1
|
Goddard KE, Fountain SJ. Characterisation of neurogenic lipolytic responses in white adipose tissue ex vivo. Br J Pharmacol 2025; 182:1975-1988. [PMID: 39894466 DOI: 10.1111/bph.17445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/26/2024] [Accepted: 12/17/2024] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND AND PURPOSE Dysfunction of the autonomic nervous system is associated with cardiovascular dysfunction, including metabolic syndrome and obesity. Understanding mechanisms of neurogenic control of white adipose tissue is key to understanding adipose physiology and pathophysiology, though there is limited research exploring this in adipose tissue using pharmacological tools, as opposed to genetic knockout models. EXPERIMENTAL APPROACH Inguinal white adipose tissue from C57BL/6J mice was used in this study. We used immunocytochemistry to determine tissue innervation and glycerol release assays to quantify lipolysis in adipose tissue and isolated adipocytes. The voltage-gated Na+ channel opener veratridine was used to stimulate nervous activity in tissue ex vivo. The role of neurotransmitters and receptors mediating veratridine-evoked lipolysis in adipose tissue was pharmacologically characterised. KEY RESULTS Veratridine evoked glycerol release in white adipose tissue but not from isolated adipocytes. This release was abolished by tetrodotoxin and propranolol. Veratridine also induced noradrenaline release from white adipose tissue. Veratridine- and noradrenaline-evoked glycerol release was blocked by the β2-adrenoceptor antagonist ICI-118551 but not by the β1-adrenoceptor antagonist CGP 20712A. Purported β3-adrenoceptor antagonists L-748337 and SR59230A stimulated glycerol release from tissue and from isolated adipocytes. Neither L-748337 or SR59230A antagonised veratridine-evoked glycerol release but SR59230A antagonised noradrenaline-evoked glycerol release. We exclude contributions of sensory neuropeptides and the autonomic neurotransmitters neuropeptide Y and ATP. CONCLUSION AND IMPLICATIONS Neurogenic lipolytic responses can be measured in white adipose tissue ex vivo using veratridine to stimulate nerve activity. The lipolytic responses are mediated by β2-adrenoceptor activation. This study provides the first evidence of neurogenic lipolysis in tissue ex vivo.
Collapse
Affiliation(s)
| | - Samuel J Fountain
- School of Biological Sciences, University of East Anglia, Norwich, UK
| |
Collapse
|
2
|
Wang Y, Ye L. Somatosensory innervation of adipose tissues. Physiol Behav 2023; 265:114174. [PMID: 36965573 PMCID: PMC11537203 DOI: 10.1016/j.physbeh.2023.114174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023]
Abstract
The increasing prevalence of obesity and type 2 diabetes has led to a greater interest in adipose tissue physiology. Adipose tissue is now understood as an organ with endocrine and thermogenic capacities in addition to its role in fat storage. It plays a critical role in systemic metabolism and energy regulation, and its activity is tightly regulated by the nervous system. Fat is now recognized to receive sympathetic innervation, which transmits information from the brain, as well as sensory innervation, which sends information into the brain. The role of sympathetic innervation in adipose tissue has been extensively studied. However, the extent and the functional significance of sensory innervation have long been unclear. Recent studies have started to reveal that sensory neurons robustly innervate adipose tissue and play an important role in regulating fat activity. This brief review will discuss both historical evidence and recent advances, as well as important remaining questions about the sensory innervation of adipose tissue.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Li Ye
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
3
|
GPCR in Adipose Tissue Function-Focus on Lipolysis. Biomedicines 2023; 11:biomedicines11020588. [PMID: 36831123 PMCID: PMC9953751 DOI: 10.3390/biomedicines11020588] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Adipose tissue can be divided anatomically, histologically, and functionally into two major entities white and brown adipose tissues (WAT and BAT, respectively). WAT is the primary energy depot, storing most of the bioavailable triacylglycerol molecules of the body, whereas BAT is designed for dissipating energy in the form of heat, a process also known as non-shivering thermogenesis as a defense against a cold environment. Importantly, BAT-dependent energy dissipation directly correlates with cardiometabolic health and has been postulated as an intriguing target for anti-obesity therapies. In general, adipose tissue (AT) lipid content is defined by lipid uptake and lipogenesis on one side, and, on the other side, it is defined by the breakdown of lipids and the release of fatty acids by lipolysis. The equilibrium between lipogenesis and lipolysis is important for adipocyte and general metabolic homeostasis. Overloading adipocytes with lipids causes cell stress, leading to the recruitment of immune cells and adipose tissue inflammation, which can affect the whole organism (metaflammation). The most important consequence of energy and lipid overload is obesity and associated pathophysiologies, including insulin resistance, type 2 diabetes, and cardiovascular disease. The fate of lipolysis products (fatty acids and glycerol) largely differs between AT: WAT releases fatty acids into the blood to deliver energy to other tissues (e.g., muscle). Activation of BAT, instead, liberates fatty acids that are used within brown adipocyte mitochondria for thermogenesis. The enzymes involved in lipolysis are tightly regulated by the second messenger cyclic adenosine monophosphate (cAMP), which is activated or inhibited by G protein-coupled receptors (GPCRs) that interact with heterotrimeric G proteins (G proteins). Thus, GPCRs are the upstream regulators of the equilibrium between lipogenesis and lipolysis. Moreover, GPCRs are of special pharmacological interest because about one third of the approved drugs target GPCRs. Here, we will discuss the effects of some of most studied as well as "novel" GPCRs and their ligands. We will review different facets of in vitro, ex vivo, and in vivo studies, obtained with both pharmacological and genetic approaches. Finally, we will report some possible therapeutic strategies to treat obesity employing GPCRs as primary target.
Collapse
|
4
|
Puente-Ruiz SC, Jais A. Reciprocal signaling between adipose tissue depots and the central nervous system. Front Cell Dev Biol 2022; 10:979251. [PMID: 36200038 PMCID: PMC9529070 DOI: 10.3389/fcell.2022.979251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
In humans, various dietary and social factors led to the development of increased brain sizes alongside large adipose tissue stores. Complex reciprocal signaling mechanisms allow for a fine-tuned interaction between the two organs to regulate energy homeostasis of the organism. As an endocrine organ, adipose tissue secretes various hormones, cytokines, and metabolites that signal energy availability to the central nervous system (CNS). Vice versa, the CNS is a critical regulator of adipose tissue function through neural networks that integrate information from the periphery and regulate sympathetic nerve outflow. This review discusses the various reciprocal signaling mechanisms in the CNS and adipose tissue to maintain organismal energy homeostasis. We are focusing on the integration of afferent signals from the periphery in neuronal populations of the mediobasal hypothalamus as well as the efferent signals from the CNS to adipose tissue and its implications for adipose tissue function. Furthermore, we are discussing central mechanisms that fine-tune the immune system in adipose tissue depots and contribute to organ homeostasis. Elucidating this complex signaling network that integrates peripheral signals to generate physiological outputs to maintain the optimal energy balance of the organism is crucial for understanding the pathophysiology of obesity and metabolic diseases such as type 2 diabetes.
Collapse
|
5
|
Melanocortin-5 Receptor: Pharmacology and Its Regulation of Energy Metabolism. Int J Mol Sci 2022; 23:ijms23158727. [PMID: 35955857 PMCID: PMC9369360 DOI: 10.3390/ijms23158727] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 12/12/2022] Open
Abstract
As the most recent melanocortin receptor (MCR) identified, melanocortin-5 receptor (MC5R) has unique tissue expression patterns, pharmacological properties, and physiological functions. Different from the other four MCR subtypes, MC5R is widely distributed in both the central nervous system and peripheral tissues and is associated with multiple functions. MC5R in sebaceous and preputial glands regulates lipid production and sexual behavior, respectively. MC5R expressed in immune cells is involved in immunomodulation. Among the five MCRs, MC5R is the predominant subtype expressed in skeletal muscle and white adipose tissue, tissues critical for energy metabolism. Activated MC5R triggers lipid mobilization in adipocytes and glucose uptake in skeletal muscle. Therefore, MC5R is a potential target for treating patients with obesity and diabetes mellitus. Melanocortin-2 receptor accessory proteins can modulate the cell surface expression, dimerization, and pharmacology of MC5R. This minireview summarizes the molecular and pharmacological properties of MC5R and highlights the progress made on MC5R in energy metabolism. We poInt. out knowledge gaps that need to be explored in the future.
Collapse
|
6
|
Ceddia RP, Collins S. A compendium of G-protein-coupled receptors and cyclic nucleotide regulation of adipose tissue metabolism and energy expenditure. Clin Sci (Lond) 2020; 134:473-512. [PMID: 32149342 PMCID: PMC9137350 DOI: 10.1042/cs20190579] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/17/2020] [Accepted: 02/24/2020] [Indexed: 12/15/2022]
Abstract
With the ever-increasing burden of obesity and Type 2 diabetes, it is generally acknowledged that there remains a need for developing new therapeutics. One potential mechanism to combat obesity is to raise energy expenditure via increasing the amount of uncoupled respiration from the mitochondria-rich brown and beige adipocytes. With the recent appreciation of thermogenic adipocytes in humans, much effort is being made to elucidate the signaling pathways that regulate the browning of adipose tissue. In this review, we focus on the ligand-receptor signaling pathways that influence the cyclic nucleotides, cAMP and cGMP, in adipocytes. We chose to focus on G-protein-coupled receptor (GPCR), guanylyl cyclase and phosphodiesterase regulation of adipocytes because they are the targets of a large proportion of all currently available therapeutics. Furthermore, there is a large overlap in their signaling pathways, as signaling events that raise cAMP or cGMP generally increase adipocyte lipolysis and cause changes that are commonly referred to as browning: increasing mitochondrial biogenesis, uncoupling protein 1 (UCP1) expression and respiration.
Collapse
Affiliation(s)
- Ryan P Ceddia
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
| |
Collapse
|
7
|
Yu H, Chhabra KH, Thompson Z, Jones GL, Kiran S, Shangguan G, Low MJ. Hypothalamic POMC deficiency increases circulating adiponectin despite obesity. Mol Metab 2020; 35:100957. [PMID: 32244188 PMCID: PMC7082555 DOI: 10.1016/j.molmet.2020.01.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/22/2020] [Accepted: 01/31/2020] [Indexed: 02/06/2023] Open
Abstract
Objective The steep rise in the prevalence of obesity and its related metabolic syndrome have become a major worldwide health concerns. Melanocortin peptides from hypothalamic arcuate nucleus (Arc) POMC neurons induce satiety to limit food intake. Consequently, Arc Pomc-deficient mice (ArcPomc−/−) exhibit hyperphagia and obesity. Previous studies demonstrated that the circulating levels of adiponectin, a protein abundantly produced and secreted by fat cells, negatively correlate with obesity in both rodents and humans. However, we found that ArcPomc−/− mice have increased circulating adiponectin levels despite obesity. Therefore, we investigated the physiological function and underlying mechanisms of hypothalamic POMC in regulating systemic adiponectin levels. Methods Circulating adiponectin was measured in obese ArcPomc−/− mice at ages 4–52 weeks. To determine whether increased adiponectin was a direct result of ArcPomc deficiency or a secondary effect of obesity, we examined plasma adiponectin levels in calorie-restricted mice with or without a history of obesity and in ArcPomc−/− mice before and after genetic restoration of Pomc expression in the hypothalamus. To delineate the mechanisms causing increased adiponectin in ArcPomc−/− mice, we determined sympathetic outflow to adipose tissue by assessing epinephrine, norepinephrine, and tyrosine hydroxylase protein levels and measured the circulating adiponectin in the mice after acute norepinephrine or propranolol treatments. In addition, adiponectin mRNA and protein levels were measured in discrete adipose tissue depots to ascertain which fat depots contributed the most to the high level of adiponectin in the ArcPomc−/− mice. Finally, we generated compound Adiopoq−/−:ArcPomc−/− mice and compared their growth, body composition, and glucose homeostasis to the individual knockout mouse strains and their wild-type controls. Results Obese ArcPomc−/− female mice had unexpectedly increased plasma adiponectin compared to wild-type siblings at all ages greater than 8 weeks. Despite chronic calorie restriction to achieve normal body weights, higher adiponectin levels persisted in the ArcPomc−/− female mice. Genetic restoration of Pomc expression in the Arc or acute treatment of the ArcPomc−/− female mice with melanotan II reduced adiponectin levels to control littermate values. The ArcPomc−/− mice had defective thermogenesis and decreased epinephrine, norepinephrine, and tyrosine hydroxylase protein levels in their fat pads, indicating reduced sympathetic outflow to adipose tissue. Injections of norepinephrine into the ArcPomc−/− female mice reduced circulating adiponectin levels, whereas injections of propranolol significantly increased adiponectin levels. Despite the beneficial effects of adiponectin on metabolism, the deletion of adiponectin alleles in the ArcPomc−/− mice did not exacerbate their metabolic abnormalities. Conclusion In summary, to the best of our knowledge, this study provides the first evidence that despite obesity, the ArcPomc−/− mouse model has high circulating adiponectin levels, which demonstrated that increased fat mass is not necessarily correlated with hypoadiponectinemia. Our investigation also found a previously unknown physiological pathway connecting POMC neurons via the sympathetic nervous system to circulating adiponectin, thereby shedding light on the biological regulation of adiponectin. Obese female hypothalamic-specific Pomc-deficient mice have unexpectedly elevated circulating adiponectin. Restoration of Pomc expression in the hypothalamus reduces plasma adiponectin. Low sympathetic output to subcutaneous fat depots in the Pomc-deficient mice contributes to high adiponectin levels. Deletion of adiponectin in hypothalamic-specific Pomc-deficient mice does not alter their metabolic phenotype.
