1
|
Furman KL, Baron L, Lyons HC, Cha T, Evans JR, Manna J, Zhu L, Mattis J, Burgess CR. Melanin-Concentrating Hormone Projections to the Nucleus Accumbens Enhance the Reward Value of Food Consumption and Do Not Induce Feeding or REM Sleep. J Neurosci 2025; 45:e1725242024. [PMID: 39746823 PMCID: PMC11884387 DOI: 10.1523/jneurosci.1725-24.2024] [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: 08/05/2024] [Revised: 11/05/2024] [Accepted: 11/27/2024] [Indexed: 01/04/2025] Open
Abstract
Regulation of food intake and energy balance is critical to survival. Hunger develops as a response to energy deficit and drives food-seeking and consumption. However, motivations to eat are varied in nature and promoted by factors other than energy deficit. When dysregulated, nonhomeostatic drives to consume can contribute to disorders of food intake, adding to the increasing prevalence of restrictive eating disorders and obesity. Melanin-concentrating hormone (MCH) neurons have been implicated in the regulation of feeding behavior, in addition to a number of other fundamental behaviors including sleep, anxiety, and maternal behavior. Several studies suggest that MCH peptide increases food consumption, while studies of MCH neurons show effects only on cued feeding, and others show no effect of MCH neuron manipulation on feeding. MCH neurons have widespread projections to diverse downstream brain regions, yet few studies have investigated the function of specific projections or differentiated the behaviors they regulate. Here we use optogenetics, in combination with different behavioral paradigms, to elucidate the role of MCH projections to the nucleus accumbens (NAc) in sleep and feeding behavior. We show that MCH neurons projecting to the NAc do not induce changes in baseline feeding or REM sleep but do enhance the preference for a food paired with optogenetic stimulation. Furthermore, this effect is diminished in female mice relative to males, in line with previous results suggesting sex differences in the functional role of MCH neurons. These results suggest that MCH projections to the NAc can enhance the rewarding value of consumed food.
Collapse
Affiliation(s)
- Katherine L Furman
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan 48109
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan 48109
| | - Lorelei Baron
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan 48109
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Hannah C Lyons
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan 48109
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109
| | - Timothy Cha
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan 48109
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Jack R Evans
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan 48109
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Jayeeta Manna
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan 48109
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Limei Zhu
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109
| | - Joanna Mattis
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan 48109
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan 48109
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109
| | - Christian R Burgess
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan 48109
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan 48109
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
2
|
Liu X, Yang H, Xu W, Wang X, Tang W, Wang X, Jiao Y, Luan X, Li P, Guo F. Melanin-concentrating hormone attenuates the hedonic feeding induced by orexin-A in the ventral tegmental area of high-fat diet male mice. Front Nutr 2024; 11:1468874. [PMID: 39758319 PMCID: PMC11697430 DOI: 10.3389/fnut.2024.1468874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Objective The ventral tegmental area (VTA), a pivotal hub in the brain's reward circuitry, receives inputs from the lateral hypothalamic area (LHA). However, it remains unclear whether melanin-concentrating hormone (MCH) and orexin-A (OX-A) neurons in the LHA exert individual or cooperative influence on palatable food consumption in the VTA. This study aims to investigate the modulatory role of MCH and OX-A in hedonic feeding within the VTA of high-fat diet (HFD) mice. Methods Male mice were subjected to an 8-week high-fat diet. To visualize the projections from the LHA to VTA, we employed fluorescent gold retrograde tracing combined with immunofluorescence staining. Immunofluorescence staining or enzyme-linked immunosorbent assay was used to detect the activity of the VTA neurons, expression of OX-A or MCH in the LHA, as well as the activity of their receptors (OXR1 and MCHR1) in the VTA following a sucrose preference test. Single-unit extracellular electrical discharge recordings were conducted to assess the effects of OX-A and MCH on VTA neurons in HFD mice. Additionally, chemogenetic inhibition of MCH neurons and immunofluorescence staining were utilized to observe the regulatory roles of MCH in changes of hedonic feeding induced by OX-A in HFD mice. Results Sucrose intake resulted in lower activation of VTA neurons in the HFD mice compared to CON mice, while OX-Aergic and MCHergic neurons project from the LHA to the VTA. Although sucrose intake increased the expression of OX-A and MCH in HFD mice, it led to diminished activation of OXR1-positive and MCHR1-positive VTA neurons compared to CON mice. Extracellular single-unit recording revealed that MCH significantly suppressed the firing rate of OX-A-responsive neurons in the VTA. MCH attenuated the hedonic feeding response induced by OX-A in HFD mice, and administration of MCHR1 antagonist (SNAP94847) significantly potentiated the effect of OX-A. Chemogenetic inhibition of MCH neurons improved the activity of OXR1-expressing neurons, which could be reversed by pretreatment with an OXR1 antagonist (SB334867). Furthermore, chemogenetic inhibition of MCH enhanced hedonic feeding behavior, which was counteracted by SB334867 treatment in HFD mice. Conclusion Melanin-concentrating hormone could attenuate the hedonic feeding behavior induced by orexin-A in the VTA of HFD mice.
Collapse
Affiliation(s)
- Xiaoning Liu
- Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Pathology, Women and Children’s Hospital, Qingdao University, Qingdao, Shandong, China
| | - Helin Yang
- Department of Spine Surgery, Peking University People’s Hospital, Women and Children’s Hospital, Qingdao University, Qingdao, Shandong, China
| | - Wenguang Xu
- Department of Gastroenterology, Affiliated Qingdao Third People’s Hospital, Qingdao University, Qingdao, Shandong, China
| | - Xuezhe Wang
- Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Wenhui Tang
- Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xiaoxuan Wang
- Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Yang Jiao
- Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xinchi Luan
- Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Pengmeng Li
- Department of Gastroenterology, Affiliated Qingdao Third People’s Hospital, Qingdao University, Qingdao, Shandong, China
| | - Feifei Guo
- Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
3
|
Furman KL, Baron L, Lyons HC, Cha T, Evans JR, Manna J, Zhu L, Mattis J, Burgess CR. Melanin concentrating hormone projections to the nucleus accumbens enhance the reward value of food consumption and do not induce feeding or REM sleep. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.622987. [PMID: 39605522 PMCID: PMC11601410 DOI: 10.1101/2024.11.11.622987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Regulation of food intake and energy balance is critical to survival. Hunger develops as a response to energy deficit and drives food-seeking and consumption. However, motivations to eat are varied in nature, and promoted by factors other than energy deficit. When dysregulated, non-homeostatic drives to consume can contribute to disorders of food intake, adding to the increasing prevalence of restrictive eating disorders and obesity. Melanin-concentrating hormone (MCH) neurons have been implicated in the regulation of feeding behavior, in addition to a number of other fundamental behaviors including sleep, anxiety, and maternal behavior. Several studies suggest that MCH peptide increases food consumption, while studies of MCH neurons show effects only on cued feeding, and others show no effect of MCH neuron manipulation on feeding. MCH neurons have widespread projections to diverse downstream brain regions yet few studies have investigated the function of specific projections or differentiated the behaviors they regulate. Here we use optogenetics, in combination with different behavioral paradigms, to elucidate the role of MCH projections to the nucleus accumbens (NAc) in sleep and feeding behavior. We show that MCH neurons projecting to the NAc do not induce changes in baseline feeding or REM sleep, but do enhance the preference for a food paired with optogenetic stimulation. Furthermore, this effect is diminished in female mice relative to males, in line with previous results suggesting sex differences in the functional role of MCH neurons. These results suggest that MCH projections to the NAc can enhance the rewarding value of consumed food.
Collapse
Affiliation(s)
- Katherine L. Furman
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI USA
| | - Lorelei Baron
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI USA
| | - Hannah C. Lyons
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI USA
- Department of Neurology, University of Michigan, Ann Arbor, MI USA
| | - Timothy Cha
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI USA
| | - Jack R. Evans
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI USA
| | - Jayeeta Manna
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI USA
| | - Limei Zhu
- Department of Neurology, University of Michigan, Ann Arbor, MI USA
| | - Joanna Mattis
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI USA
- Department of Neurology, University of Michigan, Ann Arbor, MI USA
| | - Christian R. Burgess
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI USA
| |
Collapse
|
4
|
Pham XT, Abe Y, Mukai Y, Ono D, Tanaka KF, Ohmura Y, Wake H, Yamanaka A. Glutamatergic signaling from melanin-concentrating hormone-producing neurons: A requirement for memory regulation, but not for metabolism control. PNAS NEXUS 2024; 3:pgae275. [PMID: 39035036 PMCID: PMC11259978 DOI: 10.1093/pnasnexus/pgae275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 06/29/2024] [Indexed: 07/23/2024]
Abstract
Melanin-concentrating hormone-producing neurons (MCH neurons), found mainly in the lateral hypothalamus and surrounding areas, play essential roles in various brain functions, including sleep and wakefulness, reward, metabolism, learning, and memory. These neurons coexpress several neurotransmitters and act as glutamatergic neurons. The contribution of glutamate from MCH neurons to memory- and metabolism-related functions has not been fully investigated. In a mouse model, we conditionally knocked out Slc17a6 gene, which encodes for vesicular glutamate transporter 2 (vGlut2), in the MCH neurons exclusively by using two different methods: the Cre recombinase/loxP system and in vivo genome editing using CRISPR/Cas9. Then, we evaluated several aspects of memory and measured metabolic rates using indirect calorimetry. We found that mice with MCH neuron-exclusive vGlut2 ablation had higher discrimination ratios between novel and familiar stimuli for novel object recognition, object location, and three-chamber tests. In contrast, there was no significant change in body weight, food intake, oxygen consumption, respiratory quotient, or locomotor activity. These findings suggest that glutamatergic signaling from MCH neurons is required to regulate memory, but its role in regulating metabolic rate is negligible.
Collapse
Affiliation(s)
- Xuan Thang Pham
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Department of Psychiatry, Hanoi Medical University, Hanoi 100000, Vietnam
| | - Yoshifumi Abe
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yasutaka Mukai
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Japan Society for the Promotion of Science, Tokyo 102-0083, Japan
| | - Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yu Ohmura
- Chinese Institute for Brain Research, Beijing (CIBR), Beijing 102206, China
| | - Hiroaki Wake
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Akihiro Yamanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan
- Chinese Institute for Brain Research, Beijing (CIBR), Beijing 102206, China
- National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi 444-8585, Japan
| |
Collapse
|
5
|
Concetti C, Peleg-Raibstein D, Burdakov D. Hypothalamic MCH Neurons: From Feeding to Cognitive Control. FUNCTION 2023; 5:zqad059. [PMID: 38020069 PMCID: PMC10667013 DOI: 10.1093/function/zqad059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Modern neuroscience is progressively elucidating that the classic view positing distinct brain regions responsible for survival, emotion, and cognitive functions is outdated. The hypothalamus demonstrates the interdependence of these roles, as it is traditionally known for fundamental survival functions like energy and electrolyte balance, but is now recognized to also play a crucial role in emotional and cognitive processes. This review focuses on lateral hypothalamic melanin-concentrating hormone (MCH) neurons, producing the neuropeptide MCH-a relatively understudied neuronal population with integrative functions related to homeostatic regulation and motivated behaviors, with widespread inputs and outputs throughout the entire central nervous system. Here, we review early findings and recent literature outlining their role in the regulation of energy balance, sleep, learning, and memory processes.
