1
|
Du YY, Yao MX, Yu HX, Mo HL, Yang QY, Yu JJ, Wang LX, Zhou JS, Li Y. Molecular cloning, tissue distribution, and pharmacologic function of melanocortin-3 receptor in common carp (Cyprinus carpio). Gen Comp Endocrinol 2023; 330:114149. [PMID: 36336108 DOI: 10.1016/j.ygcen.2022.114149] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
Melanocortin-3 receptor (MC3R) not only regulates energy homeostasis in animals, but also is an important regulator of inflammation. As one of the most widely farmed freshwater fish, common carp has attracted great interest for its feeding and inflammation regulation. In this study, we cloned the coding sequence (CDS) of common carp Mc3r (ccMc3r), examined its tissue expression profile, and investigated the function of this receptor in mediating downstream signaling pathways. The results showed that the CDS of ccMc3r was 975 bp, encoding a putative protein of 324 amino acids. Homology, phylogeny, and chromosomal synteny analyses revealed that ccMc3r is evolutionarily close to the orthologs of cyprinids. Quantitative real-time PCR (qPCR) indicated that ccMc3r was highly expressed in the brain and intestine. The luciferase reporter systems showed that four ligands, ACTH (1-24), α-MSH, β-MSH, and NDP-MSH, were able to activate the cAMP and MAPK/ERK signaling pathways downstream of ccMc3r with different potencies. For the cAMP signaling pathway, ACTH (1-24) had the highest activation potency; while for the MAPK/ERK signaling pathway, β-MSH had the greatest activation effect. In addition, we found that the four agonists were able to inhibit TNF-α-induced NF-κB signaling in approximately the same order of potency as cAMP signaling activation. This study may facilitate future studies on the role of Mc3r in common carp feed efficiency and immune regulation.
Collapse
Affiliation(s)
- Yu-You Du
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ming-Xing Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hui-Xia Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hao-Lin Mo
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Qi-Yuan Yang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jia-Jia Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Li-Xin Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ji-Shu Zhou
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
2
|
Ji RL, Jiang SS, Tao YX. Modulation of Canine Melanocortin-3 and -4 Receptors by Melanocortin-2 Receptor Accessory Protein 1 and 2. Biomolecules 2022; 12:biom12111608. [PMID: 36358958 PMCID: PMC9687446 DOI: 10.3390/biom12111608] [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: 10/12/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
The neural melanocortin receptors (MCRs), melanocortin-3 and -4 receptors (MC3R and MC4R), have crucial roles in regulating energy homeostasis. The melanocortin-2 receptor accessory proteins (MRAPs, MRAP1 and MRAP2) have been shown to regulate neural MCRs in a species-specific manner. The potential effects of MRAP1 and MRAP2 on canine neural MCRs have not been investigated before. Herein, we cloned canine (c) MC3R and identified one canine MRAP2 splice variant, MRAP2b, with N-terminal extension of cMRAP2a. Canine MC3R showed higher maximal responses to five agonists than those of human MC3R. We further investigated the modulation of cMRAP1, cMRAP2a, and cMRAP2b, on cMC3R and cMC4R pharmacology. For the cMC3R, all MRAPs had no effect on trafficking; cMRAP1 significantly decreased Bmax whereas cMRAP2a and cMRAP2b significantly increased Bmax. Both MRAP1 and MRAP2a decreased Rmaxs in response to α-MSH and ACTH; MRAP2b only decreased α-MSH-stimulated cAMP generation. For the MC4R, MRAP1 and MRAP2a increased cell surface expression, and MRAP1 and MRAP2a increased Bmaxs. All MRAPs had increased affinities to α-MSH and ACTH. MRAP2a increased ACTH-induced cAMP levels, whereas MRAP2b decreased α-MSH- and ACTH-stimulated cAMP production. These findings may lead to a better understanding of the regulation of neural MCRs by MRAP1 and MRAP2s.
Collapse
|
3
|
Melanocortin-5 Receptor: Pharmacology and Its Regulation of Energy Metabolism. Int J Mol Sci 2022; 23:ijms23158727. [PMID: 35955857 PMCID: PMC9369360 DOI: 10.3390/ijms23158727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 12/12/2022] Open
Abstract
As the most recent melanocortin receptor (MCR) identified, melanocortin-5 receptor (MC5R) has unique tissue expression patterns, pharmacological properties, and physiological functions. Different from the other four MCR subtypes, MC5R is widely distributed in both the central nervous system and peripheral tissues and is associated with multiple functions. MC5R in sebaceous and preputial glands regulates lipid production and sexual behavior, respectively. MC5R expressed in immune cells is involved in immunomodulation. Among the five MCRs, MC5R is the predominant subtype expressed in skeletal muscle and white adipose tissue, tissues critical for energy metabolism. Activated MC5R triggers lipid mobilization in adipocytes and glucose uptake in skeletal muscle. Therefore, MC5R is a potential target for treating patients with obesity and diabetes mellitus. Melanocortin-2 receptor accessory proteins can modulate the cell surface expression, dimerization, and pharmacology of MC5R. This minireview summarizes the molecular and pharmacological properties of MC5R and highlights the progress made on MC5R in energy metabolism. We poInt. out knowledge gaps that need to be explored in the future.
Collapse
|
4
|
Ji RL, Tao YX. Regulation of Melanocortin-3 and -4 Receptors by Isoforms of Melanocortin-2 Receptor Accessory Protein 1 and 2. Biomolecules 2022; 12:biom12020244. [PMID: 35204745 PMCID: PMC8961526 DOI: 10.3390/biom12020244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 01/27/2023] Open
Abstract
The neural melanocortin receptors (MCRs), melanocortin-3 and -4 receptors (MC3R and MC4R), play essential non-redundant roles in the regulation of energy homeostasis. Interaction of neural MCRs and melanocortin-2 receptor accessory proteins (MRAPs, MRAP1 and MRAP2) is suggested to play pivotal roles in MC3R and MC4R signaling. In the present study, we identified two new human (h) MRAP2 splice variants, MRAP2b (465 bp open reading frame) and MRAP2c (381 bp open reading frame). Human MRAP2s are different in C-termini. We investigated the effects of five isoforms of MRAPs, hMRAP1a, hMRAP1b, hMRAP2a, hMRAP2b, and hMRAP2c, on MC3R and MC4R pharmacology. At the hMC3R, hMRAP1a and hMRAP2c increased and hMRAP1b decreased the cell surface expression. hMRAP1a increased affinity to ACTH. Four MRAPs (hMRAP1a, hMRAP1b, hMRAP2a, and hMRAP2c) decreased the maximal responses in response to α-MSH and ACTH. For hMC4R, hMRAP1a, hMRAP2a, and hMRAP2c increased the cell surface expression of hMC4R. Human MRAP1b significantly increased affinity to ACTH while MRAP2a decreased affinity to ACTH. Human MRAP1a increased ACTH potency. MRAPs also affected hMC4R basal activities, with hMRAP1s increasing and hMRAP2s decreasing the basal activities. In summary, the newly identified splicing variants, hMRAP2b and hMRAP2c, could regulate MC3R and MC4R pharmacology. The two MRAP1s and three MRAP2s had differential effects on MC3R and MC4R trafficking, binding, and signaling. These findings led to a better understanding of the regulation of neural MCRs by MRAP1s and MRAP2s.
Collapse
|
5
|
Ji RL, Huang L, Wang Y, Liu T, Fan SY, Tao M, Tao YX. Topmouth culter melanocortin-3 receptor: regulation by two isoforms of melanocortin-2 receptor accessory protein 2. Endocr Connect 2021; 10:1489-1501. [PMID: 34678761 PMCID: PMC8630771 DOI: 10.1530/ec-21-0459] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022]
Abstract
Melanocortin-3 receptor (MC3R) is a regulator of energy homeostasis, and interaction of MC3R and melanocortin-2 receptor accessory protein 2 (MRAP2) plays a critical role in MC3R signaling of mammals. However, the physiological roles of MC3R in teleosts are not well understood. In this study, qRT-PCR was used to measure gene expression. Radioligand binding assay was used to study the binding properties of topmouth culter MC3R (caMC3R). Intracellular cAMP generation was determined by RIA, and caMC3R expression was quantified with flow cytometry. We showed that culter mc3r had higher expression in the CNS. All agonists could bind and stimulate caMC3R to increase dose dependently intracellular cAMP accumulation. Compared to human MC3R, culter MC3R showed higher constitutive activity, higher efficacies, and Rmax to alpha-melanocyte-stimulating hormone (α-MSH), des-α-MSH, and adrenocorticotrophic hormone. Both caMRAP2a and caMRAP2b markedly decreased caMC3R basal cAMP production. However, only caMRAP2a significantly decreased cell surface expression, Bmax, and Rmax of caMC3R. Expression analysis suggested that MRAP2a and MRAP2b might be more important in regulating MC3R/MC4R signaling during larval period, and reduced mc3r, mc4r, and pomc expression might be primarily involved in modulation of MC3R/MC4R in adults. These data indicated that the cloned caMC3R was a functional receptor. MRAP2a and MRAP2b had different effects on expression and signaling of caMC3R. In addition, expression analysis suggested that MRAP2s, receptors, and hormones might play different roles in regulating culter development and growth.
Collapse
Affiliation(s)
- Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Lu Huang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan, People’s Republic of China
| | - Yin Wang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Ting Liu
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Si-Yu Fan
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan, People’s Republic of China
| | - Min Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan, People’s Republic of China
- Correspondence should be addressed to M Tao or Y-X Tao: or
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
- Correspondence should be addressed to M Tao or Y-X Tao: or
| |
Collapse
|
6
|
Li X, Wang H. Multiple organs involved in the pathogenesis of non-alcoholic fatty liver disease. Cell Biosci 2020; 10:140. [PMID: 33372630 PMCID: PMC7720519 DOI: 10.1186/s13578-020-00507-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/27/2020] [Indexed: 02/08/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents the leading cause of chronic liver disease worldwide and the anticipated health burden is huge. There are limited therapeutic approaches for NAFLD now. It’s imperative to get a better understanding of the disease pathogenesis if new treatments are to be discovered. As the hepatic manifestation of metabolic syndrome, this disease involves complex interactions between different organs and regulatory pathways. It’s increasingly clear that brain, gut and adipose tissue all contribute to NAFLD pathogenesis and development, in view of their roles in energy homeostasis. In the present review, we try to summarize currently available data regarding NAFLD pathogenesis and to lay a particular emphasis on the inter-organ crosstalk evidence.
Collapse
Affiliation(s)
- Xiaoyan Li
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China. .,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, 230032, China.
| |
Collapse
|
7
|
Micioni Di Bonaventura E, Botticelli L, Tomassoni D, Tayebati SK, Micioni Di Bonaventura MV, Cifani C. The Melanocortin System behind the Dysfunctional Eating Behaviors. Nutrients 2020; 12:E3502. [PMID: 33202557 PMCID: PMC7696960 DOI: 10.3390/nu12113502] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022] Open
Abstract
The dysfunction of melanocortin signaling has been associated with obesity, given the important role in the regulation of energy homeostasis, food intake, satiety and body weight. In the hypothalamus, the melanocortin-3 receptor (MC3R) and melanocortin-4 receptor (MC4R) contribute to the stability of these processes, but MC3R and MC4R are also localized in the mesolimbic dopamine system, the region that responds to the reinforcing properties of highly palatable food (HPF) and where these two receptors seem to affect food reward and motivation. Loss of function of the MC4R, resulting from genetic mutations, leads to overeating in humans, but to date, a clear understanding of the underlying mechanisms and behaviors that promote overconsumption of caloric foods remains unknown. Moreover, the MC4R demonstrated to be a crucial modulator of the stress response, factor that is known to be strictly related to binge eating behavior. In this review, we will explore the preclinical and clinical studies, and the controversies regarding the involvement of melanocortin system in altered eating patterns, especially binge eating behavior, food reward and motivation.