Collapse
Affiliation(s)
- Hui Yu
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA.
| | - Kavaljit H Chhabra
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA; Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Zoe Thompson
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Graham L Jones
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Sylee Kiran
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA; School of Literature, Science, and Arts, University of Michigan, Ann Arbor, MI, USA
| | - Gary Shangguan
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Malcolm J Low
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Li N, Cao T, Wu X, Tang M, Xiang D, Cai H. Progress in Genetic Polymorphisms Related to Lipid Disturbances Induced by Atypical Antipsychotic Drugs. Front Pharmacol 2020; 10:1669. [PMID: 32116676 PMCID: PMC7011106 DOI: 10.3389/fphar.2019.01669] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 12/20/2019] [Indexed: 12/11/2022] Open
Abstract
Metabolic side effects such as weight gain and disturbed lipid metabolism are often observed in the treatment of atypical antipsychotic drugs (AAPDs), which contribute to an excessive prevalence of metabolic syndrome among schizophrenic patients. Great individual differences are observed but the underlying mechanisms are still uncertain. Research on pharmacogenomics indicates that gene polymorphisms involved in the pathways controlling food intake and lipid metabolism may play a significant role. In this review, relevant genes (HTR2C, DRD2, LEP, NPY, MC4R, BDNF, MC4R, CNR1, INSIG2, ADRA2A) and genetic polymorphisms related to metabolic side effects of AAPDs especially dyslipidemia were summarized. Apart from clinical studies, in vitro and in vivo evidence is also analyzed to support related theories. The association of central and peripheral mechanisms is emphasized, enabling the possibility of using peripheral gene expression to predict the central status. Novel methodological development of pharmacogenomics is in urgent need, so as to provide references for individualized medication and further to shed some light on the mechanisms underlying AAPD-induced lipid disturbances.
Collapse
Affiliation(s)
- Nana Li
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Ting Cao
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Xiangxin Wu
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Mimi Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,Institute of Hospital Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Hualin Cai
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
9
|
Ailanen L, Vähätalo LH, Salomäki-Myftari H, Mäkelä S, Orpana W, Ruohonen ST, Savontaus E. Peripherally Administered Y 2-Receptor Antagonist BIIE0246 Prevents Diet-Induced Obesity in Mice With Excess Neuropeptide Y, but Enhances Obesity in Control Mice. Front Pharmacol 2018; 9:319. [PMID: 29674968 PMCID: PMC5895854 DOI: 10.3389/fphar.2018.00319] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 03/20/2018] [Indexed: 12/27/2022] Open
Abstract
Neuropeptide Y (NPY) plays an important role in the regulation of energy homeostasis in the level of central and sympathetic nervous systems (SNSs). Genetic silencing of peripheral Y2-receptors have anti-obesity effects, but it is not known whether pharmacological blocking of peripheral Y2-receptors would similarly benefit energy homeostasis. The effects of a peripherally administered Y2-receptor antagonist were studied in healthy and energy-rich conditions with or without excess NPY. Genetically obese mice overexpressing NPY in brain noradrenergic nerves and SNS (OE-NPYDβH) represented the situation of elevated NPY levels, while wildtype (WT) mice represented the normal NPY levels. Specific Y2-receptor antagonist, BIIE0246, was administered (1.3 mg/kg/day, i.p.) for 2 or 4.5 weeks to OE-NPYDβH and WT mice feeding on chow or Western diet. Treatment with Y2-receptor antagonist increased body weight gain in both genotypes on chow diet and caused metabolic disturbances (e.g., hyperinsulinemia and hypercholesterolemia), especially in WT mice. During energy surplus (i.e., on Western diet), blocking of Y2-receptors induced obesity in WT mice, whereas OE-NPYDβH mice showed reduced fat mass gain, hepatic glycogen and serum cholesterol levels relative to body adiposity. Thus, it can be concluded that with normal NPY levels, peripheral Y2-receptor antagonist has no potential for treating obesity, but oppositely may even induce metabolic disorders. However, when energy-rich diet is combined with elevated NPY levels, e.g., stress combined with an unhealthy diet, Y2-receptor antagonism has beneficial effects on metabolic status.
Collapse
Affiliation(s)
- Liisa Ailanen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland.,Drug Research Doctoral Program, University of Turku, Turku, Finland
| | - Laura H Vähätalo
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Henriikka Salomäki-Myftari
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland.,Drug Research Doctoral Program, University of Turku, Turku, Finland
| | - Satu Mäkelä
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Wendy Orpana
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Suvi T Ruohonen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Eriika Savontaus
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland.,Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| |
Collapse
|
10
|
Cero C, Razzoli M, Han R, Sahu BS, Patricelli J, Guo Z, Zaidman NA, Miles JM, O'Grady SM, Bartolomucci A. The neuropeptide TLQP-21 opposes obesity via C3aR1-mediated enhancement of adrenergic-induced lipolysis. Mol Metab 2017; 6:148-158. [PMID: 28123945 PMCID: PMC5220279 DOI: 10.1016/j.molmet.2016.10.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 10/18/2016] [Accepted: 10/22/2016] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVES Obesity is characterized by excessive fat mass and is associated with serious diseases such as type 2 diabetes. Targeting excess fat mass by sustained lipolysis has been a major challenge for anti-obesity therapies due to unwanted side effects. TLQP-21, a neuropeptide encoded by the pro-peptide VGF (non-acronymic), that binds the complement 3a receptor 1 (C3aR1) on the adipocyte membrane, is emerging as a novel modulator of adipocyte functions and a potential target for obesity-associated diseases. The molecular mechanism is still largely uncharacterized. METHODS We used a combination of pharmacological and genetic gain and loss of function approaches. 3T3-L1 and mature murine adipocytes were used for in vitro experiments. Chronic in vivo experiments were conducted on diet-induced obese wild type, β1, β2, β3-adrenergic receptor (AR) deficient and C3aR1 knockout mice. Acute in vivo lipolysis experiments were conducted on Sprague Dawley rats. RESULTS We demonstrated that TLQP-21 does not possess lipolytic properties per se. Rather, it enhances β-AR activation-induced lipolysis by a mechanism requiring Ca2+ mobilization and ERK activation of Hormone Sensitive Lipase (HSL). TLQP-21 acutely potentiated isoproterenol-induced lipolysis in vivo. Finally, chronic peripheral TLQP-21 treatment decreases body weight and fat mass in diet induced obese mice by a mechanism involving β-adrenergic and C3a receptor activation without associated adverse metabolic effects. CONCLUSIONS In conclusion, our data identify an alternative pathway modulating lipolysis that could be targeted to diminish fat mass in obesity without the side effects typically observed when using potent pro-lipolytic molecules.
Collapse
Affiliation(s)
- Cheryl Cero
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN 55455, USA
| | - Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN 55455, USA
| | - Ruijun Han
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN 55455, USA
| | - Bhavani Shankar Sahu
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN 55455, USA
| | - Jessica Patricelli
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN 55455, USA
| | - ZengKui Guo
- Endocrine Research Unit, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Department of Internal Medicine, Mayo Foundation, 5-194 Joseph, Rochester, MN 55905, USA
| | - Nathan A Zaidman
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN 55455, USA
| | - John M Miles
- Endocrine Research Unit, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Department of Internal Medicine, Mayo Foundation, 5-194 Joseph, Rochester, MN 55905, USA
| | - Scott M O'Grady
- Department of Animal Science, Integrative Biology and Physiology, University of Minnesota, 480 Haecker Hall, 1364 Eckles Avenue, St Paul, MN 55108, USA
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
11
|
Chusyd DE, Wang D, Huffman DM, Nagy TR. Relationships between Rodent White Adipose Fat Pads and Human White Adipose Fat Depots. Front Nutr 2016; 3:10. [PMID: 27148535 PMCID: PMC4835715 DOI: 10.3389/fnut.2016.00010] [Citation(s) in RCA: 235] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/26/2016] [Indexed: 01/09/2023] Open
Abstract
The objective of this review was to compare and contrast the physiological and metabolic profiles of rodent white adipose fat pads with white adipose fat depots in humans. Human fat distribution and its metabolic consequences have received extensive attention, but much of what has been tested in translational research has relied heavily on rodents. Unfortunately, the validity of using rodent fat pads as a model of human adiposity has received less attention. There is a surprisingly lack of studies demonstrating an analogous relationship between rodent and human adiposity on obesity-related comorbidities. Therefore, we aimed to compare known similarities and disparities in terms of white adipose tissue (WAT) development and distribution, sexual dimorphism, weight loss, adipokine secretion, and aging. While the literature supports the notion that many similarities exist between rodents and humans, notable differences emerge related to fat deposition and function of WAT. Thus, further research is warranted to more carefully define the strengths and limitations of rodent WAT as a model for humans, with a particular emphasis on comparable fat depots, such as mesenteric fat.
Collapse
Affiliation(s)
- Daniella E Chusyd
- Department of Nutrition Science, University of Alabama at Birmingham , Birmingham, AL , USA
| | - Donghai Wang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Derek M Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tim R Nagy
- Department of Nutrition Science, University of Alabama at Birmingham , Birmingham, AL , USA
| |
Collapse
|
12
|
Ameliorating antipsychotic-induced weight gain by betahistine: Mechanisms and clinical implications. Pharmacol Res 2016; 106:51-63. [DOI: 10.1016/j.phrs.2016.02.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/08/2016] [Accepted: 02/11/2016] [Indexed: 01/08/2023]
|
13
|
Vähätalo LH, Ruohonen ST, Mäkelä S, Kovalainen M, Huotari A, Mäkelä KA, Määttä JA, Miinalainen I, Gilsbach R, Hein L, Ailanen L, Mattila M, Eerola K, Röyttä M, Ruohonen S, Herzig KH, Savontaus E. Neuropeptide Y in the noradrenergic neurones induces obesity and inhibits sympathetic tone in mice. Acta Physiol (Oxf) 2015; 213:902-19. [PMID: 25482272 DOI: 10.1111/apha.12436] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 11/21/2014] [Accepted: 11/30/2014] [Indexed: 12/21/2022]
Abstract
AIM Neuropeptide Y (NPY) co-localized with noradrenaline in central and sympathetic nervous systems seems to play a role in the control of energy metabolism. In this study, the aim was to elucidate the effects and pathophysiological mechanisms of increased NPY in catecholaminergic neurones on accumulation of body adiposity. METHODS Transgenic mice overexpressing NPY under the dopamine-beta-hydroxylase promoter (OE-NPY(DβH) ) and wild-type control mice were followed for body weight gain and body fat content. Food intake, energy expenditure, physical activity, body temperature, serum lipid content and markers of glucose homoeostasis were monitored. Thermogenic and lipolytic responses in adipose tissues, and urine catecholamine and tissue catecholamine synthesizing enzyme levels were analysed as indices of sympathetic tone. RESULTS Homozygous OE-NPY(DβH) mice showed significant obesity accompanied with impaired glucose tolerance and insulin resistance. Increased adiposity was explained by neither increased food intake or fat absorption nor by decreased total energy expenditure or physical activity. Adipocyte hypertrophy and decreased circulating lipid levels suggested decreased lipolysis and increased lipid uptake. Brown adipose tissue thermogenic capacity was decreased and brown adipocytes filled with lipids. Enhanced response to adrenergic stimuli, downregulation of catecholamine synthesizing enzyme expressions in the brainstem and lower adrenaline excretion supported the notion of low basal catecholaminergic activity. CONCLUSION Increased NPY in catecholaminergic neurones induces obesity that seems to be a result of preferential fat storage. These results support the role of NPY as a direct effector in peripheral tissues and an inhibitor of sympathetic activity in the pathogenesis of obesity.