Collapse
Affiliation(s)
- Cristina Concetti
- Neurobehavioural Dynamics Laboratory, ETH Zürich, Schorenstrasse 16, Schwerzenbach 8603, Switzerland
| | - Daria Peleg-Raibstein
- Neurobehavioural Dynamics Laboratory, ETH Zürich, Schorenstrasse 16, Schwerzenbach 8603, Switzerland
| | - Denis Burdakov
- Neurobehavioural Dynamics Laboratory, ETH Zürich, Schorenstrasse 16, Schwerzenbach 8603, Switzerland
| |
Collapse
|
6
|
Potter LE, Burgess CR. The melanin-concentrating hormone system as a target for the treatment of sleep disorders. Front Neurosci 2022; 16:952275. [PMID: 36177357 PMCID: PMC9513178 DOI: 10.3389/fnins.2022.952275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Given the widespread prevalence of sleep disorders and their impacts on health, it is critical that researchers continue to identify and evaluate novel avenues of treatment. Recently the melanin-concentrating hormone (MCH) system has attracted commercial and scientific interest as a potential target of pharmacotherapy for sleep disorders. This interest emerges from basic scientific research demonstrating a role for MCH in regulating sleep, and particularly REM sleep. In addition to this role in sleep regulation, the MCH system and the MCH receptor 1 (MCHR1) have been implicated in a wide variety of other physiological functions and behaviors, including feeding/metabolism, reward, anxiety, depression, and learning. The basic research literature on sleep and the MCH system, and the history of MCH drug development, provide cause for both skepticism and cautious optimism about the prospects of MCH-targeting drugs in sleep disorders. Extensive efforts have focused on developing MCHR1 antagonists for use in obesity, however, few of these drugs have advanced to clinical trials, and none have gained regulatory approval. Additional basic research will be needed to fully characterize the MCH system’s role in sleep regulation, for example, to fully differentiate between MCH-neuron and peptide/receptor-mediated functions. Additionally, a number of issues relating to drug design will continue to pose a practical challenge for novel pharmacotherapies targeting the MCH system.
Collapse
Affiliation(s)
- Liam E. Potter
- Department of Molecular and Integrative Physiology, Michigan Medicine, Ann Arbor, MI, United States
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Liam E. Potter,
| | - Christian R. Burgess
- Department of Molecular and Integrative Physiology, Michigan Medicine, Ann Arbor, MI, United States
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
- Christian R. Burgess,
| |
Collapse
|
7
|
Al-Massadi O, Dieguez C, Schneeberger M, López M, Schwaninger M, Prevot V, Nogueiras R. Multifaceted actions of melanin-concentrating hormone on mammalian energy homeostasis. Nat Rev Endocrinol 2021; 17:745-755. [PMID: 34608277 DOI: 10.1038/s41574-021-00559-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2021] [Indexed: 12/12/2022]
Abstract
Melanin-concentrating hormone (MCH) is a small cyclic peptide expressed in all mammals, mainly in the hypothalamus. MCH acts as a robust integrator of several physiological functions and has crucial roles in the regulation of sleep-wake rhythms, feeding behaviour and metabolism. MCH signalling has a very broad endocrine context and is involved in physiological functions and emotional states associated with metabolism, such as reproduction, anxiety, depression, sleep and circadian rhythms. MCH mediates its functions through two receptors (MCHR1 and MCHR2), of which only MCHR1 is common to all mammals. Owing to the wide variety of MCH downstream signalling pathways, MCHR1 agonists and antagonists have great potential as tools for the directed management of energy balance disorders and associated metabolic complications, and translational strategies using these compounds hold promise for the development of novel treatments for obesity. This Review provides an overview of the numerous roles of MCH in energy and glucose homeostasis, as well as in regulation of the mesolimbic dopaminergic circuits that encode the hedonic component of food intake.
Collapse
Affiliation(s)
- Omar Al-Massadi
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Spain.
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain.
| | - Carlos Dieguez
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Marc Schneeberger
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Miguel López
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience and Cognition, Laboratory of Development and Plasticity of the Neuroendocrine Brain, UMR-S1172, EGID, Lille, France
| | - Ruben Nogueiras
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain.
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain.
- Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain.
| |
Collapse
|
8
|
Ghanemi A, Yoshioka M, St-Amand J. Trefoil Factor Family Member 2: From a High-Fat-Induced Gene to a Potential Obesity Therapy Target. Metabolites 2021; 11:metabo11080536. [PMID: 34436477 PMCID: PMC8401738 DOI: 10.3390/metabo11080536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/11/2022] Open
Abstract
Obesity has its epidemiological patterns continuously increasing. With controlling both diet and exercise being the main approaches to manage the energy metabolism balance, a high-fat (HF) diet is of particular importance. Indeed, lipids have a low satiety potential but a high caloric density. Thus, focusing on pharmacologically targetable pathways remains an approach with promising therapeutic potential. Within this context, trefoil factor family member 2 (Tff2) has been characterized as specifically induced by HF diet rather than low-fat diet. TFF2 has also been linked to diverse neurological mechanisms and metabolic patterns suggesting its role in energy balance. The hypothesis is that TFF2 would be a HF diet-induced signal that regulates metabolism with a focus on lipids. Within this review, we put the spotlight on key findings highlighting this line of thought. Importantly, the hypothetical mechanisms pointed highlight TFF2 as an important contributor to obesity development via increasing lipids intestinal absorption and anabolism. Therefore, an outlook for future experimental activities and evaluation of the therapeutic potential of TFF2 inhibition is given. Indeed, its knockdown or downregulation would contribute to an antiobesity phenotype. We believe this work represents an addition to our understanding of the lipidic molecular implications in obesity, which will contribute to develop therapies aiming to manage the lipidic metabolic pathways including the absorption, storage and metabolism via targeting TFF2-related pathways. We briefly discuss important relevant concepts for both basic and clinical researchers.
Collapse
Affiliation(s)
- Abdelaziz Ghanemi
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Quebec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
| | - Mayumi Yoshioka
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Quebec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
| | - Jonny St-Amand
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Quebec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
- Correspondence:
| |
Collapse
|
9
|
Izawa S, Yoneshiro T, Kondoh K, Nakagiri S, Okamatsu-Ogura Y, Terao A, Minokoshi Y, Yamanaka A, Kimura K. Melanin-concentrating hormone-producing neurons in the hypothalamus regulate brown adipose tissue and thus contribute to energy expenditure. J Physiol 2021; 600:815-827. [PMID: 33899241 DOI: 10.1113/jp281241] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/20/2021] [Indexed: 12/17/2022] Open
Abstract
KEY POINTS Melanin-concentrating hormone (MCH) neuron-ablated mice exhibit increased energy expenditure and reduced fat weight. Increased brown adipose tissue (BAT) activity and locomotor activity-independent energy expenditure contributed to body weight reduction in MCH neuron-ablated mice. MCH neurons send inhibitory input to the medullary raphe nucleus to modulate BAT activity. ABSTRACT Hypothalamic melanin-concentrating hormone (MCH) peptide robustly affects energy homeostasis. However, it is unclear whether and how MCH-producing neurons, which contain and release a variety of neuropeptides/transmitters, regulate energy expenditure in the central nervous system and peripheral tissues. We thus examined the regulation of energy expenditure by MCH neurons, focusing on interscapular brown adipose tissue (BAT) activity. MCH neuron-ablated mice exhibited reduced body weight, increased oxygen consumption, and increased BAT activity, which improved locomotor activity-independent energy expenditure. Trans-neuronal retrograde tracing with the recombinant pseudorabies virus revealed that MCH neurons innervate BAT via the sympathetic premotor region in the medullary raphe nucleus (MRN). MRN neurons were activated by MCH neuron ablation. Therefore, endogenous MCH neuron activity negatively modulates energy expenditure via BAT inhibition. MRN neurons might receive inhibitory input from MCH neurons to suppress BAT activity.
Collapse
Affiliation(s)
- Shuntaro Izawa
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan.,Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan.,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.,JSPS Research Fellowship for Young Scientists, Tokyo, 102-0083, Japan
| | - Takeshi Yoneshiro
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan.,Division of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, 153-8904, Japan
| | - Kunio Kondoh
- Division of Endocrinology and Metabolism, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, 444-8585, Japan.,Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8585, Japan
| | - Shohei Nakagiri
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| | - Yuko Okamatsu-Ogura
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| | - Akira Terao
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan.,Department of Biology, School of Biological Sciences, Tokai University, Sapporo, 005-8601, Japan
| | - Yasuhiko Minokoshi
- Division of Endocrinology and Metabolism, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, 444-8585, Japan.,Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8585, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan.,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Kazuhiro Kimura
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| |
Collapse
|
10
|
Lord MN, Subramanian K, Kanoski SE, Noble EE. Melanin-concentrating hormone and food intake control: Sites of action, peptide interactions, and appetition. Peptides 2021; 137:170476. [PMID: 33370567 PMCID: PMC8025943 DOI: 10.1016/j.peptides.2020.170476] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022]
Abstract
Given the increased prevalence of obesity and its associated comorbidities, understanding the mechanisms through which the brain regulates energy balance is of critical importance. The neuropeptide melanin-concentrating hormone (MCH) is produced in the lateral hypothalamic area and the adjacent incerto-hypothalamic area and promotes both food intake and energy conservation, overall contributing to body weight gain. Decades of research into this system has provided insight into the neural pathways and mechanisms (behavioral and neurobiological) through which MCH stimulates food intake. Recent technological advancements that allow for selective manipulation of MCH neuron activity have elucidated novel mechanisms of action for the hyperphagic effects of MCH, implicating neural "volume" transmission in the cerebrospinal fluid and sex-specific effects of MCH on food intake control as understudied areas for future investigation. Highlighted here are historical and recent findings that illuminate the neurobiological mechanisms through which MCH promotes food intake, including the identification of various specific neural signaling pathways and interactions with other peptide systems. We conclude with a framework that the hyperphagic effects of MCH signaling are predominantly mediated through enhancement of an "appetition" process in which early postoral prandial signals promote further caloric consumption.
Collapse
Affiliation(s)
- Magen N Lord
- Department of Foods and Nutrition, University of Georgia, Athens, GA 30606, USA
| | - Keshav Subramanian
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
| | - Scott E Kanoski
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA; Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA.
| | - Emily E Noble
- Department of Foods and Nutrition, University of Georgia, Athens, GA 30606, USA.
| |
Collapse
|
11
|
Morganstern I, Gulati G, Leibowitz SF. Role of melanin-concentrating hormone in drug use disorders. Brain Res 2020; 1741:146872. [PMID: 32360868 DOI: 10.1016/j.brainres.2020.146872] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 04/17/2020] [Accepted: 04/28/2020] [Indexed: 12/22/2022]
Abstract
Melanin-concentrating hormone (MCH) is a neuropeptide primarily transcribed in the lateral hypothalamus (LH), with vast projections to many areas throughout the central nervous system that play an important role in motivated behaviors and drug use. Anatomical, pharmacological and genetic studies implicate MCH in mediating the intake and reinforcement of commonly abused substances, acting by influencing several systems including the mesolimbic dopaminergic system, glutamatergic as well as GABAergic signaling and being modulated by inflammatory neuroimmune pathways. Further support for the role of MCH in controlling behavior related to drug use will be discussed as it relates to cerebral ventricular volume transmission and intracellular molecules including cocaine- and amphetamine-regulated transcript peptide, dopamine- and cAMP-regulated phosphoprotein 32 kDa. The primary goal of this review is to introduce and summarize current literature surrounding the role of MCH in mediating the intake and reinforcement of commonly abused drugs, such as alcohol, cocaine, amphetamine, nicotine and opiates.