Collapse
Affiliation(s)
| | - Luca Botticelli
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| | - Daniele Tomassoni
- School of Bioscience and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy;
| | - Seyed Khosrow Tayebati
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| | | | - Carlo Cifani
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| |
Collapse
|
8
|
Seoane-Collazo P, Martínez-Sánchez N, Milbank E, Contreras C. Incendiary Leptin. Nutrients 2020; 12:nu12020472. [PMID: 32069871 PMCID: PMC7071158 DOI: 10.3390/nu12020472] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/06/2020] [Accepted: 02/08/2020] [Indexed: 02/08/2023] Open
Abstract
Leptin is a hormone released by adipose tissue that plays a key role in the control of energy homeostasis through its binding to leptin receptors (LepR), mainly expressed in the hypothalamus. Most scientific evidence points to leptin’s satiating effect being due to its dual capacity to promote the expression of anorexigenic neuropeptides and to reduce orexigenic expression in the hypothalamus. However, it has also been demonstrated that leptin can stimulate (i) thermogenesis in brown adipose tissue (BAT) and (ii) the browning of white adipose tissue (WAT). Since the demonstration of the importance of BAT in humans 10 years ago, its study has aroused great interest, mainly in the improvement of obesity-associated metabolic disorders through the induction of thermogenesis. Consequently, several strategies targeting BAT activation (mainly in rodent models) have demonstrated great potential to improve hyperlipidemias, hepatic steatosis, insulin resistance and weight gain, leading to an overall healthier metabolic profile. Here, we review the potential therapeutic ability of leptin to correct obesity and other metabolic disorders, not only through its satiating effect, but by also utilizing its thermogenic properties.
Collapse
Affiliation(s)
- Patricia Seoane-Collazo
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain;
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
- Correspondence: (P.S.-C.); (N.M.-S.); (C.C.); Tel.: +81-298-533-301 (P.S.-C.); +34-913-941-650 (N.M.-S.); +44-01865285890 (C.C.)
| | - Noelia Martínez-Sánchez
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
- Correspondence: (P.S.-C.); (N.M.-S.); (C.C.); Tel.: +81-298-533-301 (P.S.-C.); +34-913-941-650 (N.M.-S.); +44-01865285890 (C.C.)
| | - Edward Milbank
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain;
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Cristina Contreras
- Department of Physiology, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain
- Correspondence: (P.S.-C.); (N.M.-S.); (C.C.); Tel.: +81-298-533-301 (P.S.-C.); +34-913-941-650 (N.M.-S.); +44-01865285890 (C.C.)
| |
Collapse
|
9
|
González-García I, Milbank E, Martinez-Ordoñez A, Diéguez C, López M, Contreras C. HYPOTHesizing about central comBAT against obesity. J Physiol Biochem 2019; 76:193-211. [PMID: 31845114 DOI: 10.1007/s13105-019-00719-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 12/02/2019] [Indexed: 12/12/2022]
Abstract
The hypothalamus is a brain region in charge of many vital functions. Among them, BAT thermogenesis represents an essential physiological function to maintain body temperature. In the metabolic context, it has now been established that energy expenditure attributed to BAT function can contribute to the energy balance in a substantial extent. Thus, therapeutic interest in this regard has increased in the last years and some studies have shown that BAT function in humans can make a real contribution to improve diabetes and obesity-associated diseases. Nevertheless, how the hypothalamus controls BAT activity is still not fully understood. Despite the fact that much has been known about the mechanisms that regulate BAT activity in recent years, and that the central regulation of thermogenesis offers a very promising target, many questions remain still unsolved. Among them, the possible human application of knowledge obtained from rodent studies, and drug administration strategies able to specifically target the hypothalamus. Here, we review the current knowledge of homeostatic regulation of BAT, including the molecular insights of brown adipocytes, its central control, and its implication in the development of obesity.
Collapse
Affiliation(s)
- Ismael González-García
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.
| | - Edward Milbank
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain
| | - Anxo Martinez-Ordoñez
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain
| | - Carlos Diéguez
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain
| | - Miguel López
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain
| | - Cristina Contreras
- Department of Physiology, Pharmacy School, Complutense University of Madrid, 28040, Madrid, Spain.
| |
Collapse
|
10
|
Yu DY, Wu RZ, Zhao Y, Nie ZH, Wei L, Wang TY, Liu ZP. Polymorphisms of four candidate genes and their correlations with growth traits in blue fox (Alopex lagopus). Gene 2019; 717:143987. [PMID: 31362037 DOI: 10.1016/j.gene.2019.143987] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 07/16/2019] [Accepted: 07/16/2019] [Indexed: 11/16/2022]
Abstract
To improve the accuracy and genetic progress of blue fox breeding, the relationships between genetic polymorphisms and growth and reproductive traits of the blue fox were investigated. MC4R, MC3R, INHA and INHBA were selected as candidate genes for molecular evolution and statistical analyses. Single-factor variance analyses showed that the MC4R (g.267C > T, g.423C > T, and g.731C > A) and MC3R (g.677C > T) genotypes had significant impacts on body weight, chest circumference, abdominal perimeter and body mass index (BMI) (P < 0.05) in blue fox. The MC4R and MC3R combined genotypes had significant effects on the body weight and abdominal circumference. The different genotypes of INHA g.75G > A had significant effects on female fecundity, whereas the different genotypes of INHBA g.404G > T and g.467G > T and the INHA and INHBA combined genotypes had significant effects on male fecundity. The proteins encoded by the open reading frames (ORFs) of different polymorphic loci were predicted and analysed. The aims of this study were to identify genetic markers related to growth and reproduction in the blue fox and to provide an efficient, economical and accurate theoretical approach for auxiliary fox breeding.
Collapse
Affiliation(s)
- Dong-Yue Yu
- College of Wildlife Resources, Northeast Forestry University, Harbin, China
| | - Ru-Zi Wu
- College of Wildlife Resources, Northeast Forestry University, Harbin, China
| | - Yao Zhao
- College of Wildlife Resources, Northeast Forestry University, Harbin, China
| | - Zi-Han Nie
- College of Wildlife Resources, Northeast Forestry University, Harbin, China
| | - Lai Wei
- Hualong Blue Fox Breeding Company, Harbin, China
| | - Tian-Yi Wang
- Hualong Blue Fox Breeding Company, Harbin, China
| | - Zhi-Ping Liu
- College of Wildlife Resources, Northeast Forestry University, Harbin, China.
| |
Collapse
|
11
|
Yang LK, Zhang ZR, Wen HS, Tao YX. Characterization of channel catfish (Ictalurus punctatus) melanocortin-3 receptor reveals a potential network in regulation of energy homeostasis. Gen Comp Endocrinol 2019; 277:90-103. [PMID: 30905760 DOI: 10.1016/j.ygcen.2019.03.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 03/12/2019] [Accepted: 03/18/2019] [Indexed: 11/19/2022]
Abstract
The melanocortin-3 receptor (MC3R) is known to be involved in regulation of energy homeostasis, regulating feed efficiency and nutrient partitioning in mammals. Its physiological roles in non-mammalian vertebrates, especially economically important aquaculture species, are not well understood. Channel catfish (Ictalurus punctatus) is the main freshwater aquaculture species in North America. In this study, we characterized the channel catfish MC3R. The mc3r of channel catfish encoded a putative protein (ipMC3R) of 367 amino acids. We transfected HEK293T cells with ipMC3R plasmid for functional studies. Five agonists, including adrenocorticotropin, α-melanocyte stimulating hormone (α-MSH), β-MSH, [Nle4, D-Phe7]-α-MSH, and D-Trp8-γ-MSH, were used in the pharmacological studies. Our results showed that ipMC3R bound β-MSH with higher affinity and D-Trp8-γ-MSH with lower affinity compared with human MC3R. All agonists could stimulate ipMC3R and increase intracellular cAMP production with sub-nanomolar potencies. The extracellular signal-regulated kinases 1 and 2 (ERK1/2) activation could also be triggered by ipMC3R. The ipMC3R exhibited constitutive activities in both cAMP and ERK1/2 pathways, and Agouti-related protein served as an inverse agonist at ipMC3R, potently inhibiting the high basal cAMP level. Moreover, we showed that melanocortin receptor accessory protein 2 (MRAP2) preferentially modulated ipMC3R in cAMP production rather than ERK1/2 activation. Our study will assist further investigation of the physiological roles of the ipMC3R, especially in energy homeostasis, in channel catfish.
Collapse
Affiliation(s)
- Li-Kun Yang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Zheng-Rui Zhang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States; Key Laboratory of Marine Genetics and Breeding, Ministry of Education, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Hai-Shen Wen
- College of Fisheries, Ocean University of China, Qingdao, China
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
12
|
Gaspar RC, Muñoz VR, Kuga GK, Nakandakari SCBR, Minuzzi LG, Botezelli JD, da Silva ASR, Cintra DE, de Moura LP, Ropelle ER, Pauli JR. Acute physical exercise increases leptin-induced hypothalamic extracellular signal-regulated kinase1/2 phosphorylation and thermogenesis of obese mice. J Cell Biochem 2018; 120:697-704. [PMID: 30206970 DOI: 10.1002/jcb.27426] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/12/2018] [Indexed: 11/12/2022]
Abstract
The obesity is a result of energy imbalance and the increase in thermogenesis seems an interesting alternative for the treatment of this disease. The mechanism of energy expenditure through thermogenesis is tightly articulated in the hypothalamus by leptin. The hypothalamic extracellular signal-regulated kinase-1/2 (ERK1/2) is a key mediator of the thermoregulatory effect of leptin and mediates the sympathetic signal to the brown adipose tissue (BAT). In this context, physical exercise is one of the main interventions for the treatment of obesity. Thus, this study aimed to verify the effects of acute physical exercise on leptin-induced hypothalamic ERK1/2 phosphorylation and thermogenesis in obese mice. Here we showed that acute physical exercise reduced the fasting glucose of obese mice and increased leptin-induced hypothalamic p-ERK1/2 and uncoupling protein 1 (UCP1) content in BAT ( P < 0.05). These molecular changes are accompanied by an increased oxygen uptake (VO 2 ) and heat production in obese exercised mice ( P < 0.05). The increased energy expenditure in the obese exercised animals occurred independently of changes in spontaneous activity. Thus, this is the first study demonstrating that acute physical exercise can increase leptin-induced hypothalamic ERK1/2 phosphorylation and energy expenditure of obese mice.