Collapse
Affiliation(s)
- L. H. Vähätalo
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Drug Research Doctoral Program; University of Turku; Turku Finland
- Turku Center for Disease Modeling; University of Turku; Turku Finland
| | - S. T. Ruohonen
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Turku Center for Disease Modeling; University of Turku; Turku Finland
| | - S. Mäkelä
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Turku Center for Disease Modeling; University of Turku; Turku Finland
| | - M. Kovalainen
- Faculty of Health Sciences; School of Pharmacy; Pharmaceutical Technology; University of Eastern Finland; Kuopio Finland
- Institute of Biomedicine and Biocenter of Oulu; University of Oulu; Oulu Finland
| | - A. Huotari
- Faculty of Health Sciences; School of Pharmacy; Pharmaceutical Technology; University of Eastern Finland; Kuopio Finland
- Institute of Biomedicine and Biocenter of Oulu; University of Oulu; Oulu Finland
| | - K. A. Mäkelä
- Institute of Biomedicine and Biocenter of Oulu; University of Oulu; Oulu Finland
| | - J. A. Määttä
- Turku Center for Disease Modeling; University of Turku; Turku Finland
- Department of Cell Biology and Anatomy; Institute of Biomedicine; University of Turku; Turku Finland
| | - I. Miinalainen
- Biocenter Oulu Electron Microscopy Core Facility; University of Oulu; Oulu Finland
| | - R. Gilsbach
- Institute of Experimental and Clinical Pharmacology and Toxicology and BIOSS Centre for Biological Signalling Studies; University of Freiburg; Freiburg Germany
| | - L. Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology and BIOSS Centre for Biological Signalling Studies; University of Freiburg; Freiburg Germany
| | - L. Ailanen
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Drug Research Doctoral Program; University of Turku; Turku Finland
- Turku Center for Disease Modeling; University of Turku; Turku Finland
| | - M. Mattila
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Drug Research Doctoral Program; University of Turku; Turku Finland
- Turku Center for Disease Modeling; University of Turku; Turku Finland
| | - K. Eerola
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Turku Center for Disease Modeling; University of Turku; Turku Finland
| | - M. Röyttä
- Department of Pathology; University of Turku; Turku Finland
| | - S. Ruohonen
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine; University of Turku; Turku Finland
| | - K. -H. Herzig
- Institute of Biomedicine and Biocenter of Oulu; University of Oulu; Oulu Finland
- Medical Research Center Oulu and Oulu University Hospital; Oulu Finland
| | - E. Savontaus
- Department of Pharmacology, Drug Development and Therapeutics; University of Turku; Turku Finland
- Turku Center for Disease Modeling; University of Turku; Turku Finland
- Unit of Clinical Pharmacology; Turku University Hospital; Turku Finland
| |
Collapse
|
14
|
Mulumba M, Granata R, Marleau S, Ong H. QRFP-43 inhibits lipolysis by preventing ligand-induced complex formation between perilipin A, caveolin-1, the catalytic subunit of protein kinase and hormone-sensitive lipase in 3T3-L1 adipocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:657-66. [PMID: 25677823 DOI: 10.1016/j.bbalip.2015.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 01/21/2015] [Accepted: 02/02/2015] [Indexed: 11/30/2022]
Abstract
QRFP (RFamide) peptides are neuropeptides involved in food intake and adiposity regulation in rodents. We have previously shown that QRFP-43 (43RFa) and QRFP-26 (26RFa) inhibited isoproterenol (ISO)-induced lipolysis in adipocytes. However, the antilipolytic signaling pathways activated by QRFP peptides have not been investigated. In the present study, 3T3-L1 adipocytes were used to identify the main pathways involved in QRFP-43 decreasing ISO-induced lipolysis. Our results show that QRFP-43 reduced ISO-induced phosphorylation of perilipin A (PLIN) and hormone-sensitive lipase (HSL) on Ser660 by 43 and 25%, respectively, but increased Akt phosphorylation by 44%. However, the inhibition of phosphodiesterase 3B (PDE3B), a regulator of lipolysis activated by Akt, did not reverse the antilipolytic effect of QRFP-43. PDE3B inhibition reversed the decrease of Ser660 HSL phosphorylation associated with QRFP-43 antilipolytic effect. QRFP-43 also prevented PKC activation and ISO-induced Src kinases activation leading to the inhibition of the caveolin-1 (CAV-1) translocation on lipid droplets. Indeed, QRFP-43 attenuated phorbol 12-myristate 13-acetate-induced lipolysis and ISO-induced extracellular signal-regulated and Src kinases by 28, 37 and 48%, respectively. The attenuation of ISO-induced lipolysis by QRFP-43 was associated with a decrease of phosphorylated Ser660 HSL, PKA-catalytic (PKA-c) subunit and CAV-1 translocation on lipid droplets by 37, 50 and 46%, respectively. The decrease in ISO-induced CAV-1 and PKA-c translocation was associated with a reduction of PLIN phosphorylation by 44% in QRFP-43-treated adipocytes. These results suggest that QRFP-43 attenuated ISO-induced lipolysis by preventing the formation of an active complex on lipid droplets and the activation of Src kinases and PKC.
Collapse
Affiliation(s)
- Mukandila Mulumba
- Faculty of Pharmacy, Université de Montréal C.P. 6128, Succursale Centre-Ville, Montréal, Québec H3C 3J7, Canada
| | - Riccarda Granata
- Laboratory of Molecular and Cellular Endocrinology, Department of Medical Sciences, University of Turin, Corso Dogliotti 14, Turin 10126, Italy
| | - Sylvie Marleau
- Faculty of Pharmacy, Université de Montréal C.P. 6128, Succursale Centre-Ville, Montréal, Québec H3C 3J7, Canada
| | - Huy Ong
- Faculty of Pharmacy, Université de Montréal C.P. 6128, Succursale Centre-Ville, Montréal, Québec H3C 3J7, Canada.
| |
Collapse
|
15
|
Tang HN, Man XF, Liu YQ, Guo Y, Tang AG, Liao EY, Zhou HD. Dose-dependent effects of neuropeptide Y on the regulation of preadipocyte proliferation and adipocyte lipid synthesis via the PPARγ pathways. Endocr J 2015. [PMID: 26211472 DOI: 10.1507/endocrj.ej15-0133] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The objective of this study was to investigate the impact of neuropeptide Y (NPY) on preadipocyte proliferation and differentiation. Preadipocytes were incubated with a range of concentrations of NPY (10(-15)M - 10(-7)M). After NPY-induced differentiation, the extent of preadipocyte adipogenesis was evaluated. The expressions levels of related adipocyte markers such as PPARγ, C/EBPα and DLK-1 were examined by real-time PCR (RT-PCR) or western blot analysis. Furthermore, the mitogen-activated protein kinase (MAPK) signaling pathway proteins were also analyzed by western blot. Our results showed that low doses of NPY stimulated preadipocyte viability and proliferation, while high NPY doses inhibited cell viability. At high concentrations of NPY significantly promoted lipid accumulation and increased the size of lipid droplets. DLK-1 mRNA expression was inhibited, but the expression levels of PPARγ and C/EBPα were increased during differentiation with the presence of high concentration of NPY. High-dose NPY also suppressed the phosphorylation of the extracellular signal-regulated kinase (ERK) 1/2 protein. We conclude that NPY has a biphasic effect on preadipocyte proliferation. A high dose inhibits the proliferation of 3T3-L1 cell while promotes adipocyte differentiation, increasing lipid accumulation especially enlarged lipid droplets' size. NPY may lead to a better understanding for drug development to prevent hyperplastic obesity and hypertrophic obesity.
Collapse
Affiliation(s)
- Hao-Neng Tang
- Institute of Metabolism and Endocrinology, the Second Xiang-Ya Hospital of Central South University, Changsha 410011, China
| | | | | | | | | | | | | |
Collapse
|
16
|
Park S, Fujishita C, Komatsu T, Kim SE, Chiba T, Mori R, Shimokawa I. NPY antagonism reduces adiposity and attenuates age-related imbalance of adipose tissue metabolism. FASEB J 2014; 28:5337-48. [PMID: 25205743 DOI: 10.1096/fj.14-258384] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
An orexigenic hormone, neuropeptide Y (NPY), plays a role not only in the hypothalamic regulation of appetite, but also in the peripheral regulation of lipid metabolism. However, the intracellular mechanisms triggered by NPY to regulate lipid metabolism are poorly understood. Here we report that NPY deficiency reduces white adipose tissue (WAT) mass and ameliorates the age-related imbalance of adipose tissue metabolism in mice. Gene expression involved in adipogenesis/lipogenesis was found to decrease, whereas proteins involved in lipolysis increased in gonadal WAT (gWAT) of NPY-knockout mice. These changes were associated with an activated SIRT1- and PPARγ-mediated pathway. Moreover, the age-related decrease of de novo lipogenesis in gWAT and thermogenesis in inguinal WAT was inhibited by NPY deficiency. Further analysis using 3T3-L1 cells showed that NPY inhibited lipolysis through the Y1 receptor and enhanced lipogenesis following a reduction in cAMP response element-binding protein (CREB) and SIRT1 protein expression. Therefore, NPY appears to act as a key regulator of adipose tissue metabolism via the CREB-SIRT1 signaling pathway. Taken together, NPY deficiency reduces adiposity and ameliorates the age-related imbalance of adipose tissue metabolism, suggesting that antagonism of NPY may be a promising target for drug development to prevent age-related metabolic diseases.
Collapse
Affiliation(s)
- Seongjoon Park
- Department of Pathology, Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, Nagasaki City, Japan
| | - Chika Fujishita
- Department of Pathology, Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, Nagasaki City, Japan
| | - Toshimitsu Komatsu
- Department of Pathology, Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, Nagasaki City, Japan
| | - Sang Eun Kim
- Department of Pathology, Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, Nagasaki City, Japan
| | - Takuya Chiba
- Department of Pathology, Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, Nagasaki City, Japan
| | - Ryoichi Mori
- Department of Pathology, Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, Nagasaki City, Japan
| | - Isao Shimokawa
- Department of Pathology, Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, Nagasaki City, Japan
| |
Collapse
|
17
|
Zhang W, Cline MA, Gilbert ER. Hypothalamus-adipose tissue crosstalk: neuropeptide Y and the regulation of energy metabolism. Nutr Metab (Lond) 2014; 11:27. [PMID: 24959194 PMCID: PMC4066284 DOI: 10.1186/1743-7075-11-27] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 06/02/2014] [Indexed: 01/24/2023] Open
Abstract
Neuropeptide Y (NPY) is an orexigenic neuropeptide that plays a role in regulating adiposity by promoting energy storage in white adipose tissue and inhibiting brown adipose tissue activation in mammals. This review describes mechanisms underlying NPY's effects on adipose tissue energy metabolism, with an emphasis on cellular proliferation, adipogenesis, lipid deposition, and lipolysis in white adipose tissue, and brown fat activation and thermogenesis. In general, NPY promotes adipocyte differentiation and lipid accumulation, leading to energy storage in adipose tissue, with effects mediated mainly through NPY receptor sub-types 1 and 2. This review highlights hypothalamus-sympathetic nervous system-adipose tissue innervation and adipose tissue-hypothalamus feedback loops as pathways underlying these effects. Potential sources of NPY that mediate adipose effects include the bloodstream, sympathetic nerve terminals that innervate the adipose tissue, as well as adipose tissue-derived cells. Understanding the role of central vs. peripherally-derived NPY in whole-body energy balance could shed light on mechanisms underlying the pathogenesis of obesity. This information may provide some insight into searching for alternative therapeutic strategies for the treatment of obesity and associated diseases.