Collapse
Affiliation(s)
| | - Gazal Gulati
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA
| | - Sarah F Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
12
|
Chee MJ, Hebert AJ, Briançon N, Flaherty SE, Pissios P, Maratos-Flier E. Conditional deletion of melanin-concentrating hormone receptor 1 from GABAergic neurons increases locomotor activity. Mol Metab 2019; 29:114-123. [PMID: 31668382 PMCID: PMC6745487 DOI: 10.1016/j.molmet.2019.08.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/23/2019] [Accepted: 08/25/2019] [Indexed: 12/28/2022] Open
Abstract
Objective Melanin-concentrating hormone (MCH) plays a key role in regulating energy balance. MCH acts via its receptor MCHR1, and MCHR1 deletion increases energy expenditure and locomotor activity, which is associated with a hyperdopaminergic state. Since MCHR1 expression is widespread, the neurons supporting the effects of MCH on energy expenditure are not clearly defined. There is a high density of MCHR1 neurons in the striatum, and these neurons are known to be GABAergic. We thus determined if MCH acts via this GABAergic neurocircuit to mediate energy balance. Methods We generated a Mchr1-flox mouse and crossed it with the Vgat-cre mouse to assess if MCHR1 deletion from GABAergic neurons expressing the vesicular GABA transporter (vGAT) in female Vgat-Mchr1-KO mice resulted in lower body weights or increased energy expenditure. Additionally, we determined if MCHR1-expressing neurons within the accumbens form part of the neural circuit underlying MCH-mediated energy balance by delivering an adeno-associated virus expressing Cre recombinase to the accumbens nucleus of Mchr1-flox mice. To evaluate if a dysregulated dopaminergic tone leads to their hyperactivity, we determined if the dopamine reuptake blocker GBR12909 prolonged the drug-induced locomotor activity in Vgat-Mchr1-KO mice. Furthermore, we also performed amperometry recordings to test whether MCHR1 deletion increases dopamine output within the accumbens and whether MCH can suppress dopamine release. Results Vgat-Mchr1-KO mice have lower body weight, increased energy expenditure, and increased locomotor activity. Similarly, restricting MCHR1 deletion to the accumbens nucleus also increased locomotor activity. Vgat-Mchr1-KO mice show increased and prolonged sensitivity to GBR12909-induced locomotor activity, and amperometry recordings revealed that GBR12909 elevated accumbens dopamine levels to twice that of controls, thus MCHR1 deletion may lead to a hyperdopaminergic state that mediates their observed hyperactivity. Consistent with the inhibitory effect of MCH, we found that MCH acutely suppresses dopamine release within the accumbens. Conclusions As with established models of systemic MCH or MCHR1 deletion, we found that MCHR1 deletion from GABAergic neurons, specifically those within the accumbens nucleus, also led to increased locomotor activity. A hyperdopaminergic state underlies this increased locomotor activity, and is consistent with our finding that MCH signaling within the accumbens nucleus suppresses dopamine release. In effect, MCHR1 deletion may disinhibit dopamine release leading to the observed hyperactivity. Generation of Mchr1-flox mouse enabled cre-mediated deletion of Mchr1. Mchr1 deletion at GABAergic neurons decreased body weight. Mchr1 deletion at GABAergic neurons increased locomotor activity. Mchr1 deletion increased dopaminergic tone in the mesolimbic accumbens circuitry. MCH suppressed dopamine release in the accumbens nucleus.
Collapse
Affiliation(s)
- Melissa J Chee
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada; Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Alex J Hebert
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada; Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nadege Briançon
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Stephen E Flaherty
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Pavlos Pissios
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Eleftheria Maratos-Flier
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Diniz GB, Battagello DS, Cherubini PM, Reyes-Mendoza JD, Luna-Illades C, Klein MO, Motta-Teixeira LC, Sita LV, Miranda-Anaya M, Morales T, Bittencourt JC. Melanin-concentrating hormone peptidergic system: Comparative morphology between muroid species. J Comp Neurol 2019; 527:2973-3001. [PMID: 31152440 DOI: 10.1002/cne.24723] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 12/15/2022]
Abstract
Melanin-concentrating hormone (MCH) is a conserved neuropeptide, predominantly located in the diencephalon of vertebrates, and associated with a wide range of functions. While functional studies have focused on the use of the traditional mouse laboratory model, critical gaps exist in our understanding of the morphology of the MCH system in this species. Even less is known about the nontraditional animal model Neotomodon alstoni (Mexican volcano mouse). A comparative morphological study among these rodents may, therefore, contribute to a better understanding of the evolution of the MCH peptidergic system. To this end, we employed diverse immunohistochemical protocols to identify key aspects of the MCH system, including its spatial relationship to another neurochemical population of the tuberal hypothalamus, the orexins. Three-dimensional (3D) reconstructions were also employed to convey a better sense of spatial distribution to these neurons. Our results show that the distribution of MCH neurons in all rodents studied follows a basic plan, but individual characteristics are found for each species, such as the preeminence of a periventricular group only in the rat, the lack of posterior groups in the mouse, and the extensive presence of MCH neurons in the anterior hypothalamic area of Neotomodon. Taken together, these data suggest a strong anatomical substrate for previously described functions of the MCH system, and that particular neurochemical and morphological features may have been determinant to species-specific phenotypes in rodent evolution.
Collapse
Affiliation(s)
- Giovanne B Diniz
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniella S Battagello
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Santiago de Querétaro, Queretaro, Mexico
| | - Pedro M Cherubini
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Julio D Reyes-Mendoza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Santiago de Querétaro, Queretaro, Mexico
| | - Cesar Luna-Illades
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Santiago de Querétaro, Queretaro, Mexico
| | - Marianne O Klein
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lívia C Motta-Teixeira
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luciane V Sita
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Manuel Miranda-Anaya
- Unidad Multidisciplinaria de Docencia e Investigación, Facultad de Ciencias, Universidad Nacional Autónoma de México, Santiago de Querétaro, Queretaro, Mexico
| | - Teresa Morales
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Santiago de Querétaro, Queretaro, Mexico
| | - Jackson C Bittencourt
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Center for Neuroscience and Behavior, Institute of Psychology, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
14
|
Latifi B, Adamantidis A, Bassetti C, Schmidt MH. Sleep-Wake Cycling and Energy Conservation: Role of Hypocretin and the Lateral Hypothalamus in Dynamic State-Dependent Resource Optimization. Front Neurol 2018; 9:790. [PMID: 30344503 PMCID: PMC6183196 DOI: 10.3389/fneur.2018.00790] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/31/2018] [Indexed: 12/23/2022] Open
Abstract
The hypocretin (Hcrt) system has been implicated in a wide range of physiological functions from sleep-wake regulation to cardiovascular, behavioral, metabolic, and thermoregulagtory control. These wide-ranging physiological effects have challenged the identification of a parsimonious function for Hcrt. A compelling hypothesis suggests that Hcrt plays a role in the integration of sleep-wake neurophysiology with energy metabolism. For example, Hcrt neurons promote waking and feeding, but are also sensors of energy balance. Loss of Hcrt function leads to an increase in REM sleep propensity, but a potential role for Hcrt linking energy balance with REM sleep expression has not been addressed. Here we examine a potential role for Hcrt and the lateral hypothalamus (LH) in state-dependent resource allocation as a means of optimizing resource utilization and, as a result, energy conservation. We review the energy allocation hypothesis of sleep and how state-dependent metabolic partitioning may contribute toward energy conservation, but with additional examination of how the loss of thermoregulatory function during REM sleep may impact resource optimization. Optimization of energy expenditures at the whole organism level necessitates a top-down network responsible for coordinating metabolic operations in a state-dependent manner across organ systems. In this context, we then specifically examine the potential role of the LH in regulating this output control, including the contribution from both Hcrt and melanin concentrating hormone (MCH) neurons among a diverse LH cell population. We propose that this hypothalamic integration system is responsible for global shifts in state-dependent resource allocations, ultimately promoting resource optimization and an energy conservation function of sleep-wake cycling.
Collapse
Affiliation(s)
- Blerina Latifi
- Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Antoine Adamantidis
- Department of Neurology, Center for Experimental Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department of Biomedical Research, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Claudio Bassetti
- Department of Neurology, Center for Experimental Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Markus H Schmidt
- Department of Neurology, Center for Experimental Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Ohio Sleep Medicine Institute, Dublin, OH, United States
| |
Collapse
|
15
|
Diniz GB, Bittencourt JC. The Melanin-Concentrating Hormone as an Integrative Peptide Driving Motivated Behaviors. Front Syst Neurosci 2017; 11:32. [PMID: 28611599 PMCID: PMC5447028 DOI: 10.3389/fnsys.2017.00032] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/04/2017] [Indexed: 12/14/2022] Open
Abstract
The melanin-concentrating hormone (MCH) is an important peptide implicated in the control of motivated behaviors. History, however, made this peptide first known for its participation in the control of skin pigmentation, from which its name derives. In addition to this peripheral role, MCH is strongly implicated in motivated behaviors, such as feeding, drinking, mating and, more recently, maternal behavior. It is suggested that MCH acts as an integrative peptide, converging sensory information and contributing to a general arousal of the organism. In this review, we will discuss the various aspects of energy homeostasis to which MCH has been associated to, focusing on the different inputs that feed the MCH peptidergic system with information regarding the homeostatic status of the organism and the exogenous sensory information that drives this system, as well as the outputs that allow MCH to act over a wide range of homeostatic and behavioral controls, highlighting the available morphological and hodological aspects that underlie these integrative actions. Besides the well-described role of MCH in feeding behavior, a prime example of hypothalamic-mediated integration, we will also examine those functions in which the participation of MCH has not yet been extensively characterized, including sexual, maternal, and defensive behaviors. We also evaluated the available data on the distribution of MCH and its function in the context of animals in their natural environment. Finally, we briefly comment on the evidence for MCH acting as a coordinator between different modalities of motivated behaviors, highlighting the most pressing open questions that are open for investigations and that could provide us with important insights about hypothalamic-dependent homeostatic integration.
Collapse
Affiliation(s)
- Giovanne B. Diniz
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São PauloSão Paulo, Brazil
| | - Jackson C. Bittencourt
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São PauloSão Paulo, Brazil
- Center for Neuroscience and Behavior, Institute of Psychology, University of São PauloSão Paulo, Brazil
| |
Collapse
|
16
|
Hagar JM, Macht VA, Wilson SP, Fadel JR. Upregulation of orexin/hypocretin expression in aged rats: Effects on feeding latency and neurotransmission in the insular cortex. Neuroscience 2017; 350:124-132. [PMID: 28344067 DOI: 10.1016/j.neuroscience.2017.03.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 03/14/2017] [Accepted: 03/15/2017] [Indexed: 10/19/2022]
Abstract
Aging is associated with changes in numerous homeostatic functions, such as food intake, that are thought to be mediated by the hypothalamus. Orexin/hypocretin neurons of the hypothalamus regulate several physiological functions, including feeding, sleep and wakefulness. Evidence from both clinical and animal studies supports the notion that aging is associated with loss or dysregulation of the orexin system. Here, we used virus-mediated gene transfer to manipulate expression of orexin peptides in young and aged rats and examined behavioral and neurochemical correlates of food intake in these animals. Aged rats showed slower feeding latencies when presented with palatable food compared to young control rats, and these deficits were ameliorated by upregulation of orexin expression. Similarly, young animals treated with a virus designed to decrease preproorexin expression showed longer feeding latencies reminiscent of aged control rats. Feeding was also associated with increased acetylcholine, glutamate and GABA efflux in insular cortex of young control animals. Orexin upregulation did not restore deficits in feeding-elicited release of these neurotransmitters in aged rats, but did enhance basal neurotransmitter levels which may have contributed to the behavioral correlates of these genetic manipulations. These studies demonstrate that age-related deficits in behavioral and neurochemical measures of feeding are likely to be mediated, in part, by the orexin system. Because these same neurotransmitter systems have been shown to underlie orexin effects on cognition, treatments which increase orexin function may have potential for improving both physiological and cognitive manifestations of certain age-related disorders.