Collapse
Affiliation(s)
- Rafael Calais Gaspar
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Vitor Rosetto Muñoz
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Gabriel Keine Kuga
- Department of Physical Education, Post-graduate Program in Movement Sciences, São Paulo State University (UNESP), Rio Claro, São Paulo, Brazil
| | | | - Luciele Guerra Minuzzi
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,Research Center for Sport and Physical Activity (IUD/DTP/04213/2016), Faculty of Sports Science and Physical Education, University of Coimbra, Portugal
| | - José Diego Botezelli
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Adelino S R da Silva
- School of Physical Education and Sports of Ribeirão Preto, Post-graduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, USP, Ribeirão Preto, São Paulo, Brazil
| | - Dennys Esper Cintra
- Laboratory of Nutritional Genomics (LabGeN), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil
| | - Leandro Pereira de Moura
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,Laboratory of Nutritional Genomics (LabGeN), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil.,CEPECE - Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil.,CEPECE - Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil.,CEPECE - Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| |
Collapse
|
13
|
Michael NJ, Simonds SE, van den Top M, Cowley MA, Spanswick D. Mitochondrial uncoupling in the melanocortin system differentially regulates NPY and POMC neurons to promote weight-loss. Mol Metab 2017; 6:1103-1112. [PMID: 29031712 PMCID: PMC5641603 DOI: 10.1016/j.molmet.2017.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/29/2017] [Accepted: 07/05/2017] [Indexed: 12/13/2022] Open
Abstract
Objective The mitochondrial uncoupling agent 2,4-dinitrophenol (DNP), historically used as a treatment for obesity, is known to cross the blood-brain-barrier, but its effects on central neural circuits controlling body weight are largely unknown. As hypothalamic melanocortin neuropeptide Y/agouti-related protein (NPY/AgRP) and pro-opiomelanocortin (POMC) neurons represent key central regulators of food intake and energy expenditure we investigated the effects of DNP on these neurons, food intake and energy expenditure. Method C57BL/6 and melanocortin-4 receptor (MC4R) knock-out mice were administered DNP intracerebroventricularly (ICV) and the metabolic changes were characterized. The specific role of NPY and POMC neurons and the ionic mechanisms mediating the effects of uncoupling were examined with in vitro electrophysiology performed on NPY hrGFP or POMC eGFP mice. Results Here we show DNP-induced differential effects on melanocortin neurons including inhibiting orexigenic NPY and activating anorexigenic POMC neurons through independent ionic mechanisms coupled to mitochondrial function, consistent with an anorexigenic central effect. Central administration of DNP induced weight-loss, increased BAT thermogenesis and browning of white adipose tissue, and decreased food intake, effects that were absent in MC4R knock-out mice and blocked by the MC4R antagonist, AgRP. Conclusion These data show a novel central anti-obesity mechanism of action of DNP and highlight the potential for selective melanocortin mitochondrial uncoupling to target metabolic disorders. Mitochondrial uncoupling of the melanocortin system with DNP induced weight-loss. DNP inhibited NPY neurones via activation of ATP-sensitive potassium channels. DNP activated POMC neurones via block of inwardly rectifying potassium channels. Central DNP reduced food intake and increased WAT browning and BAT thermogenesis.
Collapse
Affiliation(s)
- Natalie Jane Michael
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Australia(5).
| | - Stephanie Elise Simonds
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Australia(5).
| | | | - Michael Alexander Cowley
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Australia(5).
| | - David Spanswick
- Neuroscience Program, Biomedicine Discovery Institute, Monash University, Australia(5); Neurosolutions, Coventry, P.O. 3517, UK; Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK.
| |
Collapse
|
14
|
Xu P, Zhu L, Saito K, Yang Y, Wang C, He Y, Yan X, Hyseni I, Tong Q, Xu Y. Melanocortin 4 receptor is not required for estrogenic regulations on energy homeostasis and reproduction. Metabolism 2017; 70:152-159. [PMID: 28403939 PMCID: PMC5407306 DOI: 10.1016/j.metabol.2016.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/02/2016] [Accepted: 12/05/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND Brain estrogen receptor-α (ERα) is essential for estrogenic regulation of energy homeostasis and reproduction. We previously showed that ERα expressed by pro-opiomelanocortin (POMC) neurons mediates estrogen's effects on food intake, body weight, negative regulation of hypothalamic-pituitary-gonadal axis (HPG axis) and fertility. RESULTS AND CONCLUSIONS We report here that global deletion of a key downstream receptor for POMC peptide, the melanocortin 4 receptor (MC4R), did not affect normal negative feedback regulation of estrogen on the HPG axis, estrous cyclicity and female fertility. Furthermore, loss of the MC4R did not influence estrogenic regulation on food intake and body weight. These results indicate that the MC4R is not required for estrogen's effects on metabolic and reproductive functions.
Collapse
Affiliation(s)
- Pingwen Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030.
| | - Liangru Zhu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Kenji Saito
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Yongjie Yang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Chunmei Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Yanlin He
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Xiaofeng Yan
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Ilirjana Hyseni
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030.
| |
Collapse
|
15
|
Yang LK, Tao YX. Biased signaling at neural melanocortin receptors in regulation of energy homeostasis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2486-2495. [PMID: 28433713 DOI: 10.1016/j.bbadis.2017.04.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/09/2017] [Accepted: 04/10/2017] [Indexed: 12/15/2022]
Abstract
The global prevalence of obesity highlights the importance of understanding on regulation of energy homeostasis. The central melanocortin system is an important intersection connecting the neural pathways controlling satiety and energy expenditure to regulate energy homeostasis by sensing and integrating the signals of external stimuli. In this system, neural melanocortin receptors (MCRs), melanocortin-3 and -4 receptors (MC3R and MC4R), play crucial roles in the regulation of energy homeostasis. Recently, multiple intracellular signaling pathways and biased signaling at neural MCRs have been discovered, providing new insights into neural MCR signaling. This review attempts to summarize biased signaling including biased receptor mutants (both naturally occurring and lab-generated) and biased ligands at neural MCRs, and to provide a better understanding of obesity pathogenesis and new therapeutic implications for obesity treatment.
Collapse
Affiliation(s)
- Li-Kun Yang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
16
|
Contreras C, Nogueiras R, Diéguez C, Rahmouni K, López M. Traveling from the hypothalamus to the adipose tissue: The thermogenic pathway. Redox Biol 2017; 12:854-863. [PMID: 28448947 PMCID: PMC5406580 DOI: 10.1016/j.redox.2017.04.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/08/2017] [Accepted: 04/11/2017] [Indexed: 01/13/2023] Open
Abstract
Brown adipose tissue (BAT) is a specialized tissue critical for non-shivering thermogenesis producing heat through mitochondrial uncoupling; whereas white adipose tissue (WAT) is responsible of energy storage in the form of triglycerides. Another type of fat has been described, the beige adipose tissue; this tissue emerges in existing WAT depots but with thermogenic ability, a phenomenon known as browning. Several peripheral signals relaying information about energy status act in the brain, particularly the hypothalamus, to regulate thermogenesis in BAT and browning of WAT. Different hypothalamic areas have the capacity to regulate the thermogenic process in brown and beige adipocytes through the sympathetic nervous system (SNS). This review discusses important concepts and discoveries about the central control of thermogenesis as a trip that starts in the hypothalamus, and taking the sympathetic roads to reach brown and beige fat to modulate thermogenic functions.
Collapse
Affiliation(s)
- Cristina Contreras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain.
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa, Iowa City, Iowa 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242, USA
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain.
| |
Collapse
|
17
|
Demidowich AP, Jun JY, Yanovski JA. Polymorphisms and mutations in the melanocortin-3 receptor and their relation to human obesity. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2468-2476. [PMID: 28363697 DOI: 10.1016/j.bbadis.2017.03.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 12/18/2022]
Abstract
Inactivating mutations in the melanocortin 3 receptor (Mc3r) have been described as causing obesity in mice, but the physiologic effects of MC3R mutations in humans have been less clear. Here we review the MC3R polymorphisms and mutations identified in humans, and the in vitro, murine, and human cohort studies examining their putative effects. Some, but not all, studies suggest that the common human MC3R variant T6K+V81I, as well as several other rare, function-altering mutations, are associated with greater adiposity and hyperleptinemia with altered energy partitioning. In vitro, the T6K+V81I variant appears to decrease MC3R expression and therefore cAMP generation in response to ligand binding. Knockin mouse studies confirm that the T6K+V81I variant increases feeding efficiency and the avidity with which adipocytes derived from bone or adipose tissue stem cells store triglycerides. Other MC3R mutations occur too infrequently in the human population to make definitive conclusions regarding their clinical effects. This article is part of a Special Issue entitled: Melanocortin Receptors - edited by Ya-Xiong Tao.
Collapse
Affiliation(s)
- Andrew P Demidowich
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Joo Yun Jun
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Jack A Yanovski
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
18
|
Contreras C, Nogueiras R, Diéguez C, Medina-Gómez G, López M. Hypothalamus and thermogenesis: Heating the BAT, browning the WAT. Mol Cell Endocrinol 2016; 438:107-115. [PMID: 27498420 DOI: 10.1016/j.mce.2016.08.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/30/2016] [Accepted: 08/01/2016] [Indexed: 12/20/2022]
Abstract
Brown adipose tissue (BAT) has been also considered as the main thermogenic organ responsible of maintenance body temperature through heat production. However, a new type of thermogenic fat has been characterized during the last years, the beige or brite fat, that is developed from white adipose tissue (WAT) in response to different stimuli by a process known as browning. The activities of brown and beige adipocytes ameliorate metabolic disease, including obesity in mice and correlate with leanness in humans. Many genes and pathways that regulate brown and beige adipocyte biology have now been identified, providing a variety of promising therapeutic targets for metabolic disease. The hypothalamus is the main central place orchestrating the outflow signals that drive the sympathetic nerve activity to BAT and WAT, controlling heat production and energy homeostasis. Recent data have revealed new hypothalamic molecular mechanisms, such as hypothalamic AMP-activated protein kinase (AMPK), that control both thermogenesis and browning. This review provides an overview of the factors influencing BAT and WAT thermogenesis, with special focus on the integration of peripheral information on hypothalamic circuits controlling thermoregulation.
Collapse
Affiliation(s)
- Cristina Contreras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain.
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Gema Medina-Gómez
- Área de Bioquímica y Biología Molecular, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28922, Alcorcón, Madrid, Spain
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain.
| |
Collapse
|
19
|
Contribution of adaptive thermogenesis to the hypothalamic regulation of energy balance. Biochem J 2016; 473:4063-4082. [DOI: 10.1042/bcj20160012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 08/13/2016] [Accepted: 08/30/2016] [Indexed: 12/12/2022]
Abstract
Obesity and its related disorders are among the most pervasive diseases in contemporary societies, and there is an urgent need for new therapies and preventive approaches. Given (i) our poor social capacity to correct unhealthy habits, and (ii) our evolutionarily genetic predisposition to store excess energy as fat, the current environment of caloric surplus makes the treatment of obesity extremely difficult. During the last few decades, an increasing number of methodological approaches have increased our knowledge of the neuroanatomical basis of the control of energy balance. Compelling evidence underlines the role of the hypothalamus as a homeostatic integrator of metabolic information and its ability to adjust energy balance. A greater understanding of the neural basis of the hypothalamic regulation of energy balance might indeed pave the way for new therapeutic targets. In this regard, it has been shown that several important peripheral signals, such as leptin, thyroid hormones, oestrogens and bone morphogenetic protein 8B, converge on common energy sensors, such as AMP-activated protein kinase to modulate sympathetic tone on brown adipose tissue. This knowledge may open new ways to counteract the chronic imbalance underlying obesity. Here, we review the current state of the art on the role of hypothalamus in the regulation of energy balance with particular focus on thermogenesis.