Collapse
Affiliation(s)
- Wei Zhang
- 3200 Litton-Reaves, Animal & Poultry Sciences Department, Virginia Tech, Blacksburg, VA 24061-0306, USA
| | - Mark A Cline
- 3200 Litton-Reaves, Animal & Poultry Sciences Department, Virginia Tech, Blacksburg, VA 24061-0306, USA
| | - Elizabeth R Gilbert
- 3200 Litton-Reaves, Animal & Poultry Sciences Department, Virginia Tech, Blacksburg, VA 24061-0306, USA
| |
Collapse
|
18
|
Bulloch JM, Daly CJ. Autonomic nerves and perivascular fat: interactive mechanisms. Pharmacol Ther 2014; 143:61-73. [PMID: 24560685 DOI: 10.1016/j.pharmthera.2014.02.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 02/03/2014] [Indexed: 12/31/2022]
Abstract
The evidence describing the autonomic innervation of body fat is reviewed with a particular focus on the role of the sympathetic neurotransmitters. In compiling the evidence, a strong case emerges for the interaction between autonomic nerves and perivascular adipose tissue (PVAT). Adipocytes have been shown to express receptors for neurotransmitters released from nearby sympathetic varicosities such as adrenoceptors (ARs), purinoceptors and receptors for neuropeptide Y (NPY). Noradrenaline can modulate both lipolysis (via α2- and β3-ARs) and lipogenesis (via α1- and β3-ARs). ATP can inhibit lipolysis (via P1 purinoceptors) or stimulate lipolysis (via P2y purinoceptors). NPY, which can be produced by adipocytes and sympathetic nerves, inhibits lipolysis. Thus the sympathetic triad of transmitters can influence adipocyte free fatty acid (FFA) content. Substance P (SP) released from sensory nerves has also been shown to promote lipolysis. Therefore, we propose a mechanism whereby sympathetic neurotransmission can simultaneously activate smooth muscle cells in the tunica media to cause vasoconstriction and alter FFA content and release from adjacent adipocytes in PVAT. The released FFA can influence endothelial function. Adipocytes also release a range of vasoactive substances, both relaxing and contractile factors, including adiponectin and reactive oxygen species. The action of adipokines (such as adiponectin) and reactive oxygen species (ROS) on cells of the vascular adventitia and nerves has yet to be fully elucidated. We hypothesise a strong link between PVAT and autonomic fibres and suggest that this poorly understood relationship is extremely important for normal vascular function and warrants a detailed study.
Collapse
Affiliation(s)
- Janette M Bulloch
- School of Science, University of the West of Scotland, Hamilton ML3 0JB, Scotland.
| | - Craig J Daly
- School of Life Sciences, University of Glasgow, Glasgow G128QQ, Scotland.
| |
Collapse
|
19
|
Zhang W, Sumners LH, Siegel PB, Cline MA, Gilbert ER. Quantity of glucose transporter and appetite-associated factor mRNA in various tissues after insulin injection in chickens selected for low or high body weight. Physiol Genomics 2013; 45:1084-94. [DOI: 10.1152/physiolgenomics.00102.2013] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chickens from lines selected for low (LWS) or high (HWS) body weight differ by 10-fold in body weight at 56 days old with differences in food intake, glucose regulation, and body composition. To evaluate if there are differences in appetite-regulatory factor and glucose transporter ( GLUT) mRNA that are accentuated by hypoglycemia, blood glucose was measured, and hypothalamus, liver, pectoralis major, and abdominal fat collected at 90 days of age from female HWS and LWS chickens, and reciprocal crosses, HL and LH, at 60 min after intraperitoneal injection of insulin. Neuropeptide Y ( NPY) and receptor ( NPYR) subtypes 1 and 5 mRNA were greater in LWS compared with HWS hypothalamus ( P < 0.05), but greater in HWS than LWS in fat ( P < 0.05). Expression of NPYR2 was greater in LWS than HWS in pectoralis major ( P < 0.05). There was greater expression in HWS than LWS for GLUT1 in hypothalamus and liver ( P < 0.05), GLUT2 in fat and liver ( P < 0.05), and GLUT9 in liver ( P < 0.05). Insulin was associated with reduced blood glucose in all populations ( P < 0.05) and reduced mRNA of insulin receptor ( IR) and GLUT 2 and 3 in liver ( P < 0.05). There was heterosis for mRNA, most notably NPYR1 (−78%) and NPYR5 (−81%) in fat and GLUT2 (−70%) in liver. Results suggest that NPY and GLUTs are associated with differences in energy homeostasis in LWS and HWS. Reduced GLUT and IR mRNA after insulin injection suggest a compensatory mechanism to prevent further hypoglycemia.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia
| | - Lindsay H. Sumners
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia
| | - Paul B. Siegel
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia
| | - Mark A. Cline
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia
| | | |
Collapse
|
20
|
Rodrigues AR, Almeida H, Gouveia AM. Alpha-MSH signalling via melanocortin 5 receptor promotes lipolysis and impairs re-esterification in adipocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1267-75. [PMID: 24046867 DOI: 10.1016/j.bbalip.2013.04.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The melanocortin system has a clear effect on the mobilisation of stored lipids in adipocytes. The aim of the current study was to investigate the role of melanocortin 5 receptor (MC5R) on alpha-melanocyte-stimulating hormone (alpha-MSH)-induced lipolysis in 3T3-L1 adipocytes. To this end, MC5R expression was decreased by small interfering RNA (siRNA), which significantly impaired the alpha-MSH stimulation of lipolysis, as determined by glycerol and nonesterified fatty-acid (NEFA) quantification. The functional role of alpha-MSH/MC5R on triglyceride (TG) hydrolysis was mediated by hormone-sensitive lipase (HSL), adipose triglyceride lipase (ATGL), perilipin 1 (PLIN1) and acetyl-CoA carboxylase (ACC). Immunofluorescence microscopy revealed that phosphorylated HSL clearly surrounded lipid droplets in alpha-MSH-stimulated adipocytes, whereas PLIN1 left the immediate periphery of lipids. These observations were lost when the expression of MC5R was suppressed. In 3T3-L1 adipocytes, alpha-MSH-activated MC5R signals through the cAMP/PKA and MAPK/ERK1/2 pathways. PKA was fundamental for HSL and PLIN1 activation and lipolysis regulation. ERK1/2 inhibition strongly interfered with the release of NEFAs but not glycerol. In addition, the intracellular TG levels, which were decreased after MC5R activation, were restored after ERK1/2 inhibition, indicating that these kinases are involved in NEFA re-esterification rather than lipolysis regulation. This notion is also supported by the observation that the alpha-MSH-mediated activation of phosphoenolpyruvate carboxykinase (PEPCK) was abolished in the presence of ERK1/2 inhibitors. Altogether, these results indicate that alpha-MSH-activated MC5R regulates two tightly coupled pathways in adipocytes: lipolysis and re-esterification. The global effect is a decrease in adipocyte fat mass, which is important for strategies to ameliorate obesity.
Collapse
Affiliation(s)
- Adriana R Rodrigues
- Department of Experimental Biology, Faculty of Medicine of Porto, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal
| | | | | |
Collapse
|
21
|
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
|
22
|
Smitka K, Papezova H, Vondra K, Hill M, Hainer V, Nedvidkova J. The role of "mixed" orexigenic and anorexigenic signals and autoantibodies reacting with appetite-regulating neuropeptides and peptides of the adipose tissue-gut-brain axis: relevance to food intake and nutritional status in patients with anorexia nervosa and bulimia nervosa. Int J Endocrinol 2013; 2013:483145. [PMID: 24106499 PMCID: PMC3782835 DOI: 10.1155/2013/483145] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 07/11/2013] [Indexed: 12/13/2022] Open
Abstract
Eating disorders such as anorexia (AN) and bulimia nervosa (BN) are characterized by abnormal eating behavior. The essential aspect of AN is that the individual refuses to maintain a minimal normal body weight. The main features of BN are binge eating and inappropriate compensatory methods to prevent weight gain. The gut-brain-adipose tissue (AT) peptides and neutralizing autoantibodies play an important role in the regulation of eating behavior and growth hormone release. The mechanisms for controlling food intake involve an interplay between gut, brain, and AT. Parasympathetic, sympathetic, and serotoninergic systems are required for communication between brain satiety centre, gut, and AT. These neuronal circuits include neuropeptides ghrelin, neuropeptide Y (NPY), peptide YY (PYY), cholecystokinin (CCK), leptin, putative anorexigen obestatin, monoamines dopamine, norepinephrine (NE), serotonin, and neutralizing autoantibodies. This extensive and detailed report reviews data that demonstrate that hunger-satiety signals play an important role in the pathogenesis of eating disorders. Neuroendocrine dysregulations of the AT-gut-brain axis peptides and neutralizing autoantibodies may result in AN and BN. The circulating autoantibodies can be purified and used as pharmacological tools in AN and BN. Further research is required to investigate the orexigenic/anorexigenic synthetic analogs and monoclonal antibodies for potential treatment of eating disorders in clinical practice.
Collapse
Affiliation(s)
- Kvido Smitka
- Institute of Endocrinology, Laboratory of Clinical and Experimental Neuroendocrinology, Narodni 8, 116 94 Prague 1, Czech Republic
| | - Hana Papezova
- Psychiatric Clinic, First Faculty of Medicine, Charles University, Ke Karlovu 11, 121 08 Prague 2, Czech Republic
| | - Karel Vondra
- Institute of Endocrinology, Laboratory of Clinical and Experimental Neuroendocrinology, Narodni 8, 116 94 Prague 1, Czech Republic
| | - Martin Hill
- Institute of Endocrinology, Laboratory of Clinical and Experimental Neuroendocrinology, Narodni 8, 116 94 Prague 1, Czech Republic
| | - Vojtech Hainer
- Institute of Endocrinology, Laboratory of Clinical and Experimental Neuroendocrinology, Narodni 8, 116 94 Prague 1, Czech Republic
| | - Jara Nedvidkova
- Institute of Endocrinology, Laboratory of Clinical and Experimental Neuroendocrinology, Narodni 8, 116 94 Prague 1, Czech Republic
- *Jara Nedvidkova:
| |
Collapse
|
23
|
Ruohonen ST, Pesonen U, Savontaus E. Neuropeptide Y in the noradrenergic neurons induces the development of cardiometabolic diseases in a transgenic mouse model. Indian J Endocrinol Metab 2012; 16:S569-S576. [PMID: 23565492 PMCID: PMC3602986 DOI: 10.4103/2230-8210.105574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Neuropeptide Y (NPY) is a neuropeptide widely expressed in the brain and a peptide transmitter of sympathetic nervous system (SNS) co-released with noradrenaline (NA) in prolonged stress. Association of a gain-of-function polymorphism in the human NPY gene with dyslipideamia, diabetes and vascular diseases suggests that increased NPY plays a role in the pathogenesis of the metabolic syndrome in humans. In the hypothalamus, NPY plays an established role in the regulation of body energy homeostasis. However, the effects of NPY elsewhere in the brain and in the SNS are less explored. In order to understand the role of NPY co-expressed with NA in the sympathetic nerves and brain noradrenergic neurons, a novel mouse model overexpressing NPY in noradrenergic neurons was generated. The mouse displays metabolic defects such as increased adiposity, hepatosteatosis, and impaired glucose tolerance as well as stress-related hypertension and increased susceptibility to vascular wall hypertrophy. The mouse phenotype closely reflects the findings of the several association studies with human NPY gene polymorphisms, and fits with the previous work on the effects of stress-induced NPY release on metabolism and vasculature. Thus, in addition of promoting feeding and obesity in the hypothalamus, NPY expressed in the noradrenergic neurons in the brain and in the SNS induces the development of cardiometabolic diseases.
Collapse
Affiliation(s)
- Suvi T. Ruohonen
- Department of Pharmacology, Drug Development and Therapeutics, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Ullamari Pesonen
- Department of Pharmacology, Drug Development and Therapeutics, Finland
| | - Eriika Savontaus
- Department of Pharmacology, Drug Development and Therapeutics, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| |
Collapse
|
24
|
Diet-induced obesity in mice overexpressing neuropeptide y in noradrenergic neurons. INTERNATIONAL JOURNAL OF PEPTIDES 2012; 2012:452524. [PMID: 23118773 PMCID: PMC3483820 DOI: 10.1155/2012/452524] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 09/06/2012] [Indexed: 12/04/2022]
Abstract
Neuropeptide Y (NPY) is a neurotransmitter associated with feeding and obesity. We have constructed an NPY transgenic mouse model (OE-NPYDBH mouse), where targeted overexpression leads to increased levels of NPY in noradrenergic and adrenergic neurons. We previously showed that these mice become obese on a normal chow. Now we aimed to study the effect of a Western-type diet in OE-NPYDBH and wildtype (WT) mice, and to compare the genotype differences in the development of obesity, insulin resistance, and diabetes. Weight gain, glucose, and insulin tolerance tests, fasted plasma insulin, and cholesterol levels were assayed. We found that female OE-NPYDBH mice gained significantly more weight without hyperphagia or decreased activity, and showed larger white and brown fat depots with no difference in UCP-1 levels. They also displayed impaired glucose tolerance and decreased insulin sensitivity. OE-NPYDBH and WT males gained weight robustly, but no difference in the degree of adiposity was observed. However, 40% of OE-NPYDBH but none of the WT males developed hyperglycaemia while on the diet. The present study shows that female OE-NPYDBH mice were not protected from the obesogenic effect of the diet suggesting that increased NPY release may predispose females to a greater risk of weight gain under high caloric conditions.
