1
|
Inozemtseva LS, Yatsenko KA, Glazova NY, Kamensky AA, Myasoedov NF, Levitskaya NG, Grivennikov IA, Dolotov OV. Antidepressant-like and antistress effects of the ACTH(4-10) synthetic analogs Semax and Melanotan II on male rats in a model of chronic unpredictable stress. Eur J Pharmacol 2024; 984:177068. [PMID: 39442746 DOI: 10.1016/j.ejphar.2024.177068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 10/19/2024] [Accepted: 10/20/2024] [Indexed: 10/25/2024]
Abstract
Current antidepressant therapy shows substantial limitations, and there is an urgent need for the development of new treatment strategies for depression. Stressful events and hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis play an important role in the pathogenesis of depression. HPA axis activity is self-regulated by negative feedback at several levels including adrenocorticotropic hormone (ACTH)-mediated feedback. Here, we investigated whether noncorticotropic synthetic analogs of the ACTH(4-10) fragment, ACTH(4-7)-Pro-Gly-Pro (Semax) and Ac-Nle4-cyclo[Asp5-His6-D-Phe7-Arg8-Trp9-Lys10]ACTH(4-10)-NH2 (Melanotan II (MTII), a potent agonist of melanocortin receptors), have potential antidepressant activity in a chronic unpredictable stress (CUS) rat model of depression. Stressed and control male adult Sprague-Dawley rats received daily intraperitoneal injections of saline or a low dose (60 nmol/kg of body weight (BW)) of Semax or MTII. Rats were monitored for BW and hedonic status, as measured in the sucrose preference test. We found that chronic treatment with Semax and MTII reversed or substantially attenuated CUS-induced anhedonia, BW gain suppression, adrenal hypertrophy and a decrease in the hippocampal levels of BDNF. In the forced swim test, no effects of the CUS procedure or peptides on the duration of rat immobility were detected. Our findings show that in the CUS paradigm, systemically administered ACTH(4-10) analogs Semax and MTII exert antidepressant-like effects on anhedonia and hippocampal BDNF levels, and attenuate markers of chronic stress load, at least in male rats. The results support the argument that ACTH(4-10) analogs and other noncorticotropic melanocortins may have promising therapeutic potential for the treatment and prevention of depression and other stress-related pathologies.
Collapse
Affiliation(s)
| | | | - Natalya Yu Glazova
- National Research Center "Kurchatov Institute", Moscow, Russia; Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Andrey A Kamensky
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | - Natalia G Levitskaya
- National Research Center "Kurchatov Institute", Moscow, Russia; Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | - Oleg V Dolotov
- National Research Center "Kurchatov Institute", Moscow, Russia; Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia.
| |
Collapse
|
2
|
Franzago M, Borrelli P, Di Nicola M, Cavallo P, D’Adamo E, Di Tizio L, Gazzolo D, Stuppia L, Vitacolonna E. From Mother to Child: Epigenetic Signatures of Hyperglycemia and Obesity during Pregnancy. Nutrients 2024; 16:3502. [PMID: 39458497 PMCID: PMC11510513 DOI: 10.3390/nu16203502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND In utero exposure to maternal hyperglycemia and obesity can trigger detrimental effects in the newborn through epigenetic programming. We aimed to assess the DNA methylation levels in the promoters of MC4R and LPL genes from maternal blood, placenta, and buccal swab samples collected in children born to mothers with and without obesity and Gestational Diabetes Mellitus (GDM). METHODS A total of 101 Caucasian mother-infant pairs were included in this study. Sociodemographic characteristics, clinical parameters, physical activity, and adherence to the Mediterranean diet were evaluated in the third trimester of pregnancy. Clinical parameters of the newborns were recorded at birth. RESULTS A negative relationship between MC4R DNA methylation on the fetal side of the GDM placenta and birth weight (r = -0.630, p = 0.011) of newborns was found. MC4R DNA methylation level was lower in newborns of GDM women (CpG1: 2.8% ± 3.0%, CpG2: 3.8% ± 3.3%) as compared to those of mothers without GDM (CpG1: 6.9% ± 6.2%, CpG2: 6.8% ± 5.6%; p < 0.001 and p = 0.0033, respectively), and it was negatively correlated with weight (r = -0.229; p = 0.035), head circumference (r = -0.236; p = 0.030), and length (r = -0.240; p = 0.027) at birth. LPL DNA methylation was higher on the fetal side of the placenta in obese patients as compared to normal-weight patients (66.0% ± 14.4% vs. 55.7% ± 15.2%, p = 0.037), and it was associated with maternal total cholesterol (r = 0.770, p = 0.015) and LDL-c (r = 0.783, p = 0.012). CONCLUSIONS These results support the role of maternal MC4R and LPL methylation in fetal programming and in the future metabolic health of children.