Collapse
|
20
|
Endospanin1 affects oppositely body weight regulation and glucose homeostasis by differentially regulating central leptin signaling. Mol Metab 2016; 6:159-172. [PMID: 28123946 PMCID: PMC5220283 DOI: 10.1016/j.molmet.2016.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 10/21/2016] [Accepted: 10/27/2016] [Indexed: 01/05/2023] Open
Abstract
The hypothalamic arcuate nucleus (ARC) is a major integration center for energy and glucose homeostasis that responds to leptin. Resistance to leptin in the ARC is an important component of the development of obesity and type 2 diabetes. Recently, we showed that Endospanin1 (Endo1) is a negative regulator of the leptin receptor (OBR) that interacts with OBR and retains the receptor inside the cell, leading to a decreased activation of the anorectic STAT3 pathway. Endo1 is up-regulated in the ARC of high fat diet (HFD)-fed mice, and its silencing in the ARC of lean and obese mice prevents and reverses the development of obesity. OBJECTIVE Herein we investigated whether decreased Endo1 expression in the hypothalamic ARC, associated with reduced obesity, could also ameliorate glucose homeostasis accordingly. METHODS We studied glucose homeostasis in lean or obese mice silenced for Endo1 in the ARC via stereotactic injection of shRNA-expressing lentiviral vectors. RESULTS We observed that despite being leaner, Endo1-silenced mice showed impaired glucose homeostasis on HFD. Mechanistically, we show that Endo1 interacts with p85, the regulatory subunit of PI3K, and mediates leptin-induced PI3K activation. CONCLUSIONS Our results thus define Endo1 as an important hypothalamic integrator of leptin signaling, and its silencing differentially regulates the OBR-dependent functions.
Collapse
Key Words
- ARC, arcuate nucleus
- BW, body weight
- CD, chow diet
- DIO, diet-induced obesity
- Diabetes
- Endo1, Endospanin1
- GTT, glucose tolerance test
- HFD, high fat diet
- Insulin
- LIF, leukemia inhibitory factor
- Leptin receptor
- OB-RGRP/Endospanin1
- OBR, leptin receptor
- Obesity
- PLA, proximity ligation assay
- T2D, type 2 diabetes
- ip, intraperitoneal
Collapse
|
21
|
Chhabra KH, Adams JM, Jones GL, Yamashita M, Schlapschy M, Skerra A, Rubinstein M, Low MJ. Reprogramming the body weight set point by a reciprocal interaction of hypothalamic leptin sensitivity and Pomc gene expression reverts extreme obesity. Mol Metab 2016; 5:869-881. [PMID: 27689000 PMCID: PMC5034612 DOI: 10.1016/j.molmet.2016.07.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 07/29/2016] [Indexed: 12/26/2022] Open
Abstract
Objective A major challenge for obesity treatment is the maintenance of reduced body weight. Diet-induced obese mice are resistant to achieving normoweight once the obesogenic conditions are reversed, in part because lowered circulating leptin leads to a reduction in metabolic rate and a rebound of hyperphagia that defend the previously elevated body weight set point. Because hypothalamic POMC is a central leptin target, we investigated whether changes in circulating leptin modify Pomc expression to maintain normal energy balance in genetically predisposed obese mice. Methods Mice with reversible Pomc silencing in the arcuate nucleus (ArcPomc−/−) become morbidly obese eating low-fat chow. We measured body composition, food intake, plasma leptin, and leptin sensitivity in ArcPomc−/− mice weight-matched to littermate controls by calorie restriction, either from weaning or after developing obesity. Pomc was reactivated by tamoxifen-dependent Cre recombinase transgenes. Long acting PASylated leptin was administered to weight-reduced ArcPomc−/− mice to mimic the super-elevated leptin levels of obese mice. Results ArcPomc−/− mice had increased adiposity and leptin levels shortly after weaning. Despite chronic calorie restriction to achieve normoweight, ArcPomc−/− mice remained moderately hyperleptinemic and resistant to exogenous leptin's effects to reduce weight and food intake. However, subsequent Pomc reactivation in weight-matched ArcPomc−/− mice normalized plasma leptin, leptin sensitivity, adiposity, and food intake. In contrast, extreme hyperleptinemia induced by PASylated leptin blocked the full restoration of hypothalamic Pomc expression in calorie restricted ArcPomc−/− mice, which consequently regained 30% of their lost body weight and attained a metabolic steady state similar to that of tamoxifen treated obese ArcPomc−/− mice. Conclusions Pomc reactivation in previously obese, calorie-restricted ArcPomc−/− mice normalized energy homeostasis, suggesting that their body weight set point was restored to control levels. In contrast, massively obese and hyperleptinemic ArcPomc−/− mice or those weight-matched and treated with PASylated leptin to maintain extreme hyperleptinemia prior to Pomc reactivation converged to an intermediate set point relative to lean control and obese ArcPomc−/− mice. We conclude that restoration of hypothalamic leptin sensitivity and Pomc expression is necessary for obese ArcPomc−/− mice to achieve and sustain normal metabolic homeostasis; whereas deficits in either parameter set a maladaptive allostatic balance that defends increased adiposity and body weight. Hypothalamic POMC-deficiency increases adiposity and induces leptin resistance. PASylated leptin blocks the normalization of Pomc expression, weight and adiposity. Interactions of leptin sensitivity and Pomc expression dictate body weight set point.
Collapse
Affiliation(s)
- Kavaljit H Chhabra
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jessica M Adams
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Graham L Jones
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Miho Yamashita
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Martin Schlapschy
- Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354 Freising (Weihenstephan), Germany
| | - Arne Skerra
- Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354 Freising (Weihenstephan), Germany
| | - Marcelo Rubinstein
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA; Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, 1428 Buenos Aires, Argentina
| | - Malcolm J Low
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
22
|
Melanocortin-4 receptor-regulated energy homeostasis. Nat Neurosci 2016; 19:206-19. [PMID: 26814590 DOI: 10.1038/nn.4202] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/13/2015] [Indexed: 12/11/2022]
Abstract
The melanocortin system provides a conceptual blueprint for the central control of energetic state. Defined by four principal molecular components--two antagonistically acting ligands and two cognate receptors--this phylogenetically conserved system serves as a prototype for hierarchical energy balance regulation. Over the last decade the application of conditional genetic techniques has facilitated the neuroanatomical dissection of the melanocortinergic network and identified the specific neural substrates and circuits that underscore the regulation of feeding behavior, energy expenditure, glucose homeostasis and autonomic outflow. In this regard, the melanocortin-4 receptor is a critical coordinator of mammalian energy homeostasis and body weight. Drawing on recent advances in neuroscience and genetic technologies, we consider the structure and function of the melanocortin-4 receptor circuitry and its role in energy homeostasis.
Collapse
|
23
|
Mutations in Melanocortin-3 Receptor Gene and Human Obesity. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 140:97-129. [DOI: 10.1016/bs.pmbts.2016.01.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
24
|
Seoane-Collazo P, Fernø J, Gonzalez F, Diéguez C, Leis R, Nogueiras R, López M. Hypothalamic-autonomic control of energy homeostasis. Endocrine 2015; 50:276-91. [PMID: 26089260 DOI: 10.1007/s12020-015-0658-y] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 06/06/2015] [Indexed: 10/23/2022]
Abstract
Regulation of energy homeostasis is tightly controlled by the central nervous system (CNS). Several key areas such as the hypothalamus and brainstem receive and integrate signals conveying energy status from the periphery, such as leptin, thyroid hormones, and insulin, ultimately leading to modulation of food intake, energy expenditure (EE), and peripheral metabolism. The autonomic nervous system (ANS) plays a key role in the response to such signals, innervating peripheral metabolic tissues, including brown and white adipose tissue (BAT and WAT), liver, pancreas, and skeletal muscle. The ANS consists of two parts, the sympathetic and parasympathetic nervous systems (SNS and PSNS). The SNS regulates BAT thermogenesis and EE, controlled by central areas such as the preoptic area (POA) and the ventromedial, dorsomedial, and arcuate hypothalamic nuclei (VMH, DMH, and ARC). The SNS also regulates lipid metabolism in WAT, controlled by the lateral hypothalamic area (LHA), VMH, and ARC. Control of hepatic glucose production and pancreatic insulin secretion also involves the LHA, VMH, and ARC as well as the dorsal vagal complex (DVC), via splanchnic sympathetic and the vagal parasympathetic nerves. Muscle glucose uptake is also controlled by the SNS via hypothalamic nuclei such as the VMH. There is recent evidence of novel pathways connecting the CNS and ANS. These include the hypothalamic AMP-activated protein kinase-SNS-BAT axis which has been demonstrated to be a key modulator of thermogenesis. In this review, we summarize current knowledge of the role of the ANS in the modulation of energy balance.
Collapse
Affiliation(s)
- Patricia Seoane-Collazo
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain.
| | - Johan Fernø
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain
- Department of Clinical Science, K. G. Jebsen Center for Diabetes Research, University of Bergen, 5021, Bergen, Norway
| | - Francisco Gonzalez
- Department of Surgery, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain
- Service of Ophthalmology, Complejo Hospitalario Universitario de Santiago de Compostela, 15706, Santiago de Compostela, Spain
| | - Carlos Diéguez
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain
| | - Rosaura Leis
- Unit of Investigation in Nutrition, Growth and Human Development of Galicia, Pediatric Department (USC), Complexo Hospitalario Universitario de Santiago (IDIS/SERGAS), Santiago de Compostela, Spain
| | - Rubén Nogueiras
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain
| | - Miguel López
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain.
| |
Collapse
|
25
|
Abstract
Thermogenesis, the production of heat energy, in brown adipose tissue is a significant component of the homeostatic repertoire to maintain body temperature during the challenge of low environmental temperature in many species from mouse to man and plays a key role in elevating body temperature during the febrile response to infection. The sympathetic neural outflow determining brown adipose tissue (BAT) thermogenesis is regulated by neural networks in the CNS which increase BAT sympathetic nerve activity in response to cutaneous and deep body thermoreceptor signals. Many behavioral states, including wakefulness, immunologic responses, and stress, are characterized by elevations in core body temperature to which central command-driven BAT activation makes a significant contribution. Since energy consumption during BAT thermogenesis involves oxidation of lipid and glucose fuel molecules, the CNS network driving cold-defensive and behavioral state-related BAT activation is strongly influenced by signals reflecting the short- and long-term availability of the fuel molecules essential for BAT metabolism and, in turn, the regulation of BAT thermogenesis in response to metabolic signals can contribute to energy balance, regulation of body adipose stores and glucose utilization. This review summarizes our understanding of the functional organization and neurochemical influences within the CNS networks that modulate the level of BAT sympathetic nerve activity to produce the thermoregulatory and metabolic alterations in BAT thermogenesis and BAT energy expenditure that contribute to overall energy homeostasis and the autonomic support of behavior.
Collapse
Affiliation(s)
- Shaun F Morrison
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon
| | | |
Collapse
|
26
|
Contreras C, Gonzalez F, Fernø J, Diéguez C, Rahmouni K, Nogueiras R, López M. The brain and brown fat. Ann Med 2015; 47:150-68. [PMID: 24915455 PMCID: PMC4438385 DOI: 10.3109/07853890.2014.919727] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 04/25/2014] [Indexed: 02/06/2023] Open
Abstract
Brown adipose tissue (BAT) is a specialized organ responsible for thermogenesis, a process required for maintaining body temperature. BAT is regulated by the sympathetic nervous system (SNS), which activates lipolysis and mitochondrial uncoupling in brown adipocytes. For many years, BAT was considered to be important only in small mammals and newborn humans, but recent data have shown that BAT is also functional in adult humans. On the basis of this evidence, extensive research has been focused on BAT function, where new molecules, such as irisin and bone morphogenetic proteins, particularly BMP7 and BMP8B, as well as novel central factors and new regulatory mechanisms, such as orexins and the canonical ventomedial nucleus of the hypothalamus (VMH) AMP- activated protein kinase (AMPK)-SNS-BAT axis, have been discovered and emerged as potential drug targets to combat obesity. In this review we provide an overview of the complex central regulation of BAT and how different neuronal cell populations co-ordinately work to maintain energy homeostasis.