Collapse
Affiliation(s)
- Janel M Hagar
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - Victoria A Macht
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA; Department of Psychology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - Steven P Wilson
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - James R Fadel
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA.
| |
Collapse
|
17
|
Blasiak A, Gundlach AL, Hess G, Lewandowski MH. Interactions of Circadian Rhythmicity, Stress and Orexigenic Neuropeptide Systems: Implications for Food Intake Control. Front Neurosci 2017; 11:127. [PMID: 28373831 PMCID: PMC5357634 DOI: 10.3389/fnins.2017.00127] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/01/2017] [Indexed: 12/23/2022] Open
Abstract
Many physiological processes fluctuate throughout the day/night and daily fluctuations are observed in brain and peripheral levels of several hormones, neuropeptides and transmitters. In turn, mediators under the “control” of the “master biological clock” reciprocally influence its function. Dysregulation in the rhythmicity of hormone release as well as hormone receptor sensitivity and availability in different tissues, is a common risk-factor for multiple clinical conditions, including psychiatric and metabolic disorders. At the same time circadian rhythms remain in a strong, reciprocal interaction with the hypothalamic-pituitary-adrenal (HPA) axis. Recent findings point to a role of circadian disturbances and excessive stress in the development of obesity and related food consumption and metabolism abnormalities, which constitute a major health problem worldwide. Appetite, food intake and energy balance are under the influence of several brain neuropeptides, including the orexigenic agouti-related peptide, neuropeptide Y, orexin, melanin-concentrating hormone and relaxin-3. Importantly, orexigenic neuropeptide neurons remain under the control of the circadian timing system and are highly sensitive to various stressors, therefore the potential neuronal mechanisms through which disturbances in the daily rhythmicity and stress-related mediator levels contribute to food intake abnormalities rely on reciprocal interactions between these elements.
Collapse
Affiliation(s)
- Anna Blasiak
- Department of Neurophysiology and Chronobiology, Institute of Zoology, Jagiellonian University Krakow, Poland
| | - Andrew L Gundlach
- Neuropeptides Division, The Florey Institute of Neuroscience and Mental HealthParkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, The University of MelbourneParkville, VIC, Australia
| | - Grzegorz Hess
- Department of Neurophysiology and Chronobiology, Institute of Zoology, Jagiellonian UniversityKrakow, Poland; Institute of Pharmacology, Polish Academy of SciencesKrakow, Poland
| | - Marian H Lewandowski
- Department of Neurophysiology and Chronobiology, Institute of Zoology, Jagiellonian University Krakow, Poland
| |
Collapse
|
18
|
Vandal M, White PJ, Tournissac M, Tremblay C, St-Amour I, Drouin-Ouellet J, Bousquet M, Traversy MT, Planel E, Marette A, Calon F. Impaired thermoregulation and beneficial effects of thermoneutrality in the 3×Tg-AD model of Alzheimer's disease. Neurobiol Aging 2016; 43:47-57. [PMID: 27255814 DOI: 10.1016/j.neurobiolaging.2016.03.024] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 10/22/2022]
Abstract
The sharp rise in the incidence of Alzheimer's disease (AD) at an old age coincides with a reduction in energy metabolism and core body temperature. We found that the triple-transgenic mouse model of AD (3×Tg-AD) spontaneously develops a lower basal body temperature and is more vulnerable to a cold environment compared with age-matched controls. This was despite higher nonshivering thermogenic activity, as evidenced by brown adipose tissue norepinephrine content and uncoupling protein 1 expression. A 24-hour exposure to cold (4 °C) aggravated key neuropathologic markers of AD such as: tau phosphorylation, soluble amyloid beta concentrations, and synaptic protein loss in the cortex of 3×Tg-AD mice. Strikingly, raising the body temperature of aged 3×Tg-AD mice via exposure to a thermoneutral environment improved memory function and reduced amyloid and synaptic pathologies within a week. Our results suggest the presence of a vicious cycle between impaired thermoregulation and AD-like neuropathology, and it is proposed that correcting thermoregulatory deficits might be therapeutic in AD.
Collapse
Affiliation(s)
- Milene Vandal
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada; Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, Québec, Canada
| | - Philip J White
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, NC, USA; Faculté de medicine, Université Laval, Québec, Québec, Canada; Institut universitaire de pneumologie et de cardiologie de Québec, Québec, Québec, Canada
| | - Marine Tournissac
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada; Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, Québec, Canada
| | - Cyntia Tremblay
- Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada
| | - Isabelle St-Amour
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada; Département de Recherche et Développement, Héma-Québec, Québec, Québec, Canada
| | - Janelle Drouin-Ouellet
- Faculté de medicine, Université Laval, Québec, Québec, Canada; John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - Melanie Bousquet
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada; Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, Québec, Canada
| | - Marie-Thérèse Traversy
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada
| | - Emmanuel Planel
- Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada; Faculté de medicine, Université Laval, Québec, Québec, Canada
| | - Andre Marette
- Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, Québec, Canada; Faculté de medicine, Université Laval, Québec, Québec, Canada; Institut universitaire de pneumologie et de cardiologie de Québec, Québec, Québec, Canada
| | - Frederic Calon
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada; Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, Québec, Canada.
| |
Collapse
|
19
|
Barson JR, Leibowitz SF. Hypothalamic neuropeptide signaling in alcohol addiction. Prog Neuropsychopharmacol Biol Psychiatry 2016; 65:321-9. [PMID: 25689818 PMCID: PMC4537397 DOI: 10.1016/j.pnpbp.2015.02.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/30/2015] [Accepted: 02/09/2015] [Indexed: 11/27/2022]
Abstract
The hypothalamus is now known to regulate alcohol intake in addition to its established role in food intake, in part through neuromodulatory neurochemicals termed neuropeptides. Certain orexigenic neuropeptides act in the hypothalamus to promote alcohol drinking, although they affect different aspects of the drinking response. These neuropeptides, which include galanin, the endogenous opioid enkephalin, and orexin/hypocretin, appear to stimulate alcohol intake not only through mechanisms that promote food intake but also by enhancing reward and reinforcement from alcohol. Moreover, these neuropeptides participate in a positive feedback relationship with alcohol, whereby they are upregulated by alcohol intake to promote even further consumption. They contrast with other orexigenic neuropeptides, such as melanin-concentrating hormone and neuropeptide Y, which promote alcohol intake under limited circumstances, are not consistently stimulated by alcohol, and do not enhance reward. They also contrast with neuropeptides that can be anorexigenic, including the endogenous opioid dynorphin, corticotropin-releasing factor, and melanocortins, which act in the hypothalamus to inhibit alcohol drinking as well as reward and therefore counter the ingestive drive promoted by orexigenic neuropeptides. Thus, while multiple hypothalamic neuropeptides may work together to regulate different aspects of the alcohol drinking response, excessive signaling from orexigenic neuropeptides or inadequate signaling from anorexigenic neuropeptides can therefore allow alcohol drinking to become dysregulated.
Collapse
Affiliation(s)
- Jessica R. Barson
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, Box 278, New York, NY, 10065 USA
| | - Sarah F. Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, Box 278, New York, NY, 10065 USA
,Corresponding author at: Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, Box 278, New York, NY, 10065 USA. Tel.: +1 212 327 8378; fax: +1 212 327 8447
| |
Collapse
|
20
|
Mediobasal hypothalamic overexpression of DEPTOR protects against high-fat diet-induced obesity. Mol Metab 2015; 5:102-112. [PMID: 26909318 PMCID: PMC4735664 DOI: 10.1016/j.molmet.2015.11.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 11/18/2015] [Accepted: 11/25/2015] [Indexed: 01/29/2023] Open
Abstract
Background/Objective The mechanistic target of rapamycin (mTOR) is a serine–threonine kinase that functions into distinct protein complexes (mTORC1 and mTORC2) that regulate energy homeostasis. DEP-domain containing mTOR-interacting protein (DEPTOR) is part of these complexes and is known to dampen mTORC1 function, consequently reducing mTORC1 negative feedbacks and promoting insulin signaling and Akt/PKB activation in several models. Recently, we observed that DEPTOR is expressed in several structures of the brain including the mediobasal hypothalamus (MBH), a region that regulates energy balance. Whether DEPTOR in the MBH plays a functional role in regulating energy balance and hypothalamic insulin signaling has never been tested. Methods We have generated a novel conditional transgenic mouse model based on the Cre-LoxP system allowing targeted overexpression of DEPTOR. Mice overexpressing DEPTOR in the MBH were subjected to a metabolic phenotyping and MBH insulin signaling was evaluated. Results We first report that systemic (brain and periphery) overexpression of DEPTOR prevents high-fat diet-induced obesity, improves glucose metabolism and protects against hepatic steatosis. These phenotypes were associated with a reduction in food intake and feed efficiency and an elevation in oxygen consumption. Strikingly, specific overexpression of DEPTOR in the MBH completely recapitulated these phenotypes. DEPTOR overexpression was associated with an increase in hypothalamic insulin signaling, as illustrated by elevated Akt/PKB activation. Conclusion Altogether, these results support a role for MBH DEPTOR in the regulation of energy balance and metabolism. Systemic (brain and peripheral) overexpression of DEPTOR promotes activity and improves glucose homeostasis. Systemic (brain and peripheral) overexpression of DEPTOR protects againts high-fat diet-induced obesity and metabolic alterations. Deptor is widely expressed in the mouse brain, with a high expression in the mediobasal hypothalamus (MBH), a key region of the brain that regulates energy balance. MBH-specific DEPTOR overexpression improves glucose metabolism and protects mice against obesity. MBH-specific DEPTOR overexpression promotes hypothalamic Akt/PKB signaling.