Collapse
|
25
|
Møller CL, Raun K, Jacobsen ML, Pedersen TÅ, Holst B, Conde-Frieboes KW, Wulff BS. Characterization of murine melanocortin receptors mediating adipocyte lipolysis and examination of signalling pathways involved. Mol Cell Endocrinol 2011; 341:9-17. [PMID: 21616121 DOI: 10.1016/j.mce.2011.03.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 02/21/2011] [Accepted: 03/15/2011] [Indexed: 11/15/2022]
Abstract
The melanocortin receptors (MCRs) belong to the G-protein coupled receptors (family A). So far, 5 different subtypes have been described (MC1R-MC5R) and of these MC2R and MC5R have been proposed to act directly in adipocytes and regulate lipolysis in rodents. Using ACTH and α-melanocyte stimulating hormone (α-MSH) generated from proopiomelanocortin (POMC), as well as synthetic MSH analogues to stimulate lipolysis in murine 3T3-L1 adipocytes it is shown that MC2R and MC5R are lipolytic mediators in differentiated 3T3-L1 adipocytes. Involvement of cAMP, phosphorylated extracellular signal-regulated kinase (ERK) 1/2, protein kinase B (PKB), adenosine 5' monophosphate activated protein kinase (AMPK) and Jun-amino-terminal kinase (JNK) in MCR mediated lipolysis were studied. Interestingly, results obtained in 3T3-L1 cells suggest that lipolysis stimulated by α-MSH, NDP-α-MSH, MT-II, SHU9119 and PG-901 is mediated through MC5R in a cAMP independent manner. Finally, we identify essential differences in MCR mediated lipolysis when using 3T3-L1 cells compared to primary adipocytes.
Collapse
MESH Headings
- 3T3-L1 Cells
- Adipocytes/drug effects
- Adipocytes/metabolism
- Adipogenesis
- Adrenocorticotropic Hormone/pharmacology
- Adrenocorticotropic Hormone/physiology
- Animals
- Binding, Competitive
- Cyclic AMP/metabolism
- Epididymis/cytology
- Epididymis/metabolism
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Fatty Acids, Nonesterified/metabolism
- Gene Expression
- Hormones/pharmacology
- Hormones/physiology
- Lipolysis
- MAP Kinase Signaling System
- Male
- Melanocortins/pharmacology
- Melanocortins/physiology
- Mice
- Mice, Inbred C57BL
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor, Melanocortin, Type 2/agonists
- Receptor, Melanocortin, Type 2/genetics
- Receptor, Melanocortin, Type 2/metabolism
- Receptors, Melanocortin/genetics
- Receptors, Melanocortin/metabolism
Collapse
|
26
|
The neuropeptide Y system: Pathophysiological and therapeutic implications in obesity and cancer. Pharmacol Ther 2011; 131:91-113. [DOI: 10.1016/j.pharmthera.2011.03.011] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Accepted: 03/07/2011] [Indexed: 12/28/2022]
|
27
|
Chaves VE, Frasson D, Kawashita NH. Several agents and pathways regulate lipolysis in adipocytes. Biochimie 2011; 93:1631-40. [PMID: 21658426 DOI: 10.1016/j.biochi.2011.05.018] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 05/23/2011] [Indexed: 01/01/2023]
Abstract
Adipose tissue is the only tissue capable of hydrolyzing its stores of triacylglycerol (TAG) and of mobilizing fatty acids and glycerol in the bloodstream so that they can be used by other tissues. The full hydrolysis of TAG depends on the activity of three enzymes, adipose triglyceride lipase (ATGL), hormone-sensitive lipase (HSL) and monoacylglycerol lipase, each of which possesses a distinct regulatory mechanism. Although more is known about HSL than about the other two enzymes, it has recently been shown that HLS and ATGL can be activated simultaneously, such that the mechanism that enables HSL to access the surface of lipid droplets also permits the stimulation of ATGL. The classical pathway of lipolysis activation in adipocytes is cAMP-dependent. The production of cAMP is modulated by G-protein-coupled receptors of the Gs/Gi family and cAMP degradation is regulated by phosphodiesterase. However, other pathways that activate TAG hydrolysis are currently under investigation. Lipolysis can also be started by G-protein-coupled receptors of the Gq family, through molecular mechanisms that involve phospholipase C, calmodulin and protein kinase C. There is also evidence that increased lipolytic activity in adipocytes occurs after stimulation of the mitogen-activated protein kinase pathway or after cGMP accumulation and activation of protein kinase G. Several agents contribute to the control of lipolysis in adipocytes by modulating the activity of HSL and ATGL. In this review, we have summarized the signalling pathways activated by several agents involved in the regulation of TAG hydrolysis in adipocytes.
Collapse
Affiliation(s)
- Valéria Ernestânia Chaves
- Department of Basic Sciences in Health, Federal University of Mato Grosso, Cuiabá, Mato Grosso, Brazil
| | | | | |
Collapse
|
28
|
Wang SCM, Myers SA, Eriksson NA, Fitzsimmons RL, Muscat GEO. Nr4a1 siRNA expression attenuates α-MSH regulated gene expression in 3T3-L1 adipocytes. Mol Endocrinol 2011; 25:291-306. [PMID: 21239615 DOI: 10.1210/me.2010-0231] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Several recent investigations have underscored the growing role of melanocortin signaling in the peripheral regulation of lipid, glucose, and energy homeostasis. In addition, the melanocortins play a critical role in the central control of satiety. These observations, and the latest reports highlighting the emerging role of the nuclear hormone receptor (NR) 4A subgroup in metabolism, have prompted us to investigate the cross talk between [Nle(4), d-Phe(7)] (NDP)-α-MSH and Nr4a signaling in adipose. We have shown that NDP-MSH strikingly and preferentially induces the expression of the NR4A subgroup (but not any other members of the NR superfamily) in differentiated 3T3-L1 adipocytes. Utilization of quantitative PCR on custom-designed metabolic TaqMan low-density arrays identified the concomitant and marked induction of the mRNAs encoding Il-6, Cox2, Pdk4, and Pck-1 after NDP-MSH treatment. Similar experiments demonstrated that the mRNA expression profile induced by cAMP and NDP-MSH treatment displayed unique but also overlapping properties and suggested that melanocortin-mediated induction of gene expression involves cAMP-dependent and -independent signaling. Nr4a1/Nur77 small interfering RNA (siRNA) expression suppressed NDP-MSH-mediated induction of Nr4a1/Nur77 and Nr4a3/Nor-1 (but not Nr4a2/Nurr1). Moreover, expression of the siRNA-attenuated NDP-MSH mediated induction of the mRNAs encoding Il-6, Cox2/Ptgs2, and Pck-1 expression. In addition, Nur77 siRNA expression attenuated NDP-MSH-mediated glucose uptake. In vivo, ip administration of NDP-MSH to C57 BL/6J (male) mice significantly induced the expression of the mRNA encoding Nur77 and increased IL-6, Cox2, Pck1, and Pdk4 mRNA expression in (inguinal) adipose tissue. We conclude that Nur77 expression is necessary for MSH-mediated induction of gene expression in differentiated adipocytes. Furthermore, this study demonstrates cross talk between MSH and Nr4a signaling in adipocytes.
Collapse
Affiliation(s)
- S-C Mary Wang
- Obesity Research Centre, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | | | | | | | | |
Collapse
|
29
|
Zhang L, Lee NJ, Nguyen AD, Enriquez RF, Riepler SJ, Stehrer B, Yulyaningsih E, Lin S, Shi YC, Baldock PA, Herzog H, Sainsbury A. Additive actions of the cannabinoid and neuropeptide Y systems on adiposity and lipid oxidation. Diabetes Obes Metab 2010; 12:591-603. [PMID: 20590734 DOI: 10.1111/j.1463-1326.2009.01193.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIMS Energy homeostasis is regulated by a complex interaction of molecules and pathways, and new antiobesity treatments are likely to require multiple pharmacological targeting of anorexigenic or orexigenic pathways to achieve effective loss of excess body weight and adiposity. Cannabinoids, acting via the cannabinoid-1 (CB1) receptor, and neuropeptide Y (NPY) are important modulators of feeding behaviour, energy metabolism and body composition. We investigated the interaction of CB1 and NPY in the regulation of energy homeostasis, hypothesizing that dual blockade of CB1 and NPY signalling will induce greater weight and/or fat loss than that induced by single blockade of either system alone. METHODS We studied the effects of the CB1 antagonist Rimonabant on food intake, body weight, body composition, energy metabolism and bone physiology in wild-type (WT) and NPY knockout (NPY(-/-)) mice. Rimonabant was administered orally at 10 mg/kg body weight twice per day for 3 weeks. Oral Rimonabant was delivered voluntarily to mice via a novel method enabling studies to be carried out in the absence of gavage-induced stress. RESULTS Mice with dual blockade of CB1 and NPY signalling (Rimonabant-treated NPY(-/-) mice) exhibited greater reductions in body weight and adiposity than mice with single blockade of either system alone (Rimonabant-treated WT or vehicle-treated NPY(-/-) mice). These changes occurred without loss of lean tissue mass or bone mass. Furthermore, Rimonabant-treated NPY(-/-) mice showed a lower respiratory exchange ratio than that seen in Rimonabant-treated WT or vehicle-treated NPY(-/-) mice, suggesting that this additive effect of dual blockade of CB1 and NPY involves promotion of lipid oxidation. On the other hand, energy expenditure and physical activity were comparable amongst all treatment groups. Interestingly, Rimonabant similarly and transiently reduced spontaneous and fasting-induced food intake in WT and NPY(-/-) mice in the first hour after administration only, suggesting independent regulation of feeding by CB1 and NPY signalling. In contrast, Rimonabant increased serum corticosterone levels in WT mice, but this effect was not seen in NPY(-/-) mice, indicating that NPY signalling may be required for effects of CB1 on the hypothalamo-pituitary-adrenal axis. CONCLUSIONS Dual blockade of CB1 and NPY signalling leads to additive reductions in body weight and adiposity without concomitant loss of lean body mass or bone mass. An additive increase in lipid oxidation in dual CB1 and NPY blockade may contribute to the effect on adiposity. These findings open new avenues for more effective treatment of obesity via dual pharmacological manipulations of the CB1 and NPY systems.
Collapse
Affiliation(s)
- L Zhang
- Neuroscience Research Program, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst, Sydney, NSW 2010, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Shrestha YB, Vaughan CH, Smith BJ, Song CK, Baro DJ, Bartness TJ. Central melanocortin stimulation increases phosphorylated perilipin A and hormone-sensitive lipase in adipose tissues. Am J Physiol Regul Integr Comp Physiol 2010; 299:R140-9. [PMID: 20410474 DOI: 10.1152/ajpregu.00535.2009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Norepinephrine (NE) released from the sympathetic nerves innervating white adipose tissue (WAT) is the principal initiator of lipolysis in mammals. Central WAT sympathetic outflow neurons express melanocortin 4-receptor (MC4-R) mRNA. Single central injection of melanotan II (MTII; MC3/4-R agonist) nonuniformly increases WAT NE turnover (NETO), increases interscapular brown adipose tissue (IBAT) NETO, and increases the circulating lipolytic products glycerol and free fatty acid. The WAT pads that contributed to this lipolysis were inferred from the increases in NETO. Because phosphorylation of perilipin A (p-perilipin A) and hormone-sensitive lipase are necessary for NE-triggered lipolysis, we tested whether MTII would increase these intracellular markers of lipolysis. Male Siberian hamsters received a single 3rd ventricular injection of MTII or saline. Trunk blood was collected at 0.5, 1.0, and 2.0 h postinjection from excised inguinal, retroperitoneal, and epididymal WAT (IWAT, RWAT, and EWAT, respectively) and IBAT pads. MTII increased circulating glycerol concentrations at 0.5 and 1.0 h, whereas free fatty acid concentrations were increased at 1.0 and 2.0 h. Western blot analysis showed that MTII specifically increased p-perilipin A and hormone-sensitive lipase only in fat pads that previously had MTII-induced increases in NETO. Phosphorylation increased in IWAT at all time points and IBAT at 0.5 h, but not RWAT or EWAT at any time point. These results show for the first time in rodents that p-perilipin A can serve as an in vivo, fat pad-specific indictor of lipolysis and extend our previous findings showing that central melanocortin stimulation increases WAT lipolysis.