Collapse
Affiliation(s)
- Cristina Contreras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria , Santiago de Compostela, 15782 , Spain
| | | | | | | | | | | | | |
Collapse
|
27
|
Melanocortin-4 receptor (MC4R) polymorphisms are associated with growth and meat quality traits in sheep. Mol Biol Rep 2014; 41:6967-74. [DOI: 10.1007/s11033-014-3583-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 07/05/2014] [Indexed: 12/21/2022]
|
28
|
Girardet C, Begriche K, Ptitsyn A, Koza RA, Butler AA. Unravelling the mysterious roles of melanocortin-3 receptors in metabolic homeostasis and obesity using mouse genetics. INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2014; 4:S37-44. [PMID: 27152165 DOI: 10.1038/ijosup.2014.10] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The central nervous melanocortin system maintains body mass and adiposity within a 'healthy' range by regulating satiety and metabolic homeostasis. Neural melanocortin-4 receptors (MC4R) modulate satiety signals and regulate autonomic outputs governing glucose and lipid metabolism in the periphery. The functions of melanocortin-3 receptors (MC3R) have been less well defined. We have observed that food anticipatory activity (FAA) is attenuated in Mc3r-/- mice housed in light:dark or constant dark conditions. Mc3r-/- mice subjected to the restricted feeding protocol that was used to induce FAA also developed insulin resistance, dyslipidaemia, impaired glucose tolerance and evidence of a cellular stress response in the liver. MC3Rs may thus function as modulators of oscillator systems that govern circadian rhythms, integrating signals from nutrient sensors to facilitate synchronizing peak foraging behaviour and metabolic efficiency with nutrient availability. To dissect the functions of MC3Rs expressed in hypothalamic and extra-hypothalamic structures, we inserted a 'lox-stop-lox' (TB) sequence into the Mc3r gene. Mc3r (TB/TB) mice recapitulate the phenotype reported for Mc3r-/- mice: increased adiposity, accelerated diet-induced obesity and attenuated FAA. The ventromedial hypothalamus exhibits high levels of Mc3r expression; however, restoring the expression of the LoxTB Mc3r allele in this nucleus did not restore FAA. However, a surprising outcome came from studies using Nestin-Cre to restore the expression of the LoxTB Mc3r allele in the nervous system. These data suggest that 'non-neural' MC3Rs have a role in the defence of body weight. Future studies examining the homeostatic functions of MC3Rs should therefore consider actions outside the central nervous system.
Collapse
Affiliation(s)
- C Girardet
- Department of Metabolism and Aging, The Scripps Research Institute , Jupiter, FL, USA
| | - K Begriche
- Department of Metabolism and Aging, The Scripps Research Institute , Jupiter, FL, USA
| | - A Ptitsyn
- The Pennington Biomedical Research Center, Louisiana State University System , Baton Rouge, LA, USA
| | - R A Koza
- The Pennington Biomedical Research Center, Louisiana State University System , Baton Rouge, LA, USA
| | - A A Butler
- Department of Metabolism and Aging, The Scripps Research Institute , Jupiter, FL, USA
| |
Collapse
|
29
|
Zegers D, Beckers S, Hendrickx R, Van Camp JK, Van Hoorenbeeck K, Desager KN, Massa G, Van Gaal LF, Van Hul W. Prevalence of rare MC3R variants in obese cases and lean controls. Endocrine 2013; 44:386-90. [PMID: 23264184 DOI: 10.1007/s12020-012-9862-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 12/12/2012] [Indexed: 01/13/2023]
Abstract
The role of mutations in the melanocortin-3 receptor (MC3R) gene, which is implicated in the regulation of energy homeostasis, is still under debate. Animal studies have clearly proven that, together with the melanocortin-4 receptor (MC4R), the MC3R is a critical receptor for melanocortin peptides within the leptin-melanocortin signaling cascade. However, as several mutations have been found in lean individuals and not all mutations seem to cause receptor dysfunction, results from mutation screens in obese humans remain controversial. In the present study, we screened for rare variants in the MC3R gene of obese children and lean controls to assess the prevalence of MC3R mutations in the Belgian population. We screened 249 severely overweight and obese children and adolescents and 239 lean adults for mutations in the coding region of MC3R. Mutation screening was performed by high resolution melting curve analysis and direct sequencing. We identified four non-synonymous coding variations in the obese population, all of which had been reported previously. In addition, we also found four novel rare MC3R variants in the lean control population, suggesting that not all MC3R mutations are disease-causing. Overall, the total prevalence of rare MC3R variants was 1 % in Belgian obese children and adolescents compared to 1.02 % in lean controls. Ultimately, cosegregation studies combined with comprehensive functional analysis is required to determine the potential pathogenic role of rare MC3R variants in causing human obesity.
Collapse
Affiliation(s)
- Doreen Zegers
- Department of Medical Genetics, University of Antwerp, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Carter S, Caron A, Richard D, Picard F. Role of leptin resistance in the development of obesity in older patients. Clin Interv Aging 2013; 8:829-44. [PMID: 23869170 PMCID: PMC3706252 DOI: 10.2147/cia.s36367] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Obesity is a global epidemic associated with aging-like cellular processes; in both aging and obesity, resistance to hormones such as insulin and leptin can be observed. Leptin is a circulating hormone/cytokine with central and peripheral effects that is released mainly by subcutaneous white adipose tissue. Centrally, leptin controls food intake, energy expenditure, and fat distribution, whereas it controls (among several others) insulin sensitivity, free fatty acids (FFAs) oxidation, and lipolysis in the periphery. Aging is associated with important changes in both the distribution and the composition of adipose tissue. Fat is redistributed from the subcutaneous to the visceral depot and increased inflammation participates in adipocyte dysfunction. This redistribution of adipose tissue in favor of visceral fat influences negatively both longevity and healthy aging as shown in numerous animal models. These modifications observed during aging are also associated with leptin resistance. This resistance blunts normal central and peripheral functions of leptin, which leads to a decrease in neuroendocrine function and insulin sensitivity, an imbalance in energy regulation, and disturbances in lipid metabolism. Here, we review how age-related leptin resistance triggers metabolic disturbances and affects the longevity of obese patients. Furthermore, we discuss the potential impacts of leptin resistance on the decline of brown adipose tissue thermogenesis observed in elderly individuals.
Collapse
Affiliation(s)
- Sophie Carter
- Faculty of Pharmacy, Department of Anatomy and Physiology, Université Laval, Québec, QC, Canada
| | | | | | | |
Collapse
|
31
|
Girardet C, Butler AA. Neural melanocortin receptors in obesity and related metabolic disorders. Biochim Biophys Acta Mol Basis Dis 2013; 1842:482-94. [PMID: 23680515 DOI: 10.1016/j.bbadis.2013.05.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 04/16/2013] [Accepted: 05/03/2013] [Indexed: 12/11/2022]
Abstract
Obesity is a global health issue, as it is associated with increased risk of developing chronic conditions associated with disorders of metabolism such as type 2 diabetes and cardiovascular disease. A better understanding of how excessive fat accumulation develops and causes diseases of the metabolic syndrome is urgently needed. The hypothalamic melanocortin system is an important point of convergence connecting signals of metabolic status with the neural circuitry that governs appetite and the autonomic and neuroendocrine system controling metabolism. This system has a critical role in the defense of body weight and maintenance of homeostasis. Two neural melanocortin receptors, melanocortin 3 and 4 receptors (MC3R and MC4R), play crucial roles in the regulation of energy balance. Mutations in the MC4R gene are the most common cause of monogenic obesity in humans, and a large literature indicates a role in regulating both energy intake through the control of satiety and energy expenditure. In contrast, MC3Rs have a more subtle role in energy homeostasis. Results from our lab indicate an important role for MC3Rs in synchronizing rhythms in foraging behavior with caloric cues and maintaining metabolic homeostasis during periods of nutrient scarcity. However, while deletion of the Mc3r gene in mice alters nutrient partitioning to favor accumulation of fat mass no obvious role for MC3R haploinsufficiency in human obesity has been reported. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.
Collapse
MESH Headings
- Animals
- Body Weight/genetics
- Cardiovascular Diseases/complications
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/pathology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Humans
- Metabolic Diseases/genetics
- Metabolic Diseases/metabolism
- Metabolic Diseases/pathology
- Mice
- Obesity/complications
- Obesity/genetics
- Obesity/metabolism
- Obesity/pathology
- Receptor, Melanocortin, Type 3/genetics
- Receptor, Melanocortin, Type 3/metabolism
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/metabolism
Collapse
Affiliation(s)
- Clemence Girardet
- Department of Metabolism and Aging, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Andrew A Butler
- Department of Metabolism and Aging, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA.
| |
Collapse
|
32
|
Asano H, Yamada T, Hashimoto O, Umemoto T, Sato R, Ohwatari S, Kanamori Y, Terachi T, Funaba M, Matsui T. Diet-induced changes in Ucp1 expression in bovine adipose tissues. Gen Comp Endocrinol 2013; 184:87-92. [PMID: 23370305 DOI: 10.1016/j.ygcen.2013.01.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 01/04/2013] [Accepted: 01/11/2013] [Indexed: 01/04/2023]
Abstract
Brown adipocytes, which regulate non-shivering thermogenesis, have been believed to exist in a limited number of mammalian species, and only under limited physiological conditions. Recent discoveries indicate that adult humans possess a significant number of functional brown adipocytes. This study explores the regulatory emergence of brown adipocytes in white adipose tissue (WAT) depots of fattening cattle. RT-PCR analyses indicated significant expression of Ucp1, a brown adipocyte-specific gene, in the WAT of 31-month-old Japanese Black steers. Immunohistochemical analysis revealed that Ucp1-positive small adipocytes were dispersed in the subcutaneous WAT. Next, we examined expression level of Ucp1 and other brown adipocyte-selective genes such as Pgc1α, Cidea, Dio2, Cox1, Cox7a1 and Cox8b in WAT of 30-month-old steers fed either diet with low protein/energy content (roughage diet) or that with high protein/energy content (concentrate diet) for 20months. Ucp1 expression in the subcutaneous WAT was significantly higher in the concentrate diet group than in the roughage diet group. Furthermore, the higher Ucp1 expression levels were limited to the subcutaneous WAT, and no differences between groups were detected in the mesenteric, perirenal, intermuscular or intramuscular WAT. Expression of Dio2, Cox1 and Cox8b was higher in the subcutaneous WAT but not in the mesenteric WAT of the concentrate diet group. Furthermore, expression of Prdm16, a positive regulator of differentiation toward brown adipocyte-lineage cells, and expression of leptin, a molecule that enhances activity of brown adipocytes, were significantly higher in the subcutaneous WAT of the concentrate diet group. This study demonstrates the presence of brown adipocytes in WAT depots of fattening cattle, and suggests the diet-related modulation of expression of genes predominantly expressed in brown adipocytes.
