical market due to the occurrence of psychiatric adverse events, new evidence suggests that peripherally restricted CB1 antagonists might be efficacious for the treatment of obesity and its associated metabolic disorders. Thus, a perspective on new promising strategies to selectively target the ECS in the context of energy balance regulation is given.
Collapse
|
200
|
Warne JP, Alemi F, Reed AS, Varonin JM, Chan H, Piper ML, Mullin ME, Myers MG, Corvera CU, Xu AW. Impairment of central leptin-mediated PI3K signaling manifested as hepatic steatosis independent of hyperphagia and obesity. Cell Metab 2011; 14:791-803. [PMID: 22152304 PMCID: PMC3240844 DOI: 10.1016/j.cmet.2011.11.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 09/13/2011] [Accepted: 11/02/2011] [Indexed: 11/22/2022]
Abstract
Hepatic steatosis is generally thought to develop via peripheral mechanisms associated with obesity. We show that chronic central infusion of leptin suppresses hepatic lipogenic gene expression and reduces triglyceride content via stimulation of hepatic sympathetic activity. This leptin function is independent of feeding and body weight but requires phosphatidylinositol 3-kinase (PI3K) signaling. Attenuation of leptin-induced PI3K signaling, brought about by transgenic expression of phosphatase and tensin homolog (PTEN) in leptin receptor neurons, leads to decreased hepatic sympathetic tone and increased triglyceride levels without affecting adiposity or hepatic insulin signaling. Central leptin's effects on hepatic norepinephrine levels and triglyceride content are blunted in these mutant mice. Simultaneous downregulation of PI3K and signal transducer and activator of transcription-3 (Stat3) in leptin receptor neurons does not exacerbate obesity but causes more severe hepatic steatosis. Together, our results indicate that central cellular leptin resistance in PI3K signaling manifests as hepatic steatosis without causing obesity.
Collapse
Affiliation(s)
- James P. Warne
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Farzad Alemi
- General Surgery, San Francisco VA Medical Center, University of California, San Francisco, San Francisco, CA 94143
| | - Alison S. Reed
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jillian M. Varonin
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Helen Chan
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Merisa L. Piper
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mark E. Mullin
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Martin G. Myers
- Departments of Internal Medicine and Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Carlos U. Corvera
- General Surgery, San Francisco VA Medical Center, University of California, San Francisco, San Francisco, CA 94143
| | - Allison W. Xu
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|