Collapse
Affiliation(s)
- Y B Shrestha
- Department of Biology, Georgia State University, Atlanta, Georgia 30302-4010, USA
| | | | | | | | | | | |
Collapse
|
31
|
Zhang L, Macia L, Turner N, Enriquez RF, Riepler SJ, Nguyen AD, Lin S, Lee NJ, Shi YC, Yulyaningsih E, Slack K, Baldock PA, Herzog H, Sainsbury A. Peripheral neuropeptide Y Y1 receptors regulate lipid oxidation and fat accretion. Int J Obes (Lond) 2009; 34:357-73. [PMID: 19918245 DOI: 10.1038/ijo.2009.232] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Neuropeptide Y and its Y receptors are important players in the regulation of energy homeostasis. However, while their functions in feeding regulation are well recognized, functions in other critical aspects of energy homeostasis are largely unknown. To investigate the function of Y1 receptors in the regulation of energy homeostasis, we examined energy expenditure, physical activity, body composition, oxidative fuel selection and mitochondrial oxidative capacity in germline Y1(-/-) mice as well as in a conditional Y1-receptor-knockdown model in which Y1 receptors were knocked down in peripheral tissues of adult mice. RESULTS Germline Y1(-/-) mice of both genders not only exhibit a decreased respiratory exchange ratio, indicative of increased lipid oxidation, but interestingly also develop late-onset obesity. However, the increased lipid oxidation is a primary effect of Y1 deletion rather than secondary to increased adiposity, as young Y1(-/-) mice are lean and show the same effect. The mechanism behind this is likely because of increased liver and muscle protein levels of carnitine palmitoyltransferase-1 (CPT-1) and maximal activity of key enzymes involved in beta-oxidation; beta-hydroxyacyl CoA dehydrogenase (betaHAD) and medium-chain acyl-CoA dehydrogenase (MCAD), leading to increased mitochondrial capacity for fatty acid transport and oxidation. These effects are controlled by peripheral Y1-receptor signalling, as adult-onset conditional Y1 knockdown in peripheral tissues also leads to increased lipid oxidation, liver CPT-1 levels and betaHAD activity. Importantly, these mice are resistant to diet-induced obesity. CONCLUSIONS This work shows the primary function of peripheral Y1 receptors in the regulation of oxidative fuel selection and adiposity, opening up new avenues for anti-obesity treatments by targeting energy utilization in peripheral tissues rather than suppressing appetite by central effects.
Collapse
Affiliation(s)
- L Zhang
- Neuroscience Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Lakaye B, Coumans B, Harray S, Grisar T. Melanin-concentrating hormone and immune function. Peptides 2009; 30:2076-80. [PMID: 19450627 DOI: 10.1016/j.peptides.2009.05.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Revised: 05/04/2009] [Accepted: 05/05/2009] [Indexed: 10/20/2022]
Abstract
To date, melanin-concentrating hormone (MCH) has been generally considered as peptide acting almost exclusively in the central nervous system. In the present paper, we revise the experimental evidence, demonstrating that MCH and its receptors are expressed by cells of the immune system and directly influence the response of these cells in some circumstances. This therefore supports the idea that, as with other peptides, MCH could be considered as a modulator of the immune system. Moreover, we suggest that this could have important implications in several immune-mediated disorders and affirm that there is a clear need for further investigation.
Collapse
|
33
|
Watson E, Fargali S, Okamoto H, Sadahiro M, Gordon RE, Chakraborty T, Sleeman MW, Salton SR. Analysis of knockout mice suggests a role for VGF in the control of fat storage and energy expenditure. BMC PHYSIOLOGY 2009; 9:19. [PMID: 19863797 PMCID: PMC2774661 DOI: 10.1186/1472-6793-9-19] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Accepted: 10/28/2009] [Indexed: 11/29/2022]
Abstract
Background Previous studies of mixed background mice have demonstrated that targeted deletion of Vgf produces a lean, hypermetabolic mouse that is resistant to diet-, lesion-, and genetically-induced obesity. To investigate potential mechanism(s) and site(s) of action of VGF, a neuronal and endocrine secreted protein and neuropeptide precursor, we further analyzed the metabolic phenotypes of two independent VGF knockout lines on C57Bl6 backgrounds. Results Unlike hyperactive VGF knockout mice on a mixed C57Bl6-129/SvJ background, homozygous mutant mice on a C57Bl6 background were hypermetabolic with similar locomotor activity levels to Vgf+/Vgf+ mice, during day and night cycles, indicating that mechanism(s) other than hyperactivity were responsible for their increased energy expenditure. In Vgf-/Vgf- knockout mice, morphological analysis of brown and white adipose tissues (BAT and WAT) indicated decreased fat storage in both tissues, and decreased adipocyte perimeter and area in WAT. Changes in gene expression measured by real-time RT-PCR were consistent with increased fatty acid oxidation and uptake in BAT, and increased lipolysis, decreased lipogenesis, and brown adipocyte differentiation in WAT, suggesting that increased sympathetic nervous system activity in Vgf-/Vgf- mice may be associated with or responsible for alterations in energy expenditure and fat storage. In addition, uncoupling protein 1 (UCP1) and UCP2 protein levels, mitochondrial number, and mitochondrial cristae density were upregulated in Vgf-/Vgf- BAT. Using immunohistochemical and histochemical techniques, we detected VGF in nerve fibers innervating BAT and Vgf promoter-driven reporter expression in cervical and thoracic spinal ganglia that project to and innervate the chest wall and tissues including BAT. Moreover, VGF peptide levels were quantified by radioimmunoassay in BAT, and were found to be down-regulated by a high fat diet. Lastly, despite being hypermetabolic, VGF knockout mice were cold intolerant. Conclusion We propose that VGF and/or VGF-derived peptides modulate sympathetic outflow pathways to regulate fat storage and energy expenditure.
Collapse
Affiliation(s)
- Elizabeth Watson
- Department of Neuroscience, Mount Sinai School of Medicine, New York, NY, USA.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Kos K, Baker AR, Jernas M, Harte AL, Clapham JC, O'Hare JP, Carlsson L, Kumar S, McTernan PG. DPP-IV inhibition enhances the antilipolytic action of NPY in human adipose tissue. Diabetes Obes Metab 2009; 11:285-92. [PMID: 19175376 DOI: 10.1111/j.1463-1326.2008.00909.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CONTEXT Dipeptidyl peptidase IV (DPP-IV) inactivates the incretin hormone glucagon-like peptide. It can also affect the orexigenic hormone neuropeptide Y (NPY(1-36)) which is truncated by DPP-IV to NPY(3-36), as a consequence NPY's affinity changes from receptor Y1, which mediates the antilipolytic function of NPY, to other NPY receptors. Little is known whether DPP-IV inhibitors for the treatment of type 2 diabetic (T2DM) patients could influence these pathways. AIMS To investigate the in vitro effects of NPY with DPP-IV inhibition in isolated abdominal subcutaneous (AbdSc) adipocytes on fat metabolism, and assessment of NPY receptor and DPP-IV expression in adipose tissue (AT). METHODS Ex vivo human AT was taken from women undergoing elective surgery (body mass index: 27.5 (mean +/- s.d.) +/- 5 kg/m2, age: 43.7 +/- 10 years, n = 36). Isolated AbdSc adipocytes were treated with human recombinant (rh)NPY (1-100 nM) with and without DPP-IV inhibitor (1 M); glycerol release and tissue distribution of DPP-IV, Y1 and Y5 messenger RNA (mRNA) were measured and compared between lean and obese subjects. RESULTS AND CONCLUSION rhNPY reduced glycerol release, an effect that was further enhanced by co-incubation with a DPP-IV inhibitor [control: 224 (mean +/- s.e.) +/- 37 micromol/l; NPY, 100 nM: 161 +/- 27 micromol/l**; NPY 100 nM/DPP-IV inhibitor, 1 M: 127 +/- 14 micromol/l**; **p < 0.01, n = 14]. DPP-IV was expressed in AbdSc AT and omental AT with relative DPP-IV mRNA expression lower in AbdSc AT taken from obese [77 +/- 6 signal units (SU)] vs. lean subjects (186 +/- 29 SU*, n = 10). Y1 was predominantly expressed in fat and present in all fat depots but higher in obese subjects, particularly the AbdSc AT-depot (obese: 1944 +/- 111 SU vs. lean: 711 +/- 112 SU**, n = 10). NPY appears to be regulated by AT-derived DPP-IV. DPP-IV inhibitors augment the antilipolytic effect of NPY in AT. Further studies are required to show whether this explains the lack of weight loss in T2DM patients treated with DPP-IV inhibitors.
Collapse
Affiliation(s)
- K Kos
- Unit for Diabetes and Metabolism, Clinical Sciences Research Institute (CSRI), Warwick Medical School, Coventry, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zechner R, Kienesberger PC, Haemmerle G, Zimmermann R, Lass A. Adipose triglyceride lipase and the lipolytic catabolism of cellular fat stores. J Lipid Res 2009; 50:3-21. [PMID: 18952573 DOI: 10.1194/jlr.r800031-jlr200] [Citation(s) in RCA: 416] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Fatty acids (FAs) are essential components of all lipid classes and pivotal substrates for energy production in all vertebrates. Additionally, they act directly or indirectly as signaling molecules and, when bonded to amino acid side chains of peptides, anchor proteins in biological membranes. In vertebrates, FAs are predominantly stored in the form of triacylglycerol (TG) within lipid droplets of white adipose tissue. Lipid droplet-associated TGs are also found in most nonadipose tissues, including liver, cardiac muscle, and skeletal muscle. The mobilization of FAs from all fat depots depends on the activity of TG hydrolases. Currently, three enzymes are known to hydrolyze TG, the well-studied hormone-sensitive lipase (HSL) and monoglyceride lipase (MGL), discovered more than 40 years ago, as well as the relatively recently identified adipose triglyceride lipase (ATGL). The phenotype of HSL- and ATGL-deficient mice, as well as the disease pattern of patients with defective ATGL activity (due to mutation in ATGL or in the enzyme's activator, CGI-58), suggest that the consecutive action of ATGL, HSL, and MGL is responsible for the complete hydrolysis of a TG molecule. The complex regulation of these enzymes by numerous, partially uncharacterized effectors creates the "lipolysome," a complex metabolic network that contributes to the control of lipid and energy homeostasis. This review focuses on the structure, function, and regulation of lipolytic enzymes with a special emphasis on ATGL.
Collapse
Affiliation(s)
- Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Austria.
| | | | | | | | | |
Collapse
|
36
|
Bahar B, Sweeney T. Mapping of the transcription start site (TSS) and identification of SNPs in the bovine neuropeptide Y (NPY) gene. BMC Genet 2008; 9:91. [PMID: 19105820 PMCID: PMC2657160 DOI: 10.1186/1471-2156-9-91] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Accepted: 12/23/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neuropeptide Y is a key neurotransmitter of the central nervous system which plays a vital role in the feed energy homeostasis in mammals. Mutations in the regulatory and coding regions of the bovine NPY gene can potentially affect the neuronal regulation of appetite and feeding behaviour in cattle. The objectives of this experiment were to: a) fully characterize the bovine NPY gene transcript and b) identify the SNP diversity in both coding and non-coding regions of the NPY gene in a panel of Bos taurus and B. indicus cattle. RESULTS Bovine NPY gene consists of four exons (99, 188, 81 and 195 nucleotides) and three introns. The promoter region of the NPY gene consists of TATA and GC boxes which are separated from the transcription start site (TSS) by 29 and ~100 nt, respectively. Analyses of the tissue specific expression of the bovine NPY gene revealed the presence of highly abundant NPY gene transcripts in the arcuate nucleus, cerebral and cerebellar regions of the bovine brain. We identified a total of 59 SNPs in the 8.4 kb region of the bovine NPY gene. Seven out of nine total SNPs in the promoter region affect binding of putative transcription factors. A high level of nucleotide diversity was evident in the promoter regions (2.84 x 10(-3)) compared to the exonic (1.44 x 10(-3)), intronic (1.30 x 10(-3)) and 3' untranslated (1.26 x 10(-3)) regions. CONCLUSION The SNPs identified in different regions of bovine NPY gene may serve as a basis for understanding the regulation of the expression of the bovine NPY gene under a variety of physiological conditions and identification of genotypes with high feed energy conversion efficiency.