Collapse
|
21
|
Chee MJS, Douris N, Forrow AB, Monnard A, Lu S, Flaherty SE, Adams AC, Maratos-Flier E. Melanin-concentrating hormone is necessary for olanzapine-inhibited locomotor activity in male mice. Eur Neuropsychopharmacol 2015; 25:1808-16. [PMID: 26092201 PMCID: PMC4609648 DOI: 10.1016/j.euroneuro.2015.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 04/07/2015] [Accepted: 05/25/2015] [Indexed: 01/01/2023]
Abstract
Olanzapine (OLZ), an atypical antipsychotic, can be effective in treating patients with restricting type anorexia nervosa who exercise excessively. Clinical improvements include weight gain and reduced pathological hyperactivity. However the neuronal populations and mechanisms underlying OLZ actions are not known. We studied the effects of OLZ on hyperactivity using male mice lacking the hypothalamic neuropeptide melanin-concentrating hormone (MCHKO) that are lean and hyperactive. We compared the in vivo effects of systemic or intra-accumbens nucleus (Acb) OLZ administration on locomotor activity in WT and MCHKO littermates. Acute systemic OLZ treatment in WT mice significantly reduced locomotor activity, an effect that is substantially attenuated in MCHKO mice. Furthermore, OLZ infusion directly into the Acb of WT mice reduced locomotor activity, but not in MCHKO mice. To identify contributing neuronal mechanisms, we assessed the effect of OLZ treatment on Acb synaptic transmission ex vivo and in vitro. Intraperitoneal OLZ treatment reduced Acb GABAergic activity in WT but not MCHKO neurons. This effect was also seen in vitro by applying OLZ to acute brain slices. OLZ reduced the frequency and amplitude of GABAergic activity that was more robust in WT than MCHKO Acb. These findings indicate that OLZ reduced Acb GABAergic transmission and that MCH is necessary for the hypolocomotor effects of OLZ.
Collapse
Affiliation(s)
- Melissa J S Chee
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Nicholas Douris
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Avery B Forrow
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Arnaud Monnard
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Shuangyu Lu
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Stephen E Flaherty
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew C Adams
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Eleftheria Maratos-Flier
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
22
|
Chometton S, Cvetkovic-Lopes V, Houdayer C, Franchi G, Mariot A, Poncet F, Fellmann D, Risold PY. Anatomical organization of MCH connections with the pallidum and dorsal striatum in the rat. Front Syst Neurosci 2014; 8:185. [PMID: 25324738 PMCID: PMC4181234 DOI: 10.3389/fnsys.2014.00185] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 09/11/2014] [Indexed: 12/27/2022] Open
Abstract
Neurons producing the melanin-concentrating hormone (MCH) are distributed in the posterior hypothalamus, but project massively throughout the forebrain. Many aspects regarding the anatomical organization of these projections are still obscure. The present study has two goals: first to characterize the topographical organization of neurons projecting into the cholinergic basal forebrain (globus pallidus, medial septal complex), and second to verify if MCH neurons may indirectly influence the dorsal striatum (caudoputamen) by innervating afferent sources to this structure. In the first series of experiments, the retrograde tracer fluorogold was injected into multiple sites in the pallidal and medial septal regions and the distribution of retrogradely labeled neurons were analyzed in the posterior lateral hypothalamus. In the second series of experiments, fluorogold was injected into the caudoputamen, and the innervation by MCH axons of retrogradely labeled cells was analyzed. Our results revealed that the MCH system is able to interact with the basal nuclei in several different ways. First, MCH neurons provide topographic inputs to the globus pallidus, medial septal complex, and substantia innominata. Second, striatal projecting neurons in the cortex, thalamus, and substantia nigra presumably receive only sparse inputs from MCH neurons. Third, the subthalamic nucleus is heavily innervated by MCH projections, thus, presumably serves as one important intermediate station to mediate MCH influence on other parts of the basal nuclei.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Pierre-Yves Risold
- EA3922, SFR FED 4234, UFR Sciences Médicales et Pharmaceutiques, Université de Franche-ComtéBesançon, France
| |
Collapse
|
23
|
Monge-Roffarello B, Labbe SM, Roy MC, Lemay ML, Coneggo E, Samson P, Lanfray D, Richard D. The PVH as a site of CB1-mediated stimulation of thermogenesis by MC4R agonism in male rats. Endocrinology 2014; 155:3448-58. [PMID: 24949658 DOI: 10.1210/en.2013-2092] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The present study was designed to investigate the involvement of the cannabinoid receptor 1 (CB1) in the stimulating effects of the melanocortin-4 receptor (MC4R) agonism on whole-body and brown adipose tissue (BAT) thermogenesis. In a first series of experiments, whole-body and BAT thermogenesis were investigated in rats infused in the third ventricle of the brain with the MC4R agonist melanotan II (MTII) and the CB1 agonist δ9-tetrahydrocannabinol (δ(9)-THC) or the CB1 antagonist AM251. Whole-body thermogenesis was measured by indirect calorimetry and BAT thermogenesis assessed from interscapular BAT (iBAT) temperature. δ(9)-THC blunted the effects of MTII on energy expenditure and iBAT temperature, whereas AM251 tended to potentiate the MTII effects. δ(9)-THC also blocked the stimulating effect of MTII on (14)C-bromopalmitate and (3)H-deoxyglucose uptakes in iBAT. Additionally, δ(9)-THC attenuated the stimulating effect of MTII on the expression of peroxisome proliferator-activated receptor-γ coactivator 1-α (Pgc1α), type II iodothyronine deiodinase (Dio2), carnitine palmitoyltransferase 1B (Cpt1b), and uncoupling protein 1 (Ucp1). In a second series of experiments, we addressed the involvement of the paraventricular hypothalamic nucleus (PVH) in the CB1-mediated effects of MTII on iBAT thermogenesis, which were assessed following the infusion of MTII in the PVH and δ(9)-THC or AM251 in the fourth ventricle of the brain. We demonstrated the ability of δ(9)-THC to blunt MTII-induced iBAT temperature elevation. δ(9)-THC also blocked the PVH effect of MTII on (14)C-bromopalmitate uptake as well as on Pgc1α and Dio2 expression in iBAT. Altogether the results of this study demonstrate the involvement of the PVH in the CB1-mediated stimulating effects of the MC4R agonist MTII on whole-body and BAT thermogenesis.
Collapse
MESH Headings
- Adipose Tissue, Brown/metabolism
- Animals
- Male
- Paraventricular Hypothalamic Nucleus/metabolism
- Peptides, Cyclic/metabolism
- Piperidines
- Pyrazoles
- Rats
- Rats, Wistar
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Melanocortin, Type 4/agonists
- Receptor, Melanocortin, Type 4/metabolism
- Thermogenesis
- alpha-MSH/analogs & derivatives
- alpha-MSH/metabolism
Collapse
Affiliation(s)
- Boris Monge-Roffarello
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Canada G1V 4G5
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Naufahu J, Cunliffe AD, Murray JF. The roles of melanin-concentrating hormone in energy balance and reproductive function: Are they connected? Reproduction 2013; 146:R141-50. [PMID: 23884861 DOI: 10.1530/rep-12-0385] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Melanin-concentrating hormone (MCH) is an anabolic neuropeptide with multiple and diverse physiological functions including a key role in energy homoeostasis. Rodent studies have shown that the ablation of functional MCH results in a lean phenotype, increased energy expenditure and resistance to diet-induced obesity. These findings have generated interest among pharmaceutical companies vigilant for potential anti-obesity agents. Nutritional status affects reproductive physiology and behaviours, thereby optimising reproductive success and the ability to meet energetic demands. This complex control system entails the integration of direct or indirect peripheral stimuli with central effector systems and involves numerous mediators. A role for MCH in the reproductive axis has emerged, giving rise to the premise that MCH may serve as an integratory mediator between those discrete systems that regulate energy balance and reproductive function. Hence, this review focuses on published evidence concerning i) the role of MCH in energy homoeostasis and ii) the regulatory role of MCH in the reproductive axis. The question as to whether the MCH system mediates the integration of energy homoeostasis with the neuroendocrine reproductive axis and, if so, by what means has received limited coverage in the literature; evidence to date and current theories are summarised herein.
Collapse
Affiliation(s)
- Jane Naufahu
- Department of Human and Health Sciences, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK and
| | | | | |
Collapse
|
25
|
Abstract
The propensity to select and consume palatable nutrients is strongly influenced by the rewarding effects of food. Neural processes integrating reward, emotional states and decision-making can supersede satiety signals to promote excessive caloric intake and weight gain. While nutritional habits are influenced by reward-based neural mechanisms, nutrition and its impact on energy metabolism, in turn, plays an important role in the control of food reward. Feeding modulates the release of metabolic hormones that have an important influence on central controls of appetite. Nutrients themselves are also an essential source of energy fuel, while serving as key metabolites and acting as signalling molecules in the neural pathways that control feeding and food reward. Along these lines, this review discusses the impact of nutritionally regulated hormones and select macronutrients on the behavioural and neural processes underlying the rewarding effects of food.
Collapse
|
26
|
Stengel A, Rivier J, Taché Y. Central actions of somatostatin-28 and oligosomatostatin agonists to prevent components of the endocrine, autonomic and visceral responses to stress through interaction with different somatostatin receptor subtypes. Curr Pharm Des 2013; 19:98-105. [PMID: 22950508 DOI: 10.2174/13816128130114] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 07/30/2012] [Indexed: 01/23/2023]
Abstract
Somatostatin was discovered four decades ago and since then its physiological role has been extensively investigated, first in relation with its inhibitory effect on growth hormone secretion but soon it expanded to extrapituitary actions influencing various stressresponsive systems. Somatostatin is expressed in distinct brain nuclei and binds to five somatostatin receptor subtypes which are also widely expressed in the brain with a distinct distribution pattern. The last few years witnessed the discovery of highly selective peptide somatostatin receptor agonists and antagonists representing valuable tools to delineate the respective pathways of somatostatin signaling. Here we review the centrally mediated actions of somatostatin and related selective somatostatin receptor subtype agonists to influence the endocrine, autonomic, and visceral components of the stress response and basal behavior as well as thermogenesis.
Collapse
Affiliation(s)
- Andreas Stengel
- CURE: Digestive Diseases Research Center and Center for Neurovisceral Sciences & Women's Health, Digestive Diseases Division, David Geffen School of Medicine at UCLA and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | | | | |
Collapse
|
27
|
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
|
28
|
MacNeil DJ. The role of melanin-concentrating hormone and its receptors in energy homeostasis. Front Endocrinol (Lausanne) 2013; 4:49. [PMID: 23626585 PMCID: PMC3631741 DOI: 10.3389/fendo.2013.00049] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 04/09/2013] [Indexed: 01/25/2023] Open
Abstract
Extensive studies in rodents with melanin-concentrating hormone (MCH) have demonstrated that the neuropeptide hormone is a potent orexigen. Acutely, MCH causes an increase in food intake, while chronically it leads to increased weight gain, primarily as an increase in fat mass. Multiple knockout mice models have confirmed the importance of MCH in modulating energy homeostasis. Animals lacking MCH, MCH-containing neurons, or the MCH receptor all are resistant to diet-induced obesity. These genetic and pharmacologic studies have prompted a large effort to identify potent and selective MCH receptor antagonists, initially as tool compounds to probe pharmacology in models of obesity, with an ultimate goal to identify novel anti-obesity drugs. In animal models, MCH antagonists have consistently shown efficacy in reducing food intake acutely and inhibiting body-weight gain when given chronically. Five compounds have proceeded into clinical testing. Although they were reported as well-tolerated, none has advanced to long-term efficacy and safety studies.