Collapse
Affiliation(s)
- Hiroki Asano
- Division of Applied Biosciences, Kyoto University Graduate School of Agriculture, Kyoto 606-8502, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Begriche K, Girardet C, McDonald P, Butler AA. Melanocortin-3 receptors and metabolic homeostasis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 114:109-46. [PMID: 23317784 DOI: 10.1016/b978-0-12-386933-3.00004-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Attenuated activity of the central nervous melanocortin system causes obesity and insulin resistance. Obese rodents treated with melanocortins exhibit improvements in obesity and metabolic homeostasis that are not mutually dependent, suggesting metabolic actions that are independent of weight changes. These responses are generally thought to involve G-protein-coupled receptors expressed in the brain. Melanocortin-4 receptors (MC4Rs) regulate satiety and autonomic nervous system and thyroid function. MC3Rs are expressed in hypothalamic and limbic regions involved in controlling ingestive behaviors and autonomic function. Mc3r-/- mice exhibit increased adiposity and an accelerated diet-induced obesity. While this phenotype is not dependent on hyperphagia, data on the regulation of food intake by MC3Rs are inconsistent. Recent investigations by our laboratory suggest a unique combination of behavioral and metabolic disorders in Mc3r-/- mice. MC3Rs are critical for the expression of the anticipatory response and metabolic homeostasis when food intake occurs outside the normal voluntary rhythms driven by photoperiod. Using a Cre-Lox strategy, we can now investigate MC3Rs expressed in different brain regions and organ systems in the periphery. While focusing on the functions of neural MC3Rs, early results suggest an additional layer of complexity with central and peripheral MC3Rs involved in the defense of body weight.
Collapse
Affiliation(s)
- Karima Begriche
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, Florida, USA
| | | | | | | |
Collapse
|
34
|
Yang F, Tao YX. Functional characterization of nine novel naturally occurring human melanocortin-3 receptor mutations. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1752-61. [PMID: 22884546 DOI: 10.1016/j.bbadis.2012.07.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Revised: 07/12/2012] [Accepted: 07/27/2012] [Indexed: 12/20/2022]
Abstract
The melanocortin-3 receptor (MC3R) is a member of family A rhodopsin-like G protein-coupled receptors. Mouse genetic studies suggested that MC3R and the related MC4R are non-redundant regulators of energy homeostasis. Lack of Mc3r leads to higher feed efficiency and fat mass. However, until now only a few MC3R mutations have been identified in humans and the role of MC3R in the pathogenesis of obesity was unclear. In the present study, we performed detailed functional studies on nine naturally occurring MC3R mutations recently reported. We found that all nine mutants had decreased cell surface expression. A260V, M275T, and L297V had decreased total expression whereas the other six mutants had normal total expression. Mutants S69C and T280S exhibited significant defects in ligand binding and signaling. The dramatic defects of T280S might be partially caused by decreased cell surface expression. In addition, we found mutants M134I and M275T had decreased maximal binding but displayed similar signaling properties as wild-type MC3R. All the other mutants had normal binding and signaling activities. Co-expression studies showed that all mutants except L297V did not affect wild-type MC3R signaling. Multiple mutations at T280 demonstrated the necessity of Thr for cell surface expression, ligand binding, and signaling. In summary, we provided detailed data of these novel human MC3R mutations leading to a better understanding of structure-function relationship of MC3R and the role of MC3R mutation in obesity.
Collapse
Affiliation(s)
- Fan Yang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | | |
Collapse
|
35
|
Henagan TM, Forney L, Dietrich MA, Harrell BR, Stewart LK. Melanocortin receptor expression is associated with reduced CRP in response to resistance training. J Appl Physiol (1985) 2012; 113:393-400. [PMID: 22678961 PMCID: PMC4422369 DOI: 10.1152/japplphysiol.00107.2012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 06/03/2012] [Indexed: 12/18/2022] Open
Abstract
The existing paradigm of exercise-induced decreases in chronic inflammation focuses on the expression of inflammatory receptors on systemic monocytes in response to exercise training, with the role of anti-inflammatory receptors largely ignored. Our recent preliminary studies indicate that the anti-inflammatory melanocortin receptors (MCRs) may play a role in modulating exercise-induced decreases in chronic inflammation. Here, we present a study designed to determine the effect of intense, resistance exercise training on systemic monocyte MCR expression. Because low-grade chronic inflammation is associated with elevated cardiometabolic risk in healthy populations and exercise decreases chronic inflammation, we investigated the associations between systemic monocyte cell surface expression of MCRs and inflammatory markers as a possible mechanism for the beneficial anti-inflammatory effects of resistance training. To this end, the present study includes 40 adults (aged 19-27 yr) and implements a 12-wk periodized, intensive resistance training intervention. Melanocortin 1 and 3 receptor expression on systemic monocytes and inflammatory markers, including C-reactive protein (CRP), interleukin (IL)-6, IL-1β, and IL-10, were measured before and after the intervention. Resistance training significantly altered MCR systemic monocyte cell surface expression, had no chronic effects on IL-6, IL-1β, or IL-10 expression, but significantly decreased CRP levels from a moderate to a low cardiovascular disease risk category. More specifically, decreased melanocortin 3 receptor expression significantly correlated with decreased CRP, independent of changes in adiposity. These data suggest that the observed responses in MCR expression and decreases in cardiovascular disease risk in response to resistance training represent an important anti-inflammatory mechanism in regulating exercise-induced decreases in chronic inflammation that occur independent of chronic changes in systemic cytokines.
Collapse
Affiliation(s)
- Tara M Henagan
- Neurosignaling Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | | | | | | | | |
Collapse
|
36
|
Abstract
The melanocortin-3 receptor-deficient (MC3-R(-/-)) mouse exhibits mild obesity without hyperphagia or hypometabolism. MC3-R deletion is reported to increase adiposity, reduce lean mass and white adipose tissue inflammation, and increase sensitivity to salt-induced hypertension. We show here that the MC3-R(-/-) mouse exhibits defective fasting-induced white adipose tissue lipolysis, fasting-induced liver triglyceride accumulation, fasting-induced refeeding, and fasting-induced regulation of the adipostatic and hypothalamic-adrenal-pituitary axes. Close examination of the hypothalamic-pituitary-adrenal axis showed that MC3-R(-/-) mice exhibit elevated nadir corticosterone as well as a blunted fasting-induced activation of the axis. The previously described phenotypes of this animal and the reduced bone density reported here parallel those of Cushing syndrome. Thus, MC3-R is required for communicating nutritional status to both central and peripheral tissues involved in nutrient partitioning, and this defect explains much of the metabolic phenotype in the model.
Collapse
|
37
|
Richard D, Monge-Roffarello B, Chechi K, Labbé SM, Turcotte EE. Control and physiological determinants of sympathetically mediated brown adipose tissue thermogenesis. Front Endocrinol (Lausanne) 2012; 3:36. [PMID: 22654862 PMCID: PMC3356074 DOI: 10.3389/fendo.2012.00036] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 02/13/2012] [Indexed: 02/05/2023] Open
Abstract
Brown adipose tissue (BAT) represents a remarkable heat-producing tissue. The thermogenic potential of BAT is conferred by uncoupling protein 1, a protein found uniquely in brown adipocytes. BAT activity and capacity is controlled by the sympathetic nervous system (SNS), which densely innervates brown fat depots. SNS-mediated BAT thermogenesis is essentially governed by hypothalamic and brainstem neurons. BAT activity is also modulated by brain energy balance pathways including the very significant brain melanocortin system, suggesting a genuine involvement of SNS-mediated BAT thermogenesis in energy homeostasis. The use of positron emission tomography/computed tomography scanning has revealed the presence of well-defined BAT depots in the cervical, clavicular, and paraspinal areas in adult humans. The prevalence of these depots is higher in subjects exposed to low temperature and is also higher in women compared to men. Moreover, the prevalence of BAT decreases with age and body fat mass, suggesting that BAT could be involved in energy balance regulation and obesity in humans. This short review summarizes recent progress made in our understanding of the control of SNS-mediated BAT thermogenesis and of the determinants of BAT prevalence or detection in humans.
Collapse
Affiliation(s)
- Denis Richard
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec et Groupe interdisciplinaire de Recherche sur l’Obésité de l’Université LavalQuébec, QC, Canada
- *Correspondence: Denis Richard, Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, 2725 Chemin Sainte-Foy, Québec, QC, Canada G1V 4G5. e-mail:
| | - Boris Monge-Roffarello
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec et Groupe interdisciplinaire de Recherche sur l’Obésité de l’Université LavalQuébec, QC, Canada
| | - Kanta Chechi
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec et Groupe interdisciplinaire de Recherche sur l’Obésité de l’Université LavalQuébec, QC, Canada
| | - Sébastien M. Labbé
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec et Groupe interdisciplinaire de Recherche sur l’Obésité de l’Université LavalQuébec, QC, Canada
| | - Eric E. Turcotte
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec et Groupe interdisciplinaire de Recherche sur l’Obésité de l’Université LavalQuébec, QC, Canada
| |
Collapse
|
38
|
Bechtold DA, Sidibe A, Saer BRC, Li J, Hand LE, Ivanova EA, Darras VM, Dam J, Jockers R, Luckman SM, Loudon ASI. A role for the melatonin-related receptor GPR50 in leptin signaling, adaptive thermogenesis, and torpor. Curr Biol 2011; 22:70-7. [PMID: 22197240 DOI: 10.1016/j.cub.2011.11.043] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 11/03/2011] [Accepted: 11/21/2011] [Indexed: 12/21/2022]
Abstract
The ability of mammals to maintain a constant body temperature has proven to be a profound evolutionary advantage, allowing members of this class to thrive in most environments on earth. Intriguingly, some mammals employ bouts of deep hypothermia (torpor) to cope with reduced food supply and harsh climates [1, 2]. During torpor, physiological processes such as respiration, cardiac function, and metabolic rate are severely depressed, yet the neural mechanisms that regulate torpor remain unclear [3]. Hypothalamic responses to energy signals, such as leptin, influence the expression of torpor [4-7]. We show that the orphan receptor GPR50 plays an important role in adaptive thermogenesis and torpor. Unlike wild-type mice, Gpr50(-/-) mice readily enter torpor in response to fasting and 2-deoxyglucose administration. Decreased thermogenesis in Gpr50(-/-) mice is not due to a deficit in brown adipose tissue, the principal site of nonshivering thermogenesis in mice [8]. GPR50 is highly expressed in the hypothalamus of several species, including man [9, 10]. In line with this, altered thermoregulation in Gpr50(-/-) mice is associated with attenuated responses to leptin and a suppression of thyrotropin-releasing hormone. Thus, our findings identify hypothalamic circuits involved in torpor and reveal GPR50 to be a novel component of adaptive thermogenesis in mammals.