Collapse
Affiliation(s)
- Bojlul Bahar
- Cell and Molecular Biology Lab, School of Agriculture, Food Science & Veterinary Medicine, Veterinary Science Centre, University College Dublin, Belfield, Dublin 4, Ireland.
| | | |
Collapse
|
37
|
Catecholamine-induced lipolysis in adipose tissue and skeletal muscle in obesity. Physiol Behav 2008; 94:219-30. [DOI: 10.1016/j.physbeh.2008.01.002] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2007] [Revised: 01/07/2008] [Accepted: 01/07/2008] [Indexed: 12/20/2022]
|
38
|
Yang K, Guan H, Arany E, Hill DJ, Cao X. Neuropeptide Y is produced in visceral adipose tissue and promotes proliferation of adipocyte precursor cells
via
the Y1 receptor. FASEB J 2008; 22:2452-64. [DOI: 10.1096/fj.07-100735] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Kaiping Yang
- Children's Health Research InstituteUniversity of Western OntarioLondon OntarioCanada
- Department of Obstetrics and GynecologyUniversity of Western OntarioLondon OntarioCanada
- Department of Physiology and PharmacologyUniversity of Western OntarioLondon OntarioCanada
| | - Haiyan Guan
- Children's Health Research InstituteUniversity of Western OntarioLondon OntarioCanada
- Department of Obstetrics and GynecologyUniversity of Western OntarioLondon OntarioCanada
- Department of Physiology and PharmacologyUniversity of Western OntarioLondon OntarioCanada
| | - Edith Arany
- Department of Physiology and PharmacologyUniversity of Western OntarioLondon OntarioCanada
- Department of MedicineLawson Health Research InstituteUniversity of Western OntarioLondon OntarioCanada
| | - David J. Hill
- Department of Physiology and PharmacologyUniversity of Western OntarioLondon OntarioCanada
- Department of MedicineLawson Health Research InstituteUniversity of Western OntarioLondon OntarioCanada
| | - Xiang Cao
- Children's Health Research InstituteUniversity of Western OntarioLondon OntarioCanada
- Department of Obstetrics and GynecologyUniversity of Western OntarioLondon OntarioCanada
- Department of Physiology and PharmacologyUniversity of Western OntarioLondon OntarioCanada
| |
Collapse
|
39
|
Karagiannides I, Torres D, Tseng YH, Bowe C, Carvalho E, Espinoza D, Pothoulakis C, Kokkotou E. Substance P as a novel anti-obesity target. Gastroenterology 2008; 134:747-55. [PMID: 18325388 PMCID: PMC2359157 DOI: 10.1053/j.gastro.2007.12.032] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Accepted: 12/06/2007] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Substance P (SP) is an 11-amino acid peptide that belongs to the tachykinin family of peptides. SP acts in the brain and in the periphery as a neuropeptide, neurotransmitter, and hormone affecting diverse physiologic pathways, mainly via its high-affinity neurokinin-1 receptor (NK-1R). Its presence in the hypothalamus and other areas of the brain that regulate feeding as well as in the stomach and small intestine prompted us to investigate its role on appetite control and energy balance. METHODS CJ 012,255 (CJ), a SP antagonist that binds to NK-1R, was injected into lean, diet-induced obese (DIO), and genetically obese (ob/ob) mice, and its effects on body weight, adiposity, and insulin sensitivity were investigated. RESULTS CJ administration prevented weight gain and accumulation of fat after 2 weeks of high-fat feeding, whereas similar CJ treatment in obese mice (following 3 months of high-fat diet) resulted in weight loss, reduction in adiposity, and improvement of insulin sensitivity, in part because of inhibition of food intake. The effects of SP in the control of energy balance are, at least in part, leptin independent because CJ treatment was also effective in leptin-deficient mice. Peripheral SP administration resulted in a mild, dose-dependent increase in food intake, evident 3 hours post-SP injection. CONCLUSIONS CJ reduces appetite and promotes weight loss in mice. We speculate that NK-1R antagonists, already tested in clinical trials for various diseases, may represent a potential target against obesity.
Collapse
Affiliation(s)
- Iordanes Karagiannides
- Gastrointestinal Neuropeptide Center, Gastroenterology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Torres
- Gastrointestinal Neuropeptide Center, Gastroenterology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Yu-Hua Tseng
- Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Collin Bowe
- Gastrointestinal Neuropeptide Center, Gastroenterology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Eugenia Carvalho
- Gastrointestinal Neuropeptide Center, Gastroenterology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Espinoza
- Gastrointestinal Neuropeptide Center, Gastroenterology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Charalabos Pothoulakis
- Gastrointestinal Neuropeptide Center, Gastroenterology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Efi Kokkotou
- Gastrointestinal Neuropeptide Center, Gastroenterology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
40
|
Kos K, Harte AL, James S, Snead DR, O'Hare JP, McTernan PG, Kumar S. Secretion of neuropeptide Y in human adipose tissue and its role in maintenance of adipose tissue mass. Am J Physiol Endocrinol Metab 2007; 293:E1335-40. [PMID: 17785501 DOI: 10.1152/ajpendo.00333.2007] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
NPY is an important central orexigenic hormone, but little is known about its peripheral actions in human adipose tissue (AT) or its potential paracrine effects. Our objective was to examine NPY's role in AT, specifically addressing NPY protein expression, the effect of NPY on adipokine secretion, and the influence of insulin and rosiglitazone (RSG) on adipocyte-derived NPY in vitro. Ex vivo human AT was obtained from women undergoing elective surgery [age: 42.7 +/- 1.5 yr (mean +/- SE), BMI: 26.2 +/- 0.7 kg/m(2); n = 38]. Western blot analysis was used to determine NPY protein expression in AT depots. Abdominal subcutaneous (AbSc) adipocytes were isolated and treated with recombinant (rh) NPY, insulin, and RSG. NPY and adipokine levels were measured by ELISA. Our results were that NPY was localized in human AT and adipocytes and confirmed by immunohistochemistry. Depot-specific NPY expression was noted as highest in AbSc AT (1.87 +/- 0.23 ODU) compared with omental (Om; 1.03 +/- 0.15 ODU, P = 0.029) or thigh AT (Th; 1.0 +/- 0.29 ODU, P = 0.035). Insulin increased NPY secretion (control: 0.22 +/- 0.024 ng/ml; 1 nM insulin: 0.26 +/- 0.05 ng/ml; 100 nM insulin: 0.29 +/- 0.04 ng/ml; 1,000 nM insulin: 0.3 +/- 0.04 ng/ml; P < 0.05, n = 13), but cotreatment of RSG (10 nM) with insulin (100 nM) had no effect on NPY secretion. Furthermore, adipocyte treatment with rh-NPY downregulated leptin secretion (control: 6.99 +/- 0.89 ng/ml; 1 nmol/l rh-NPY: 4.4 +/- 0.64 ng/ml; 10 nmol/l rh-NPY: 4.3 +/- 0.61 ng/ml, 100 nmol/l rh-NPY: 4.2 +/- 0.67 ng/ml; P < 0.05, n = 10) but had no effect on adiponectin or TNF-alpha secretion. We conclude that NPY is expressed and secreted by human adipocytes. NPY secretion is stimulated by insulin, but this increment was limited by cotreatment with RSG. NPY's antilipolytic action may promote an increase in adipocyte size in hyperinsulinemic conditions. Adipose-derived NPY mediates reduction of leptin secretion and may have implications for central feedback of adiposity signals.
Collapse
Affiliation(s)
- Katarina Kos
- Unit for Diabetes and Metabolism, Clinical Sciences Research Institute, Warwick Medical School, Coventry, UK
| | | | | | | | | | | | | |
Collapse
|
41
|
Brito MN, Brito NA, Baro DJ, Song CK, Bartness TJ. Differential activation of the sympathetic innervation of adipose tissues by melanocortin receptor stimulation. Endocrinology 2007; 148:5339-47. [PMID: 17702843 DOI: 10.1210/en.2007-0621] [Citation(s) in RCA: 144] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Melanocortins are implicated in the control of energy intake/expenditure. Centrally administered melanotan II (MTII), a synthetic melanocortin 3/4-receptor agonist, decreases adiposity beyond that accountable by food intake decreases. Melanocortin-4 receptor (MC4-R) mRNA is expressed on sympathetic nervous system (SNS) outflow neurons to white adipose tissue (WAT) in Siberian hamsters, suggesting a role in lipid mobilization. Therefore, we tested whether third ventricular injections of MTII increased sympathetic drive to WAT and interscapular brown adipose tissue (IBAT) using norepinephrine turnover (NETO) as a measure of sympathetic drive. We also tested for MTII-induced changes in lipolysis-related WAT gene expression (beta3-adrenoceptors, hormone sensitive lipase) and IBAT thermogenesis (beta3-adrenoceptor, uncoupling protein-1). Finally, we tested whether third ventricularly injected MTII, a highly selective MC4-R agonist (cyclo[beta-Ala-His-D-Phe-Arg-Trp-Glu]NH2) increased or agouti-related protein decreased IBAT temperature in hamsters implanted with sc IBAT temperature transponders. Centrally administered MTII provoked differential sympathetic drives to WAT and IBAT (increased inguinal WAT, dorsosubcutaneous WAT and IBAT NETO, but not epididymal WAT and retroperitoneal WAT NETO). MTII also increased circulating concentrations of the lipolytic products free fatty acids and glycerol but not plasma catecholamines, suggesting lipid mobilization via WAT SNS innervation and not via adrenal medullary catecholamines. WAT or IBAT gene expression was largely unaffected by acute MTII treatment, but IBAT temperature was increased by MTII and the MC4-R agonist and decreased by agouti-related protein. Collectively, this is the first demonstration of central melanocortin agonist stimulation of WAT lipolysis through the SNS and confirms melanocortin-induced changes in BAT thermogenesis.
Collapse
Affiliation(s)
- Márcia N Brito
- Department of Morphophysiological Sciences, State University of Maringá, Maringá, Brazil
| | | | | | | | | |
Collapse
|
42
|
Nogueiras R, Wiedmer P, Perez-Tilve D, Veyrat-Durebex C, Keogh JM, Sutton GM, Pfluger PT, Castaneda TR, Neschen S, Hofmann SM, Howles PN, Morgan DA, Benoit SC, Szanto I, Schrott B, Schürmann A, Joost HG, Hammond C, Hui DY, Woods SC, Rahmouni K, Butler AA, Farooqi IS, O’Rahilly S, Rohner-Jeanrenaud F, Tschöp MH. The central melanocortin system directly controls peripheral lipid metabolism. J Clin Invest 2007; 117:3475-88. [PMID: 17885689 PMCID: PMC1978426 DOI: 10.1172/jci31743] [Citation(s) in RCA: 316] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Accepted: 07/30/2007] [Indexed: 12/21/2022] Open
Abstract
Disruptions of the melanocortin signaling system have been linked to obesity. We investigated a possible role of the central nervous melanocortin system (CNS-Mcr) in the control of adiposity through effects on nutrient partitioning and cellular lipid metabolism independent of nutrient intake. We report that pharmacological inhibition of melanocortin receptors (Mcr) in rats and genetic disruption of Mc4r in mice directly and potently promoted lipid uptake, triglyceride synthesis, and fat accumulation in white adipose tissue (WAT), while increased CNS-Mcr signaling triggered lipid mobilization. These effects were independent of food intake and preceded changes in adiposity. In addition, decreased CNS-Mcr signaling promoted increased insulin sensitivity and glucose uptake in WAT while decreasing glucose utilization in muscle and brown adipose tissue. Such CNS control of peripheral nutrient partitioning depended on sympathetic nervous system function and was enhanced by synergistic effects on liver triglyceride synthesis. Our findings offer an explanation for enhanced adiposity resulting from decreased melanocortin signaling, even in the absence of hyperphagia, and are consistent with feeding-independent changes in substrate utilization as reflected by respiratory quotient, which is increased with chronic Mcr blockade in rodents and in humans with loss-of-function mutations in MC4R. We also reveal molecular underpinnings for direct control of the CNS-Mcr over lipid metabolism. These results suggest ways to design more efficient pharmacological methods for controlling adiposity.