Collapse
Affiliation(s)
- Douglas J. MacNeil
- Department of In Vitro Pharmacology, Merck Research LaboratoriesKenilworth, NJ, USA
- *Correspondence: Douglas J. MacNeil, Department of In Vitro Pharmacology, Merck Research Laboratories, K15-3-309D, 2015 Galloping Hill Road, Kenilworth, NJ 07033, USA. e-mail:
| |
Collapse
|
29
|
Barson JR, Morganstern I, Leibowitz SF. Complementary roles of orexin and melanin-concentrating hormone in feeding behavior. Int J Endocrinol 2013; 2013:983964. [PMID: 23935621 PMCID: PMC3727095 DOI: 10.1155/2013/983964] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 06/21/2013] [Indexed: 11/18/2022] Open
Abstract
Transcribed within the lateral hypothalamus, the neuropeptides orexin/hypocretin (OX) and melanin-concentrating hormone (MCH) both promote palatable food intake and are stimulated by palatable food. While these two neuropeptides share this similar positive relationship with food, recent evidence suggests that this occurs through different albeit complementary effects on behavior, with OX promoting food seeking and motivation for palatable food and MCH functioning during ongoing food intake, reinforcing the consumption of calorically dense foods. Further differences are evident in their effects on physiological processes, which are largely opposite in nature. For example, activation of OX receptors, which is neuronally excitatory, promotes waking, increases energy expenditure, and enhances limbic dopamine levels and reward. In contrast, activation of MCH receptors, which is neuronally inhibitory, promotes paradoxical sleep, enhances energy conservation, reduces limbic dopamine, and increases depressive behavior. This review describes these different effects of the neuropeptides, developing the hypothesis that they stimulate the consumption of palatable food through excessive seeking in the case of OX and through excessive energy conservation in the case of MCH. It proposes that OX initiates food intake and subsequently stimulates MCH which then acts to prolong the consumption of palatable, energy-dense food.
Collapse
Affiliation(s)
- Jessica R. Barson
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Irene Morganstern
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Sarah F. Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
- *Sarah F. Leibowitz:
| |
Collapse
|
30
|
Harrold JA, Dovey TM, Blundell JE, Halford JC. CNS regulation of appetite. Neuropharmacology 2012; 63:3-17. [DOI: 10.1016/j.neuropharm.2012.01.007] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 01/05/2012] [Accepted: 01/10/2012] [Indexed: 12/14/2022]
|
31
|
Morganstern I, Barson JR, Leibowitz SF. Regulation of drug and palatable food overconsumption by similar peptide systems. ACTA ACUST UNITED AC 2012; 4:163-73. [PMID: 21999690 DOI: 10.2174/1874473711104030163] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Revised: 08/04/2011] [Accepted: 08/04/2011] [Indexed: 11/22/2022]
Abstract
This review is aimed at understanding some of the common neurochemical, behavioral and physiological determinants of drug and food overconsumption. Much current work has been devoted to determining the similarities between the brain circuits controlling excessive use of addictive drugs and the overconsumption of palatable foods. The brain systems involved likely include peptides of both mesolimbic and hypothalamic origin. Evidence gathered from expression and injection studies suggests that the consumption of drugs, such as ethanol and nicotine, and also of palatable foods rich in fat is stimulated by different orexigenic peptides, such as enkephalin, galanin, orexin, and melaninconcentrating hormone, acting within the hypothalamus or various limbic structures, while another peptide, neuropeptide Y, is closely related to carbohydrate consumption and shows an inverse relationship with ethanol and nicotine consumption. Moreover, studies in animal models suggest that a propensity to overconsume these reinforcing substances may result from preexisting disturbances in these same peptide systems. These neurochemical disturbances, in turn, may also be closely linked to specific behaviors associated with excessive consummatory behavior, such as hyperactivity or novelty-seeking, palatable food preference, and also fluctuations in circulating lipid levels. Clear understanding of the relationship between these various determinants of consummatory behavior will allow researchers to effectively predict and examine at early stages of exposure animals that are prone to drug and food overconsumption. This work may ultimately aid in the identification of inherent traits that increase the risk for drug abuse and palatable food overconsumption.
Collapse
Affiliation(s)
- Irene Morganstern
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA
| | | | | |
Collapse
|
32
|
Lopes APF, Ganzer L, Borges AC, Kochenborger L, Januário AC, Faria MS, Marino-Neto J, Paschoalini MA. Effects of GABA ligands injected into the nucleus accumbens shell on fear/anxiety-like and feeding behaviours in food-deprived rats. Pharmacol Biochem Behav 2012; 101:41-8. [DOI: 10.1016/j.pbb.2011.11.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 10/24/2011] [Accepted: 11/15/2011] [Indexed: 10/15/2022]
|
33
|
Tuziak SM, Volkoff H. A preliminary investigation of the role of melanin-concentrating hormone (MCH) and its receptors in appetite regulation of winter flounder (Pseudopleuronectes americanus). Mol Cell Endocrinol 2012; 348:281-96. [PMID: 21945816 DOI: 10.1016/j.mce.2011.09.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 08/25/2011] [Accepted: 09/07/2011] [Indexed: 10/17/2022]
Abstract
In order to better understand the role of melanin-concentrating hormone (MCH) in the regulation of appetite in fish, the mRNAs of two forms of MCH, prepro-MCH and MCH2, and two forms of MCH receptors, MCH-R1 and MCH-R2, were isolated from winter flounder (Pseudopleuronectes americanus). In addition, the mRNA expressions of these peptides and their receptors were determined under fed and fasted conditions. Both MCHs are expressed in forebrain and midbrain, as well as peripheral tissues including gut and gonads. Both MCH-Rs are ubiquitously expressed in the brain and periphery. Fasting induced an increase in the expression levels of MCH and MCH-R1 mRNAs in optic tectum/thalamus and hypothalamus but had no effect on either MCH2 or MCH-R2 mRNA expressions. Our results suggest that MCH and MCH-R1, but not MCH2 and MCH-R2 might have a role in the regulation of appetite in flounder.
Collapse
Affiliation(s)
- Sarah M Tuziak
- Department of Biology, Memorial University of Newfoundland, St. John's, Newfoundland, Canada A1B-3X9
| | | |
Collapse
|
34
|
Lopez CA, Guesdon B, Baraboi ED, Roffarello BM, Hétu M, Richard D. Involvement of the opioid system in the orexigenic and hedonic effects of melanin-concentrating hormone. Am J Physiol Regul Integr Comp Physiol 2011; 301:R1105-11. [DOI: 10.1152/ajpregu.00076.2011] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Melanin-concentrating hormone (MCH) exerts an orexigenic effect that resembles that of opioids, suggesting that the MCH and opioid systems could interact in controlling the food intake behavior. Three series of experiments were conducted in male Wistar rats: 1) to test the ability of the κ-, μ-, and δ-opioid receptor antagonists binaltorphimine (nor-BNI-κ), β-funaltrexamine (β-FNA-μ), and naltrindole (NTI-δ), respectively, to block the stimulating effects of MCH on food intake; 2) to verify the ability of MCH to induce a positive hedonic response to a sweet stimulus when injected into the nucleus accumbens shell (NAcSh) or right lateral ventricle (LV) of the brain; and 3) to assess the ability of nor-BNI, β-FNA, and NTI to block the effects of MCH on the hedonic response to a sweet stimulus. Nor-BNI, NTI (0, 10 and 40 nmol), and β-FNA (0, 10 and 50 nmol) were administered into the LV prior to injecting MCH (2.0 nmol). To assess the hedonic response, rats were implanted with an intraoral cannula allowing for the infusion of a sweet solution into the oral cavity. Food intake was assessed in sated rats during the first 3 h following the MCH or vehicle (i.e., artificial cerebrospinal fluid) injection. The hedonic response to a sweet stimulus was assessed by examining facial mimics, following the intraoral administration of a sucrose solution. Blockade of each of the three opioid receptors by selective antagonists prevented MCH-induced feeding. Furthermore, MCH-injections into the NAcSh and right LV resulted in enhanced hedonic responses. Finally, antagonism of the three opioid receptors blunted the LV-injected, MCH-induced, facial-liking expressions in response to an intraoral sweet stimulus. Overall, the present study provides evidence to link the MCH and opioid systems in the food intake behavior.
Collapse
Affiliation(s)
- Carlos Andres Lopez
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Canada
| | - Benjamin Guesdon
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Canada
| | - Elena-Dana Baraboi
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Canada
| | - Boris Monge Roffarello
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Canada
| | - Marylène Hétu
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Canada
| | - Denis Richard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Canada
| |
Collapse
|
35
|
van Dijk G, Evers SS, Guidotti S, Thornton SN, Scheurink AJ, Nyakas C. The lateral hypothalamus: A site for integration of nutrient and fluid balance. Behav Brain Res 2011; 221:481-7. [DOI: 10.1016/j.bbr.2011.01.047] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Revised: 01/20/2011] [Accepted: 01/27/2011] [Indexed: 11/26/2022]
|
36
|
Bittencourt JC. Anatomical organization of the melanin-concentrating hormone peptide family in the mammalian brain. Gen Comp Endocrinol 2011; 172:185-97. [PMID: 21463631 DOI: 10.1016/j.ygcen.2011.03.028] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 03/23/2011] [Accepted: 03/28/2011] [Indexed: 11/24/2022]
Abstract
More than 20 years ago, melanin-concentrating hormone (MCH) and its peptide family members - neuropeptide EI (NEI) and neuropeptide GE (NGE) - were described in various species, including mammals (rodents, humans, and non-human primates). Since then, most studies have focused on the role of MCH as an orexigenic peptide, as well as on its participation in learning, spatial memory, neuroendocrine control, and sleep. It has been shown that MCH mRNA or the neuropeptide MCH are present in neurons of the prosencephalon, hypothalamus and brainstem. However, most of the neurons containing MCH/NEI are within the incerto-hypothalamic and lateral hypothalamic areas. In addition, the terminals of those neurons are distributed widely throughout the central nervous system. In this review, we will discuss the relationship between those territories and the roles played by MCH/NEI, as well as the importance of MCH receptor 1 in the respective terminal fields. Certain neurochemical features of MCH- and NEI-immunoreactive (MCH-ir and NEI-ir) neurons will also be discussed. The overarching theme is the anatomical organization of an inhibitory neuropeptide colocalized with an inhibitory neurotransmitter in integrative territories of the central nervous system, such as the IHy and LHA. Although these territories have connections to few brain regions, the regions to which they are connected are relevant, being responsible for the organization of motivated behaviors. All available information on this peptidergic system (anatomical, neurochemical, hodological, physiological, pharmacological and behavioral data) suggests that MCH is intimately involved in arousal and the initiation of motivated behaviors.
Collapse
Affiliation(s)
- Jackson C Bittencourt
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil.
| |
Collapse
|
37
|
Mul JD, la Fleur SE, Toonen PW, Afrasiab-Middelman A, Binnekade R, Schetters D, Verheij MMM, Sears RM, Homberg JR, Schoffelmeer ANM, Adan RAH, DiLeone RJ, De Vries TJ, Cuppen E. Chronic loss of melanin-concentrating hormone affects motivational aspects of feeding in the rat. PLoS One 2011; 6:e19600. [PMID: 21573180 PMCID: PMC3088702 DOI: 10.1371/journal.pone.0019600] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 04/11/2011] [Indexed: 12/20/2022] Open
Abstract
Current epidemic obesity levels apply great medical and financial pressure to the strenuous economy of obesity-prone cultures, and neuropeptides involved in body weight regulation are regarded as attractive targets for a possible treatment of obesity in humans. The lateral hypothalamus and the nucleus accumbens shell (AcbSh) form a hypothalamic-limbic neuropeptide feeding circuit mediated by Melanin-Concentrating Hormone (MCH). MCH promotes feeding behavior via MCH receptor-1 (MCH1R) in the AcbSh, although this relationship has not been fully characterized. Given the AcbSh mediates reinforcing properties of food, we hypothesized that MCH modulates motivational aspects of feeding. Here we show that chronic loss of the rat MCH-precursor Pmch decreased food intake predominantly via a reduction in meal size during rat development and reduced high-fat food-reinforced operant responding in adult rats. Moreover, acute AcbSh administration of Neuropeptide-GE and Neuropeptide-EI (NEI), both additional neuropeptides derived from Pmch, or chronic intracerebroventricular infusion of NEI, did not affect feeding behavior in adult pmch+/+ or pmch−/− rats. However, acute administration of MCH to the AcbSh of adult pmch−/− rats elevated feeding behavior towards wild type levels. Finally, adult pmch−/− rats showed increased ex vivo electrically evoked dopamine release and increased limbic dopamine transporter levels, indicating that chronic loss of Pmch in the rat affects the limbic dopamine system. Our findings support the MCH-MCH1R system as an amplifier of consummatory behavior, confirming this system as a possible target for the treatment of obesity. We propose that MCH-mediated signaling in the AcbSh positively mediates motivational aspects of feeding behavior. Thereby it provides a crucial signal by which hypothalamic neural circuits control energy balance and guide limbic brain areas to enhance motivational or incentive-related aspects of food consumption.