Collapse
|
39
|
Begriche K, Levasseur PR, Zhang J, Rossi J, Skorupa D, Solt LA, Young B, Burris TP, Marks DL, Mynatt RL, Butler AA. Genetic dissection of the functions of the melanocortin-3 receptor, a seven-transmembrane G-protein-coupled receptor, suggests roles for central and peripheral receptors in energy homeostasis. J Biol Chem 2011; 286:40771-81. [PMID: 21984834 PMCID: PMC3220494 DOI: 10.1074/jbc.m111.278374] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 09/05/2011] [Indexed: 01/29/2023] Open
Abstract
The melanocortin-3 receptor (MC3R) gene is pleiotropic, influencing body composition, natriuresis, immune function, and entrainment of circadian rhythms to nutrient intake. MC3Rs are expressed in hypothalamic and limbic regions of the brain and in peripheral tissues. To investigate the roles of central MC3Rs, we inserted a "lox-stop-lox" (LoxTB) 5' of the translation initiation codon of the mouse Mc3r gene and reactivated transcription using neuron-specific Cre transgenic mice. As predicted based on earlier observations of Mc3r knock-out mice, Mc3r(TB/TB) mice displayed reduced lean mass, increased fat mass, and accelerated diet-induced obesity. Surprisingly, rescuing Mc3r expression in the nervous system using the Nestin-Cre transgene only partially rescued obesity in chow-fed conditions and had no impact on the accelerated diet-induced obesity phenotype. The ventromedial hypothalamus (VMH), a critical node in the neural networks regulating feeding-related behaviors and metabolic homeostasis, exhibits dense Mc3r expression relative to other brain regions. To target VMH MC3R expression, we used the steroidogenic factor-1 Cre transgenic mouse. Although restoring VMH MC3R signaling also had a modest impact on obesity, marked improvements in metabolic homeostasis were observed. VMH MC3R signaling was not sufficient to rescue the lean mass phenotype or the regulation of behaviors anticipating food anticipation. These results suggest that actions of MC3Rs impacting on energy homeostasis involve both central and peripheral sites of action. The impact of central MC3Rs on behavior and metabolism involves divergent pathways; VMH MC3R signaling improves metabolic homeostasis but does not significantly impact on the expression of behaviors anticipating nutrient availability.
Collapse
Affiliation(s)
| | - Peter R. Levasseur
- the Department of Pediatrics, Oregon Health and Science University, Portland, Oregon 97239, and
| | - Jingying Zhang
- the Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808
| | - Jari Rossi
- From the Departments of Metabolism and Aging and
| | | | | | - Brandon Young
- the Genomics Core, The Scripps Research Institute, Jupiter, Florida 33458
| | | | - Daniel L. Marks
- the Department of Pediatrics, Oregon Health and Science University, Portland, Oregon 97239, and
| | - Randall L. Mynatt
- the Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808
| | | |
Collapse
|
40
|
Leptin action in the dorsomedial hypothalamus increases sympathetic tone to brown adipose tissue in spite of systemic leptin resistance. J Neurosci 2011; 31:12189-97. [PMID: 21865462 DOI: 10.1523/jneurosci.2336-11.2011] [Citation(s) in RCA: 231] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Leptin regulates body weight in mice by decreasing appetite and increasing sympathetic nerve activity (SNA), which increases energy expenditure in interscapular brown adipose tissue (iBAT). Diet-induced obese mice (DIO) are resistant to the anorectic actions of leptin. We evaluated whether leptin still stimulated sympathetic outflow in DIO mice. We measured iBAT temperature as a marker of SNA. We found that obese hyperleptinemic mice have higher iBAT temperature than mice on regular diet. Conversely, obese leptin-deficient ob/ob mice have lower iBAT temperature. Additionally, leptin increased SNA in obese (DIO and ob/ob) and control mice, despite DIO mice being resistant to anorectic action of leptin. We demonstrated that neurons in the dorsomedial hypothalamus (DMH) of DIO mice mediate the thermogenic responses to hyperleptinemia in obese mammals because blockade of leptin receptors in the DMH prevented the thermogenic effects of leptin. Peripheral Melotan II (MTII) injection increased iBAT temperature, but it was blunted by blockade of DMH melanocortin receptors (MC4Rs) by injecting agouti-related peptide (AgRP) directly into the DMH, suggesting a physiological role of the DMH on temperature regulation in animals with normal body weight. Nevertheless, obese mice without a functional melanocortin system (MC4R KO mice) have an increased sympathetic outflow to iBAT compared with their littermates, suggesting that higher leptin levels drive sympathoexcitation to iBAT by a melanocortin-independent pathway. Because the sympathetic nervous system contributes in regulating blood pressure, heart rate, and hepatic glucose production, selective leptin resistance may be a crucial mechanism linking adiposity and metabolic syndrome.
Collapse
|
41
|
Yang J, Bromage TG, Zhao Q, Xu BH, Gao WL, Tian HF, Tang HJ, Liu DW, Zhao XQ. Functional evolution of leptin of Ochotona curzoniae in adaptive thermogenesis driven by cold environmental stress. PLoS One 2011; 6:e19833. [PMID: 21698227 PMCID: PMC3116822 DOI: 10.1371/journal.pone.0019833] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Accepted: 04/18/2011] [Indexed: 12/20/2022] Open
Abstract
Background Environmental stress can accelerate the directional selection and evolutionary rate of specific stress-response proteins to bring about new or altered functions, enhancing an organism's fitness to challenging environments. Plateau pika (Ochotona curzoniae), an endemic and keystone species on Qinghai-Tibetan Plateau, is a high hypoxia and low temperature tolerant mammal with high resting metabolic rate and non-shivering thermogenesis to cope in this harsh plateau environment. Leptin is a key hormone related to how these animals regulate energy homeostasis. Previous molecular evolutionary analysis helped to generate the hypothesis that adaptive evolution of plateau pika leptin may be driven by cold stress. Methodology/Principal Findings To test the hypothesis, recombinant pika leptin was first purified. The thermogenic characteristics of C57BL/6J mice injected with pika leptin under warm (23±1°C) and cold (5±1°C) acclimation is investigated. Expression levels of genes regulating adaptive thermogenesis in brown adipose tissue and the hypothalamus are compared between pika leptin and human leptin treatment, suggesting that pika leptin has adaptively and functionally evolved. Our results show that pika leptin regulates energy homeostasis via reduced food intake and increased energy expenditure under both warm and cold conditions. Compared with human leptin, pika leptin demonstrates a superior induced capacity for adaptive thermogenesis, which is reflected in a more enhanced β-oxidation, mitochondrial biogenesis and heat production. Moreover, leptin treatment combined with cold stimulation has a significant synergistic effect on adaptive thermogenesis, more so than is observed with a single cold exposure or single leptin treatment. Conclusions/Significance These findings support the hypothesis that cold stress has driven the functional evolution of plateau pika leptin as an ecological adaptation to the Qinghai-Tibetan Plateau.
Collapse
Affiliation(s)
- Jie Yang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Timothy G. Bromage
- Department of Biomaterials and Biomimetics, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York, United States of America
| | - Qian Zhao
- Graduate School of the Chinese Academy of Sciences, The Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Bao Hong Xu
- Microbiology Department, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang, Hebei, People's Republic of China
| | - Wei Li Gao
- Microbiology Department, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang, Hebei, People's Republic of China
| | - Hui Fang Tian
- Microbiology Department, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang, Hebei, People's Republic of China
| | - Hui Jun Tang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Dian Wu Liu
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
- * E-mail: (DWL); (XQZ)
| | - Xin Quan Zhao
- Key Laboratory of Qinghai-Tibetan Plateau Biological Evolution and Adaptation, Northwest Plateau Institute of Biology, The Chinese Academy of Sciences, Xining, Qinghai, People's Republic of China
- * E-mail: (DWL); (XQZ)
| |
Collapse
|
42
|
Begriche K, Sutton GM, Butler AA. Homeostastic and non-homeostatic functions of melanocortin-3 receptors in the control of energy balance and metabolism. Physiol Behav 2011; 104:546-54. [PMID: 21497617 DOI: 10.1016/j.physbeh.2011.04.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 04/04/2011] [Accepted: 04/08/2011] [Indexed: 01/05/2023]
Abstract
The central nervous melanocortin system is a neural network linking nutrient-sensing systems with hypothalamic, limbic and hindbrain neurons regulating behavior and metabolic homeostasis. Primary melanocortin neurons releasing melanocortin receptor ligands residing in the hypothalamic arcuate nucleus are regulated by nutrient-sensing and metabolic signals. A smaller group of primary neurons releasing melanocortin agonists in the nucleus tractus solitarius in the brainstem are also regulated by signals of metabolic state. Two melanocortin receptors regulate energy homeostasis. Melanocortin-4 receptors regulate satiety and autonomic outputs controlling peripheral metabolism. The functions of melanocortin-3 receptors (MC3R) expressed in hypothalamic and limbic structures are less clear. Here we discuss published data and preliminary observations from our laboratory suggesting that neural MC3R regulate inputs into systems governing the synchronization of rhythms in behavior and metabolism with nutrient intake. Mice subjected to a restricted feeding protocol, where a limited number of calories are presented at a 24h interval, rapidly exhibit bouts of increased wakefulness and activity which anticipate food presentation. The full expression of these responses is dependent on MC3R. Moreover, MC3R knockout mice are unique in exhibiting a dissociation of weight loss from improved glucose homeostasis when subject to a restricted feeding protocol. While mice lacking MC3R fed ad libitum exhibit normal to moderate hyperinsulinemia, when subjected to a restricted protocol they develop hyperglycemia, glucose intolerance, and dyslipidemia. Collectively, our data suggest that the central nervous melanocortin system is a point convergence in the control of energy balance and the expression of rhythms anticipating nutrient intake.
Collapse
Affiliation(s)
- Karima Begriche
- Department of Metabolism and Aging, The Scripps Research Institute-Florida, Jupiter, FL 33458, USA
| | | | | |
Collapse
|
43
|
Henagan TM, Phillips MD, Cheek DJ, Kirk KM, Barbee JJ, Stewart LK. The melanocortin 3 receptor: a novel mediator of exercise-induced inflammation reduction in postmenopausal women? J Aging Res 2011; 2011:512593. [PMID: 21253483 PMCID: PMC3022199 DOI: 10.4061/2011/512593] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 11/09/2010] [Accepted: 12/14/2010] [Indexed: 12/30/2022] Open
Abstract
The purpose of this study was to determine whether resistance exercise training-induced reductions in inflammation are mediated via melanocortin 3 receptor expression in obese (BMI 32.7 ± 3.7) women (65.6 ± 2.8 yrs) randomized to either a control (N = 11) or resistance training group (N = 12). The resistance trained group performed resistance training 3 days/week for 12 weeks. Resting blood samples were collected before and after the training intervention in both resistance trained and control groups. Resistance training upregulated melanocortin 3 receptor mRNA by 16-fold (P = .035) and decreased monocyte count, without changing leukocyte number, body composition, or body weight. Resistance trained individuals exhibited increased sensitivity to inflammatory stimuli, whereas control individuals exhibited no change. While there was no change in whole blood tumor necrosis factor alpha mRNA between the groups, whole blood interleukin 10 mRNA was higher in the resistance trained group following the intervention period. In summary, it appears that resistance training may modulate melanocortin 3 receptor expression, providing a possible mechanism for the anti-inflammatory effects of exercise training.