Collapse
Affiliation(s)
- Ruben Nogueiras
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Petra Wiedmer
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Diego Perez-Tilve
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Christelle Veyrat-Durebex
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Julia M. Keogh
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Gregory M. Sutton
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Paul T. Pfluger
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Tamara R. Castaneda
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Susanne Neschen
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Susanna M. Hofmann
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Philip N. Howles
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Donald A. Morgan
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Stephen C. Benoit
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Ildiko Szanto
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Brigitte Schrott
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Annette Schürmann
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Hans-Georg Joost
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Craig Hammond
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - David Y. Hui
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Stephen C. Woods
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Kamal Rahmouni
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Andrew A. Butler
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - I. Sadaf Farooqi
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Stephen O’Rahilly
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Françoise Rohner-Jeanrenaud
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| | - Matthias H. Tschöp
- Obesity Research Center, Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
University Departments of Medicine and Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
Center of Arteriosclerosis Studies, Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA.
Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
Eli Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, USA
| |
Collapse
|
43
|
Strader AD, Shi H, Ogawa R, Seeley RJ, Reizes O. The effects of the melanocortin agonist (MT-II) on subcutaneous and visceral adipose tissue in rodents. J Pharmacol Exp Ther 2007; 322:1153-61. [PMID: 17567964 DOI: 10.1124/jpet.107.123091] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The melanocortin system is a critical pathway in the regulation of energy balance. In this study, we analyzed the peripheral effects of the synthetic melanocortin agonist melanotan-II (MT-II) in rodents fed a low-fat or high-fat diet. MT-II-treated high-fat diet-induced obese (DIO) mice lost weight and body fat, whereas MT-II-treated low-fat-fed mice maintained their original body weight. Specifically, MT-II treatment led to a general reduction in both visceral and subcutaneous adipose tissue in high-fat-fed mice compared with Vehicle (ad libitum) controls. Vehicle-treated pair-fed DIO mice lost an equivalent amount of body weight compared with MT-II-treated mice but retained more adipose tissue. Pair-fed mice showed a reduction in visceral adipose tissue and no effect on subcutaneous adipose tissue compared with MT-II-treated mice. It is surprising that subcutaneous lean mass was significantly reduced in the pairfed mice. The data were replicated in DIO rats and indicated that MT-II treatment led to a generalized reduction in adipose tissue. These results indicate that peripheral MT-II treatment leads to weight loss that affects both the visceral and subcutaneous fat compartments. This finding illustrates the complexity of analyzing weight-reducing compounds. Although the present data suggest that the anorectic effect of MT-II is primarily a consequence of reduced food intake, the body composition data suggest that other mechanisms are involved.
Collapse
Affiliation(s)
- April D Strader
- University of Cincinnati School of Medicine, Cincinnati, Ohio, USA.
| | | | | | | | | |
Collapse
|
44
|
|
45
|
Kuo LE, Kitlinska JB, Tilan JU, Li L, Baker SB, Johnson MD, Lee EW, Burnett MS, Fricke ST, Kvetnansky R, Herzog H, Zukowska Z. Neuropeptide Y acts directly in the periphery on fat tissue and mediates stress-induced obesity and metabolic syndrome. Nat Med 2007; 13:803-11. [PMID: 17603492 DOI: 10.1038/nm1611] [Citation(s) in RCA: 459] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Accepted: 05/25/2007] [Indexed: 12/29/2022]
Abstract
The relationship between stress and obesity remains elusive. In response to stress, some people lose weight, whereas others gain. Here we report that stress exaggerates diet-induced obesity through a peripheral mechanism in the abdominal white adipose tissue that is mediated by neuropeptide Y (NPY). Stressors such as exposure to cold or aggression lead to the release of NPY from sympathetic nerves, which in turn upregulates NPY and its Y2 receptors (NPY2R) in a glucocorticoid-dependent manner in the abdominal fat. This positive feedback response by NPY leads to the growth of abdominal fat. Release of NPY and activation of NPY2R stimulates fat angiogenesis, macrophage infiltration, and the proliferation and differentiation of new adipocytes, resulting in abdominal obesity and a metabolic syndrome-like condition. NPY, like stress, stimulates mouse and human fat growth, whereas pharmacological inhibition or fat-targeted knockdown of NPY2R is anti-angiogenic and anti-adipogenic, while reducing abdominal obesity and metabolic abnormalities. Thus, manipulations of NPY2R activity within fat tissue offer new ways to remodel fat and treat obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Lydia E Kuo
- Department of Physiology & Biophysics, Georgetown University Medical Center, 3900 Reservoir Rd. NW, BSB 234, Washington, DC 20057, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Lee M, Kim A, Chua SC, Obici S, Wardlaw SL. Transgenic MSH overexpression attenuates the metabolic effects of a high-fat diet. Am J Physiol Endocrinol Metab 2007; 293:E121-31. [PMID: 17374695 DOI: 10.1152/ajpendo.00555.2006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To determine whether long-term melanocortinergic activation can attenuate the metabolic effects of a high fat diet, mice overexpressing an NH(2)-terminal POMC transgene that includes alpha- and gamma(3)-MSH were studied on either a 10% low-fat diet (LFD) or 45% high-fat diet (HFD). Weight gain was modestly reduced in transgenic (Tg-MSH) male and female mice vs. wild type (WT) on HFD (P < 0.05) but not LFD. Substantial reductions in body fat percentage were found in both male and female Tg-MSH mice on LFD (P < 0.05) and were more pronounced on HFD (P < 0.001). These changes occurred in the absence of significant feeding differences in most groups, consistent with effects of Tg-MSH on energy expenditure and partitioning. This is supported by indirect calorimetry studies demonstrating higher resting oxygen consumption and lower RQ in Tg-MSH mice on the HFD. Tg-MSH mice had lower fasting insulin levels and improved glucose tolerance on both diets. Histological and biochemical analyses revealed that hepatic fat accumulation was markedly reduced in Tg-MSH mice on the HFD. Tg-MSH also attenuated the increase in corticosterone induced by the HFD. Higher levels of Agrp mRNA, which might counteract effects of the transgene, were measured in Tg-MSH mice on LFD (P = 0.02) but not HFD. These data show that long-term melanocortin activation reduces body weight, adiposity, and hepatic fat accumulation and improves glucose metabolism, particularly in the setting of diet-induced obesity. Our results suggest that long-term melanocortinergic activation could serve as a potential strategy for the treatment of obesity and its deleterious metabolic consequences.
Collapse
Affiliation(s)
- Michelle Lee
- Department of Medicine, Columbia University College of Physicians and Surgeons, 630 West 168th St., New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
47
|
Coll AP, Fassnacht M, Klammer S, Hahner S, Schulte DM, Piper S, Tung YCL, Challis BG, Weinstein Y, Allolio B, O'Rahilly S, Beuschlein F. Peripheral administration of the N-terminal pro-opiomelanocortin fragment 1-28 to Pomc-/- mice reduces food intake and weight but does not affect adrenal growth or corticosterone production. J Endocrinol 2006; 190:515-25. [PMID: 16899584 PMCID: PMC2638022 DOI: 10.1677/joe.1.06749] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Pro-opiomelanocortin (POMC) is a polypeptide precursor that undergoes extensive processing to yield a range of peptides with biologically diverse functions. POMC-derived ACTH is vital for normal adrenal function and the melanocortin alpha-MSH plays a key role in appetite control and energy homeostasis. However, the roles of peptide fragments derived from the highly conserved N-terminal region of POMC are less well characterized. We have used mice with a null mutation in the Pomc gene (Pomc(-/-)) to determine the in vivo effects of synthetic N-terminal 1-28 POMC, which has been shown previously to possess adrenal mitogenic activity. 1-28 POMC (20 mug) given s.c. for 10 days had no effect on the adrenal cortex of Pomc(-/-) mice, with resultant cortical morphology and plasma corticosterone levels being indistinguishable from sham treatment. Concurrent administration of 1-28 POMC and 1-24 ACTH (30 mug/day) resulted in changes identical to 1-24 ACTH treatment alone, which consisted of upregulation of steroidogenic enzymes, elevation of corticosterone levels, hypertrophy of the zona fasciculate, and regression of the X-zone. However, treatment of corticosterone-depleted Pomc(-/-) mice with 1-28 POMC reduced cumulative food intake and total body weight. These anorexigenic effects were ameliorated when the peptide was administered to Pomc(-/-) mice with circulating corticosterone restored either to a low physiological level by corticosterone-supplemented drinking water (CORT) or to a supraphysiological level by concurrent 1-24 ACTH administration. Further, i.c.v. administration of 1-28 POMC to CORT-treated Pomc(-/-) mice had no effect on food intake or body weight. In wild-type mice, the effects of 1-28 POMC upon food intake and body weight were identical to sham treatment, but 1-28 POMC was able to ameliorate the hyperphagia induced by concurrent 1-24 ACTH treatment. In a mouse model which lacks all endogenous POMC peptides, s.c. treatment with synthetic 1-28 POMC alone can reduce food intake and body weight, but has no impact upon adrenal growth or steroidogenesis.
Collapse
Affiliation(s)
- Anthony P. Coll
- Departments of Clinical Biochemistry and Medicine, Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Martin Fassnacht
- Division of Endocrinology, Department of Internal Medicine I, University Hospital Würzburg, Germany
| | - Steffen Klammer
- Division of Endocrinology and Diabetes, Department of Internal Medicine II, University Hospital Freiburg, Germany
| | - Stephanie Hahner
- Division of Endocrinology, Department of Internal Medicine I, University Hospital Würzburg, Germany
| | - Dominik M. Schulte
- Division of Endocrinology and Diabetes, Department of Internal Medicine II, University Hospital Freiburg, Germany
| | - Sarah Piper
- Departments of Clinical Biochemistry and Medicine, Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - YC Loraine Tung
- Departments of Clinical Biochemistry and Medicine, Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Benjamin G. Challis
- Departments of Clinical Biochemistry and Medicine, Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Yacob Weinstein
- Faculty of Health Sciences, Department of Microbiology and Immunology, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Bruno Allolio
- Division of Endocrinology, Department of Internal Medicine I, University Hospital Würzburg, Germany
| | - Stephen O'Rahilly
- Departments of Clinical Biochemistry and Medicine, Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Felix Beuschlein
- Division of Endocrinology and Diabetes, Department of Internal Medicine II, University Hospital Freiburg, Germany
| |
Collapse
|
48
|
Schäffler A, Schölmerich J, Buechler C. The role of 'adipotropins' and the clinical importance of a potential hypothalamic–pituitary–adipose axis. ACTA ACUST UNITED AC 2006; 2:374-83. [PMID: 16932320 DOI: 10.1038/ncpendmet0197] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2005] [Accepted: 02/09/2006] [Indexed: 01/17/2023]
Abstract
Since adipocytes express specific receptors for pituitary hormones and hypothalamic releasing factors, adipose tissue has to be regarded as a fast-acting endocrine gland under the control of the brain. Expanding on this suggestion, the existence and clinical impact of a hypothalamic-pituitary-adipose axis is reviewed. The term 'adipotropins' is introduced in order to describe pituitary and hypothalamic hormones or releasing factors that directly target adipocytes by their specific receptors.
Collapse
Affiliation(s)
- Andreas Schäffler
- Department of Internal Medicine I, University of Regensburg, Germany.
| | | | | |
Collapse
|
49
|
Hervieu GJ. Further insights into the neurobiology of melanin-concentrating hormone in energy and mood balances. Expert Opin Ther Targets 2006; 10:211-29. [PMID: 16548771 DOI: 10.1517/14728222.10.2.211] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Melanin-concentrating hormone (MCH) is a critical hypothalamic anabolic neuropeptide, with key central and peripheral actions on energy balance regulation. The actions of MCH are, so far, known to be transduced through two seven-transmembrane-like receptor paralogues, named MCH1R and MCH2R. MCH2R is not functional in rodents. MCH1R is an important receptor involved in mediating feeding behaviour modulation by MCH in rodents. Pharmacological antagonism at MCH1R in rodents diminishes food intake and results in significant and sustained weight loss in fat tissues, particularly in obese animals. Additionally, MCH1R antagonists have been shown to have anxiolytic and antidepressant properties. The purpose of this review is to highlight the recent numerous pieces of evidence showing that pharmacological blockade at MCH1R could be a potential treatment for obesity and its related metabolic syndrome, as well as for various psychiatric disorders.
Collapse
Affiliation(s)
- Guillaume J Hervieu
- GlaxoSmithKline R&D, Neurology Centre of Excellence for Drug Discovery, NFSP-North, HW1713 Building H17, L1-130 C06 Third Avenue, Harlow, Essex CM19 5AW, UK.
| |
Collapse
|