Collapse
Affiliation(s)
- Joram D. Mul
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Susanne E. la Fleur
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Pim W. Toonen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anthonieke Afrasiab-Middelman
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboud University, Nijmegen, The Netherlands
| | - Rob Binnekade
- Department of Anatomy and Neurosciences, Center for Neurogenomics and Cognitive Research, Free University Medical Center, Amsterdam, The Netherlands
| | - Dustin Schetters
- Department of Anatomy and Neurosciences, Center for Neurogenomics and Cognitive Research, Free University Medical Center, Amsterdam, The Netherlands
| | - Michel M. M. Verheij
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition, and Behavior, Radboud University, Nijmegen, The Netherlands
| | - Robert M. Sears
- Department of Psychiatry, Ribicoff Research Facilities, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Judith R. Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboud University, Nijmegen, The Netherlands
| | - Anton N. M. Schoffelmeer
- Department of Anatomy and Neurosciences, Center for Neurogenomics and Cognitive Research, Free University Medical Center, Amsterdam, The Netherlands
| | - Roger A. H. Adan
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ralph J. DiLeone
- Department of Psychiatry, Ribicoff Research Facilities, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Taco J. De Vries
- Department of Anatomy and Neurosciences, Center for Neurogenomics and Cognitive Research, Free University Medical Center, Amsterdam, The Netherlands
| | - Edwin Cuppen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
38
|
Barson JR, Morganstern I, Leibowitz SF. Similarities in hypothalamic and mesocorticolimbic circuits regulating the overconsumption of food and alcohol. Physiol Behav 2011; 104:128-37. [PMID: 21549731 DOI: 10.1016/j.physbeh.2011.04.054] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 04/26/2011] [Indexed: 12/26/2022]
Abstract
Historically, studies of food intake regulation started with the hypothalamus and gradually expanded to mesocorticolimbic regions, while studies of drug use began with mesocorticolimbic regions and now include the hypothalamus. As research on ingestive behavior has progressed, it has uncovered more and more similarities between the regulation of palatable food and drug intake. It has also identified specific neurochemicals involved in palatable food and drug intake. Hypothalamic orexigenic neurochemicals specifically involved in controlling fat ingestion, including galanin, enkephalin, orexin and melanin-concentrating hormone, show positive feedback with this macronutrient, with these peptides both increasing fat intake and being further stimulated by its intake. This positive relationship offers some explanation for why foods high in fat are so often overconsumed. Research in Bart Hoebel's laboratory in conjunction with our own has shown that consumption of ethanol, a drug of abuse that also contains calories, is similarly driven by these neurochemical systems involved in fat intake, consistent with evidence closely relating fat and ethanol consumption. Both fat and ethanol intake are also regulated by dopamine and acetylcholine acting in mesocorticolimbic nuclei. This close relationship of fat and ethanol is likely driven in part by circulating lipids, which are increased by fat and ethanol intake, known to increase expression and levels of the neurochemicals, and found to promote further intake of fat and ethanol. Compellingly, recent studies suggest that these systems may already be dysregulated in animals prone to consuming excess fat or ethanol, even before they have ever been exposed to these substances. Further understanding of these systems involved in consummatory behavior will allow researchers to develop effective therapies for the treatment of overeating as well as drug abuse.
Collapse
Affiliation(s)
- Jessica R Barson
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA
| | | | | |
Collapse
|
39
|
Morganstern I, Chang GQ, Chen YW, Barson JR, Zhiyu Y, Hoebel BG, Leibowitz SF. Role of melanin-concentrating hormone in the control of ethanol consumption: Region-specific effects revealed by expression and injection studies. Physiol Behav 2010; 101:428-37. [PMID: 20670637 PMCID: PMC2949500 DOI: 10.1016/j.physbeh.2010.07.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Revised: 05/27/2010] [Accepted: 07/21/2010] [Indexed: 11/23/2022]
Abstract
The peptide melanin-concentrating hormone (MCH), produced mainly by cells in the lateral hypothalamus (LH), perifornical area (PF) and zona incerta (ZI), is suggested to have a role in the consumption of rewarding substances, such as ethanol, sucrose and palatable food. However, there is limited information on the specific brain sites where MCH acts to stimulate intake of these rewarding substances and on the feedback effects that their consumption has on the expression of endogenous MCH. The current study investigated MCH in relation to ethanol consumption, in Sprague-Dawley rats. In Experiment 1, chronic consumption of ethanol (from 0.70 to 2.7 g/kg/day) dose-dependently reduced MCH gene expression in the LH. In Experiments 2-4, the opposite effect was observed with acute oral ethanol, which stimulated MCH expression specifically in the LH but not the ZI. In Experiment 5, the effect of MCH injection in brain-cannulated rats on ethanol consumption was examined. Compared to saline, MCH injected in the paraventricular nucleus (PVN) and nucleus accumbens (NAc) selectively stimulated ethanol consumption without affecting food or water intake. In contrast, it reduced ethanol intake when administered into the LH, while having no effect in the ZI. These results demonstrate that voluntary, chronic consumption of ethanol leads to local negative feedback control of MCH expression in the LH. However, with a brief exposure, ethanol stimulates MCH-expressing neurons in this region, which through projections to the feeding-related PVN and reward-related NAc can promote further drinking behavior.
Collapse
Affiliation(s)
- I Morganstern
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Stengel A, Coskun T, Goebel M, Wang L, Craft L, Alsina-Fernandez J, Rivier J, Taché Y. Central injection of the stable somatostatin analog ODT8-SST induces a somatostatin2 receptor-mediated orexigenic effect: role of neuropeptide Y and opioid signaling pathways in rats. Endocrinology 2010; 151:4224-35. [PMID: 20610566 PMCID: PMC2940496 DOI: 10.1210/en.2010-0195] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Somatostatin and octreotide injected into the brain have been reported to modulate food intake. However, little is known regarding the underlying mechanisms. The stable oligosomatostatin analog, des-AA(1,2,4,5,12,13)-[DTrp(8)]-somatostatin (ODT8-SST), like somatostatin, binds to all five somatostatin receptors (sst(1-5)). We characterized the effects of ODT8-SST injected intracerebroventricularly (i.c.v.) on food consumption and related mechanisms of action in freely fed rats. ODT8-SST (0.3 and 1 microg per rat, i.c.v.) injected during the light or dark phase induced an early onset (within 1 h) and long-lasting (4 h) increase in food intake in nonfasted rats. By contrast, i.p. injection (0.3-3 mg/kg) or i.c.v. injection of selective sst(1) or sst(4) agonists (1 microg per rat) had no effect. The 2 h food intake response during the light phase was blocked by i.c.v. injection of a sst(2) antagonist, the neuropeptide Y (NPY) Y(1) receptor antagonist, BIBP-3226, and ip injection of the mu-opioid receptor antagonist, naloxone, and not associated with changes in plasma ghrelin levels. ODT8-SST (1 microg per rat, i.c.v.) stimulated gastric emptying of a solid meal which was also blocked by naloxone. The increased food intake was accompanied by a sustained increase in respiratory quotient, energy expenditure, and drinking as well as mu-opioid receptor-independent grooming behavior and hyperthermia, while ambulatory movements were not altered after ODT8-SST (1 microg per rat, i.c.v.). These data show that ODT8-SST acts primarily through brain sst(2) receptors to induce a long-lasting orexigenic effect that involves the activation of Y(1) and opiate-receptors, accompanied by enhanced gastric transit and energy expenditure suggesting a modulation of NPYergic and opioidergic orexigenic systems by brain sst(2) receptors.
Collapse
MESH Headings
- Animals
- Anti-Anxiety Agents/administration & dosage
- Anti-Anxiety Agents/pharmacology
- Arginine/administration & dosage
- Arginine/analogs & derivatives
- Arginine/pharmacology
- Body Temperature/drug effects
- Eating/drug effects
- Eating/physiology
- Energy Metabolism/drug effects
- Gastric Emptying/drug effects
- Grooming/drug effects
- Injections, Intraperitoneal
- Injections, Intraventricular
- Male
- Mice
- Mice, Inbred C57BL
- Naloxone/administration & dosage
- Naloxone/pharmacology
- Peptide Fragments/administration & dosage
- Peptide Fragments/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, Neuropeptide Y/antagonists & inhibitors
- Receptors, Neuropeptide Y/physiology
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/physiology
- Receptors, Somatostatin/agonists
- Receptors, Somatostatin/antagonists & inhibitors
- Receptors, Somatostatin/physiology
- Somatostatin/administration & dosage
- Somatostatin/analogs & derivatives
- Somatostatin/pharmacology
Collapse
Affiliation(s)
- Andreas Stengel
- Department of Medicine, Center for Ulcer Research and Education, Digestive Diseases Division, University of California Los Angeles, and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California 90073, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abbenante G, Becker B, Blanc S, Clark C, Condie G, Fraser G, Grathwohl M, Halliday J, Henderson S, Lam A, Liu L, Mann M, Muldoon C, Pearson A, Premraj R, Ramsdale T, Rossetti T, Schafer K, Le Thanh G, Tometzki G, Vari F, Verquin G, Waanders J, West M, Wimmer N, Yau A, Zuegg J, Meutermans W. Biological Diversity from a Structurally Diverse Library: Systematically Scanning Conformational Space Using a Pyranose Scaffold. J Med Chem 2010; 53:5576-86. [DOI: 10.1021/jm1002777] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
| | - Bernd Becker
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | | | - Chris Clark
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | - Glenn Condie
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | | | | | - Judy Halliday
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | | | - Ann Lam
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | - Ligong Liu
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | - Maretta Mann
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | - Craig Muldoon
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | - Andrew Pearson
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | | | | | - Tony Rossetti
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | - Karl Schafer
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | - Giang Le Thanh
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | | | - Frank Vari
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | | | | | - Michael West
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | - Norbert Wimmer
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | - Annika Yau
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | - Johannes Zuegg
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| | - Wim Meutermans
- Alchemia Ltd, Eight Mile Plains, Queensland 4113, Australia
| |
Collapse
|
42
|
Paglialunga S, Fisette A, Munkonda M, Gao Y, Richard D, Cianflone K. The effects of acylation stimulating protein supplementation VS antibody neutralization on energy expenditure in wildtype mice. BMC PHYSIOLOGY 2010; 10:4. [PMID: 20416070 PMCID: PMC2875207 DOI: 10.1186/1472-6793-10-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Accepted: 04/23/2010] [Indexed: 11/10/2022]
Abstract
BACKGROUND Acylation stimulating protein (ASP) is an adipogenic hormone that stimulates triglyceride (TG) synthesis and glucose transport in adipocytes. Previous studies have shown that ASP-deficient C3 knockout mice are hyperphagic yet lean, as they display increased oxygen consumption and fatty acid oxidation compared to wildtype mice. In the present study, antibodies against ASP (Anti-ASP) and human recombinant ASP (rASP) were tested in vitro and in vivo. Continuous administration for 4 weeks via osmotic mini-pump of Anti-ASP or rASP was evaluated in wildtype mice on a high-fat diet (HFD) to examine their effects on body weight, food intake and energy expenditure. RESULTS In mature murine adipocytes, rASP significantly stimulated fatty acid uptake (+243% vs PBS, P < 0.05) while Anti-ASP neutralized the rASP response. Mice treated with Anti-ASP showed elevated energy expenditure (P < 0.0001), increased skeletal muscle glucose oxidation (+141%, P < 0.001), reduced liver glycogen (-34%, P < 0.05) and glucose-6-phosphate content (-64%, P = 0.08) compared to control mice. There was no change in body weight, food intake, fasting insulin, adiponectin, CRP or TG levels compared to controls. Interestingly, HFD mice treated with rASP showed the opposite phenotype with reduced energy expenditure (P < 0.0001) and increased body weight (P < 0.05), cumulative food intake (P < 0.0001) and liver glycogen content (+59%, P < 0.05). Again, there was no change in circulating insulin, adiponectin, CRP or TG levels, however, plasma free fatty acids were reduced (-48%, P < 0.05). CONCLUSION In vitro, Anti-ASP effectively neutralized ASP stimulated fatty acid uptake. In vivo, Anti-ASP treatment increased whole body energy utilization while rASP increased energy storage. Therefore, ASP is a potent anabolic hormone that may also be a mediator of energy expenditure.