Collapse
Affiliation(s)
- Tara M Henagan
- Department of Kinesiology, Louisiana State University, 112 Long Fieldhouse, Baton Rouge, LA 70803, USA
| | | | | | | | | | | |
Collapse
|
44
|
Zegers D, Beckers S, de Freitas F, Peeters AV, Mertens IL, Verhulst SL, Rooman RP, Timmermans JP, Desager KN, Massa G, Van Gaal LF, Van Hul W. Identification of three novel genetic variants in the melanocortin-3 receptor of obese children. Obesity (Silver Spring) 2011; 19:152-9. [PMID: 20539302 DOI: 10.1038/oby.2010.127] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The melanocortin-3 receptor (MC3R), a G-protein-coupled receptor expressed in the hypothalamus, is a key component of the leptin-melanocortin pathway that regulates energy homeostasis. It is suggested that an MC3R defect leads to an increased feed efficiency, by which nutrients are partitioned preferentially into fat. In this study, we hypothesized that early-onset obesity could be induced by mutations in MC3R. To investigate this hypothesis, we screened the entire coding region of the MC3R gene for mutations in obese subjects. A total of 404 overweight and obese children and adolescents, 86 severely obese adults (BMI ≥40 kg/m²), and 150 normal-weight control adults were included. Besides three synonymous coding variations in the MC3R gene (S69S, L95L, I226I), we were able to identify three novel heterozygous, nonsynonymous, coding mutations (N128S, V211I, L299V) in three unrelated obese children. None of these mutations were found in any of the control subjects. Functional studies assessing localization and signaling properties of the mutant receptors provided proof for impaired function of the L299V mutated receptor, whereas no conclusive evidence for functional impairment of the N128S and V211I mutated receptors could be established. First, these results provide supporting evidence for a role of the MC3R gene in the pathogenesis of obesity in a small subset of patients. Second, they show that caution is called for the interpretation of newly discovered mutations in MC3R.
Collapse
Affiliation(s)
- Doreen Zegers
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
|
46
|
White PB, True EM, Ziegler KM, Wang SS, Swartz-Basile DA, Pitt HA, Zyromski NJ. Insulin, leptin, and tumoral adipocytes promote murine pancreatic cancer growth. J Gastrointest Surg 2010; 14:1888-93; discussion 1893-4. [PMID: 20859700 DOI: 10.1007/s11605-010-1349-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Accepted: 08/23/2010] [Indexed: 01/31/2023]
Abstract
BACKGROUND Obesity accelerates development and growth of human pancreatic cancer. We recently reported similar findings in a novel murine model of pancreatic cancer in congenitally obese mice. The current experiments were designed to evaluate the effects of diet-induced obesity on pancreatic cancer growth. METHODS Thirty C57BL/6J female mice were fed either control 10% fat (n = 10) or 60% fat diet (n = 20) starting at age 6 weeks. At 11 weeks, 2.5 × 10(5) PAN02 murine pancreatic cancer cells were inoculated. After 6 weeks, tumors were harvested. Serum adiponectin, leptin, insulin, and glucose concentrations were measured. Tumor proliferation, apoptosis, adipocyte content, and tumor-infiltrating lymphocytes were evaluated. RESULTS The diet-induced obesity diet led to significant weight gain (control 21.3 ± 0.6 g; diet-induced obesity 23.1 ± 0.5 g; p = 0.03). Mice heavier than 23.1 g were considered "Overweight." Tumors grew significantly larger in overweight (1.3 ± 0.3 g) compared to lean (0.5 ± 0.2 g; p = 0.03) mice; tumor size correlated positively with body weight (R = 0.56; p < 0.02). Serum leptin (3.1 ± 0.7 vs. 1.4 ± 0.2 ng/ml) and insulin (0.5 ± 0.2 vs. 0.18 ± 0.02 ng/ml) were significantly greater in overweight mice. Tumor proliferation, apoptosis, and tumor adipocyte volume were similar. T and B lymphocytes were observed infiltrating tumors from lean and overweight mice in similar number. CONCLUSION These data show that diet-induced obesity accelerates the growth of murine pancreatic cancer.
Collapse
Affiliation(s)
- Patrick B White
- Department of Surgery, Indiana University School of Medicine, 535 Barnhill Dr. RT 130, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
Plaisance EP, Henagan TM, Echlin H, Boudreau A, Hill KL, Lenard NR, Hasek BE, Orentreich N, Gettys TW. Role of beta-adrenergic receptors in the hyperphagic and hypermetabolic responses to dietary methionine restriction. Am J Physiol Regul Integr Comp Physiol 2010; 299:R740-50. [PMID: 20554934 DOI: 10.1152/ajpregu.00838.2009] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Dietary methionine restriction (MR) limits fat deposition and decreases plasma leptin, while increasing food consumption, total energy expenditure (EE), plasma adiponectin, and expression of uncoupling protein 1 (UCP1) in brown and white adipose tissue (BAT and WAT). beta-adrenergic receptors (beta-AR) serve as conduits for sympathetic input to adipose tissue, but their role in mediating the effects of MR on energy homeostasis is unclear. Energy intake, weight, and adiposity were modestly higher in beta(3)-AR(-/-) mice on the Control diet compared with wild-type (WT) mice, but the hyperphagic response to the MR diet and the reduction in fat deposition did not differ between the genotypes. The absence of beta(3)-ARs also did not diminish the ability of MR to increase total EE and plasma adiponectin or decrease leptin mRNA, but it did block the MR-dependent increase in UCP1 mRNA in BAT but not WAT. In a further study, propranolol was used to antagonize remaining beta-adrenergic input (beta(1)- and beta(2)-ARs) in beta(3)-AR(-/-) mice, and this treatment blocked >50% of the MR-induced increase in total EE and UCP1 induction in both BAT and WAT. We conclude that signaling through beta-adrenergic receptors is a component of the mechanism used by dietary MR to increase EE, and that beta(1)- and beta(2)-ARs are able to substitute for beta(3)-ARs in mediating the effect of dietary MR on EE. These findings are consistent with the involvement of both UCP1-dependent and -independent mechanisms in the physiological responses affecting energy balance that are produced by dietary MR.
Collapse
Affiliation(s)
- Eric P Plaisance
- Laboratory of Nutrient Sensing and Adipocyte Signaling, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Hasek BE, Stewart LK, Henagan TM, Boudreau A, Lenard NR, Black C, Shin J, Huypens P, Malloy VL, Plaisance EP, Krajcik RA, Orentreich N, Gettys TW. Dietary methionine restriction enhances metabolic flexibility and increases uncoupled respiration in both fed and fasted states. Am J Physiol Regul Integr Comp Physiol 2010; 299:R728-39. [PMID: 20538896 DOI: 10.1152/ajpregu.00837.2009] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Dietary methionine restriction (MR) is a mimetic of chronic dietary restriction (DR) in the sense that MR increases rodent longevity, but without food restriction. We report here that MR also persistently increases total energy expenditure (EE) and limits fat deposition despite increasing weight-specific food consumption. In Fischer 344 (F344) rats consuming control or MR diets for 3, 9, and 20 mo, mean EE was 1.5-fold higher in MR vs. control rats, primarily due to higher EE during the night at all ages. The day-to-night transition produced a twofold higher heat increment of feeding (3.0 degrees C vs. 1.5 degrees C) in MR vs. controls and an exaggerated increase in respiratory quotient (RQ) to values greater than 1, indicative of the interconversion of glucose to lipid by de novo lipogenesis. The simultaneous inhibition of glucose utilization and shift to fat oxidation during the day was also more complete in MR (RQ approximately 0.75) vs. controls (RQ approximately 0.85). Dietary MR produced a rapid and persistent increase in uncoupling protein 1 expression in brown (BAT) and white adipose tissue (WAT) in conjunction with decreased leptin and increased adiponectin levels in serum, suggesting that remodeling of the metabolic and endocrine function of adipose tissue may have an important role in the overall increase in EE. We conclude that the hyperphagic response to dietary MR is matched to a coordinated increase in uncoupled respiration, suggesting the engagement of a nutrient-sensing mechanism, which compensates for limited methionine through integrated effects on energy homeostasis.
Collapse
Affiliation(s)
- Barbara E Hasek
- Laboratory of Nutrient Sensing and Adipocyte Signaling, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Gautron L, Lee C, Funahashi H, Friedman J, Lee S, Elmquist J. Melanocortin-4 receptor expression in a vago-vagal circuitry involved in postprandial functions. J Comp Neurol 2010; 518:6-24. [PMID: 19882715 DOI: 10.1002/cne.22221] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Vagal afferents regulate energy balance by providing a link between the brain and postprandial signals originating from the gut. In the current study, we investigated melanocortin-4 receptor (MC4R) expression in the nodose ganglion, where the cell bodies of vagal sensory afferents reside. By using a line of mice expressing green fluorescent protein (GFP) under the control of the MC4R promoter, we found GFP expression in approximately one-third of nodose ganglion neurons. By using immunohistochemistry combined with in situ hybridization, we also demonstrated that approximately 20% of GFP-positive neurons coexpressed cholecystokinin receptor A. In addition, we found that the GFP is transported to peripheral tissues by both vagal sensory afferents and motor efferents, which allowed us to assess the sites innervated by MC4R-GFP neurons. GFP-positive efferents that co-expressed choline acetyltransferase specifically terminated in the hepatic artery and the myenteric plexus of the stomach and duodenum. In contrast, GFP-positive afferents that did not express cholinergic or sympathetic markers terminated in the submucosal plexus and mucosa of the duodenum. Retrograde tracing experiments confirmed the innervation of the duodenum by GFP-positive neurons located in the nodose ganglion. Our findings support the hypothesis that MC4R signaling in vagal afferents may modulate the activity of fibers sensitive to satiety signals such as cholecystokinin, and that MC4R signaling in vagal efferents may contribute to the control of the liver and gastrointestinal tract.
Collapse
Affiliation(s)
- Laurent Gautron
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-9077, USA
| | | | | | | | | | | |
Collapse
|
50
|
Savastano DM, Tanofsky-Kraff M, Han JC, Ning C, Sorg RA, Roza CA, Wolkoff LE, Anandalingam K, Jefferson-George KS, Figueroa RE, Sanford EL, Brady S, Kozlosky M, Schoeller DA, Yanovski JA. Energy intake and energy expenditure among children with polymorphisms of the melanocortin-3 receptor. Am J Clin Nutr 2009; 90:912-20. [PMID: 19656839 PMCID: PMC2744620 DOI: 10.3945/ajcn.2009.27537] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Homozygosity for 2 protein-altering polymorphisms in the melanocortin-3 receptor gene (MC3R) coding sequence, C17A and G241A, has been reported to be associated with an obesity phenotype in children, yet how these polymorphisms affect energy homeostasis is unknown. Association between adult body weight and +2138InsCAGACC, another variant in the 3' untranslated region of MC3R, has also been described. OBJECTIVE The objective of this study was to examine associations of C17A + G241A and +2138InsCAGACC MC3R variants with children's energy balance. DESIGN Children aged 6-19 y were genotyped for MC3R C17A, G241A, and +2138InsCAGACC. Subjects underwent studies of energy intake from a 9835-kcal food array (n = 185), resting energy expenditure (REE) by using indirect calorimetry (n = 302), or total daily energy expenditure (TEE) by using doubly labeled water (n = 120). Linear regression was used to examine the associations between MC3R polymorphisms and the measures of energy balance. RESULTS Body mass index and fat mass were greater in those with double homozygosity for C17A + G241A (P = 0.001). After accounting for covariates (including body composition), the number of minor C17A + G241A alleles was associated with significantly greater energy intake (beta = +0.15, P = 0.02) but not altered REE or TEE. No significant associations were observed between +2138InsCAGACC and measures of either fat mass or energy balance. CONCLUSIONS C17A + G241A polymorphisms may be associated with pediatric obesity because of greater energy intake rather than because of diminished energy expenditure. +2138InsCAGACC does not appear to be associated with obesity or measures of energy balance in children.
Collapse
Affiliation(s)
- David M Savastano
- Unit on Growth and Obesity, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1103, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|