Collapse
Affiliation(s)
- Sabina Paglialunga
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, G1V 4G5, Canada
| | | | | | | | | | | |
Collapse
|
43
|
Mul JD, Yi CX, van den Berg SAA, Ruiter M, Toonen PW, van der Elst MCJ, Voshol PJ, Ellenbroek BA, Kalsbeek A, la Fleur SE, Cuppen E. Pmch expression during early development is critical for normal energy homeostasis. Am J Physiol Endocrinol Metab 2010; 298:E477-88. [PMID: 19934402 DOI: 10.1152/ajpendo.00154.2009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Postnatal development and puberty are times of strong physical maturation and require large quantities of energy. The hypothalamic neuropeptide melanin-concentrating hormone (MCH) regulates nutrient intake and energy homeostasis, but the underlying mechanisms are not completely understood. Here we use a novel rat knockout model in which the MCH precursor Pmch has been inactivated to study the effects of loss of MCH on energy regulation in more detail. Pmch(-/-) rats are lean, hypophagic, osteoporotic, and although endocrine parameters were changed in pmch(-/-) rats, endocrine dynamics were normal, indicating an adaptation to new homeostatic levels rather than disturbed metabolic mechanisms. Detailed body weight growth and feeding behavior analysis revealed that Pmch expression is particularly important during early rat development and puberty, i.e., the first 8 postnatal weeks. Loss of Pmch resulted in a 20% lower set point for body weight that was determined solely during this period and remained unchanged during adulthood. Although the final body weight is diet dependent, the Pmch-deficiency effect was similar for all diets tested in this study. Loss of Pmch affected energy expenditure in both young and adult rats, although these effects seem secondary to the observed hypophagia. Our findings show an important role for Pmch in energy homeostasis determination during early development and indicate that the MCH receptor 1 system is a plausible target for childhood obesity treatment, currently a major health issue in first world countries.
Collapse
Affiliation(s)
- Joram D Mul
- Hubrecht Institute-Koninklijke Nederlandse Akademie van Wetenschappen and University Medical Center Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Blouet C, Schwartz GJ. Hypothalamic nutrient sensing in the control of energy homeostasis. Behav Brain Res 2009; 209:1-12. [PMID: 20035790 DOI: 10.1016/j.bbr.2009.12.024] [Citation(s) in RCA: 220] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Accepted: 12/16/2009] [Indexed: 12/15/2022]
Abstract
The hypothalamus is a center of convergence and integration of multiple nutrient-related signals. It can sense changes in circulating adiposity hormones, gastric hormones and nutrients, and receives neuroanatomical projections from other nutrient sensors, mainly within the brainstem. The hypothalamus also integrates these signals with various cognitive forebrain-descending information and reward/motivation-related signals coming from the midbrain-dopamine system, to coordinate neuroendocrine, behavioral and metabolic effectors of energy balance. Some of the key nutrient-sensing hypothalamic neurons have been identified in the arcuate, the ventro-medial and the lateral nuclei of the hypothalamus, and the molecular mechanisms underlying intracellular integration of nutrient-related signals in these neurons are currently under intensive investigation. However, little is known about the neural pathways downstream from hypothalamic nutrient sensors, and how they drive effectors of energy homeostasis under physiological conditions. This manuscript will review recent progress from molecular, genetic and neurophysiological studies that identify and characterize the critical intracellular signalling pathways and neurocircuits involved in determining hypothalamic nutrient detection, and link these circuits to behavioral and metabolic effectors of energy balance. We will provide a critical analysis of current data to identify ongoing challenges for future research in this field.
Collapse
Affiliation(s)
- Clémence Blouet
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| | | |
Collapse
|
45
|
Glick M, Segal-Lieberman G, Cohen R, Kronfeld-Schor N. Chronic MCH infusion causes a decrease in energy expenditure and body temperature, and an increase in serum IGF-1 levels in mice. Endocrine 2009; 36:479-85. [PMID: 19859841 DOI: 10.1007/s12020-009-9252-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Accepted: 09/11/2009] [Indexed: 10/20/2022]
Abstract
Melanin concentrating hormone (MCH) is an orexigenic peptide secreted from the lateral hypothalamus. Various observations suggest a role for MCH in energy expenditure in transgenic mice; however, the influence of MCH on energy expenditure and body temperature in WT mice was inadequately studied. Therefore, our first goal was to characterize the influence of chronic intracerebroventrical MCH infusion on energy homeostasis in mice. Our second goal was to explore the effect of MCH on the GH-insulin like growth factor 1 (IGF-1) axis in vivo. We have recently published that MCH directly increased GH-secretion from pituitary cells in vitro, suggesting that MCH may exert part of its effects on energy balance via direct pituitary hormone regulation. Mice were centrally infused with MCH for 14 days, resulting in a significant increase in food intake, body weight, fat mass and plasma IGF-1 levels, while decreasing body temperature and energy expenditure. Our data emphasize the role of MCH as a key regulator of energy homeostasis by means of appetite regulation, regulation of energy expenditure, and an integrator of energy balance with the neuroendocrine system regulating pituitary hormone secretion. They also support the notion that MCH may have a physiologic role in GH regulation that may, in turn, contribute to its effect on body weight.
Collapse
Affiliation(s)
- Moran Glick
- Department of Zoology, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | | | |
Collapse
|
46
|
Pissios P. Animals models of MCH function and what they can tell us about its role in energy balance. Peptides 2009; 30:2040-4. [PMID: 19447150 PMCID: PMC2977959 DOI: 10.1016/j.peptides.2009.05.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 05/04/2009] [Accepted: 05/05/2009] [Indexed: 11/16/2022]
Abstract
Melanin-concentrating hormone (MCH) has attracted considerable attention because of its effects on food intake and body weight and the MCH receptor (MCHR1) remains one of the viable targets for obesity therapy. This review summarizes the literature examining the effects of MCH on body weight, food intake and energy expenditure in rodent models, and the central sites where MCH acts in regulating energy homeostasis. Emphasis is given on the discrepancies between the genetic and pharmacologic models of MCHR1 inactivation. We propose some solutions to resolve these discrepancies and discuss some future directions in MCH research.
Collapse
Affiliation(s)
- Pavlos Pissios
- Beth Israel Deaconess Medical Center, Boston, MA 02446, United States.
| |
Collapse
|
47
|
Griffond B, Risold PY. MCH and feeding behavior-interaction with peptidic network. Peptides 2009; 30:2045-51. [PMID: 19619600 DOI: 10.1016/j.peptides.2009.07.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 04/17/2009] [Accepted: 07/09/2009] [Indexed: 12/20/2022]
Abstract
Numerous works associate the MCH peptide, and the hypothalamic neurons that produce it, to the feeding behavior and energy homeostasis. It is commonly admitted that MCH is an orexigenic peptide, and MCH neurons could be under the control of arcuate NPY and POMC neurons. However, the literature data is not always concordant. In particular questions about the intrahypothalamic circuit involving other neuropeptides and about the mechanisms through which MCH could act are not yet clearly answered. For example, which receptors mediate a MCH response to NPY or alpha-MSH, does MCH act alone, is there any local anatomical organization within the tuberal LHA? A review of the current literature is then needed to help focus attention on these unresolved and often neglected issues.
Collapse
Affiliation(s)
- B Griffond
- Université de Franche-Comté, Besançon, France
| | | |
Collapse
|
48
|
Ito M, Ishihara A, Gomori A, Egashira S, Matsushita H, Mashiko S, Ito J, Ito M, Nakase K, Haga Y, Iwaasa H, Suzuki T, Ohtake N, Moriya M, Sato N, MacNeil DJ, Takenaga N, Tokita S, Kanatani A. Melanin-concentrating hormone 1-receptor antagonist suppresses body weight gain correlated with high receptor occupancy levels in diet-induced obesity mice. Eur J Pharmacol 2009; 624:77-83. [PMID: 19836369 DOI: 10.1016/j.ejphar.2009.10.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 07/18/2009] [Accepted: 10/06/2009] [Indexed: 11/25/2022]
Abstract
Melanin-concentrating hormone (MCH), which is a neuropeptide expressed in the hypothalamus of the brain, is involved in regulating feeding behavior and energy homeostasis via the MCH(1) receptor in rodents. It is widely considered that MCH(1) receptor antagonists are worthy of development for medical treatment of obesity. Here we report on the development of an ex vivo receptor occupancy assay using a new radiolabeled MCH(1) receptor antagonist, [(35)S]-compound D. An MCH(1) receptor antagonist inhibited the binding of [(35)S]-compound D to brain slices in a dose-dependent manner. The result showed a good correlation between the receptor occupancy levels and plasma or brain levels of the MCH(1) receptor antagonist, suggesting that the ex vivo receptor binding assay using this radioligand is practical. Quantitative analysis in diet-induced obese mice showed that the efficacy of body weight reduction correlated with the receptor occupancy levels at 24h. Furthermore, more than 90% occupancy levels of MCH(1) receptor antagonists during 24h post-dosing are required for potent efficacy on body weight reduction. The present occupancy assay could be a useful pharmacodynamic marker to quantitatively estimate anti-obese efficacy, and would accelerate the development of MCH(1) receptor antagonists for treatment of obesity.
Collapse
Affiliation(s)
- Masahiko Ito
- Tsukuba Research Institute, Banyu Pharmaceutical Co., Ltd., Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|