Collapse
Affiliation(s)
- Marica Franzago
- Department of Medicine and Aging, School of Medicine, and Health Sciences, “G. D’Annunzio” University, Via dei Vestini, Chieti-Pescara, 66100 Chieti, Italy; (M.F.); (D.G.)
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy;
| | - Paola Borrelli
- Laboratory of Biostatistics, Department of Medical, Oral and Biotechnological Sciences, “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy; (P.B.); (M.D.N.)
| | - Marta Di Nicola
- Laboratory of Biostatistics, Department of Medical, Oral and Biotechnological Sciences, “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy; (P.B.); (M.D.N.)
| | - Pierluigi Cavallo
- Department of Medicine and Aging, School of Medicine, and Health Sciences, “G. D’Annunzio” University, Via dei Vestini, Chieti-Pescara, 66100 Chieti, Italy; (M.F.); (D.G.)
| | - Ebe D’Adamo
- Neonatal Intensive Care Unit, “G. D’Annunzio” University, 66100 Chieti, Italy;
| | - Luciano Di Tizio
- Department of Obstetrics and Gynaecology, SS. Annunziata Hospital, “G. D’Annunzio” University, 66100 Chieti, Italy;
| | - Diego Gazzolo
- Department of Medicine and Aging, School of Medicine, and Health Sciences, “G. D’Annunzio” University, Via dei Vestini, Chieti-Pescara, 66100 Chieti, Italy; (M.F.); (D.G.)
- Neonatal Intensive Care Unit, “G. D’Annunzio” University, 66100 Chieti, Italy;
| | - Liborio Stuppia
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy;
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy
| | - Ester Vitacolonna
- Department of Medicine and Aging, School of Medicine, and Health Sciences, “G. D’Annunzio” University, Via dei Vestini, Chieti-Pescara, 66100 Chieti, Italy; (M.F.); (D.G.)
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy;
| |
Collapse
|
3
|
Steiner AM, Roscoe RF, Booze RM, Mactutus CF. Motivational dysregulation with melanocortin 4 receptor haploinsufficiency. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2024; 3:237-250. [PMID: 39741559 PMCID: PMC11683877 DOI: 10.1515/nipt-2024-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 10/18/2024] [Indexed: 01/03/2025]
Abstract
Obesity, by any standard, is a global health crisis. Both genetic and dietary contributions to the development and maintenance of obesity were integral factors of our experimental design. As mutations of the melanocortin 4 receptors (MC4R) are the leading monogenetic cause of obesity, MC4R haploinsufficient rats were fed a range of dietary fat (0-12 %) in a longitudinal design. Physiological and motivational assessments were performed using a locomotor task, a 5-choice sucrose preference task, an operant task with fixed and progressive ratios, as well as a distraction operant task. Dendritic spine morphology of medium spiny neurons (MSNs) of the nucleus accumbens (NAc), cells with ample D1 and D2 receptors, was also assessed. The percentage of lipid deposits in the liver of each rat was also analyzed using the Area Fraction Fractionator probe for stereological measurements. MC4R haploinsufficiency resulted in a phenotypic resemblance for adult-onset obesity that was exacerbated by the consumption of a high-fat diet. Results from the operant tasks indicate that motivational deficits due to MC4R haploinsufficiency were apparent prior to the onset of obesity and exacerbated by dietary fat consumption after obesity was well established. Moreover, MSN morphology shifted to longer spines with smaller head diameters for the MC4R+/- animals under the high-fat diet, suggesting a potential mechanism for the dysregulation of motivation to work for food. Increasing our knowledge of the neural circuitry/mechanisms responsible for the rewarding properties of food has significant implications for understanding energy balance and the development of obesity.
Collapse
Affiliation(s)
- Alex M. Steiner
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, University of South Carolina, Columbia, SC, USA
| | - Robert F. Roscoe
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, University of South Carolina, Columbia, SC, USA
| | - Rosemarie M. Booze
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, University of South Carolina, Columbia, SC, USA
| | - Charles F. Mactutus
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
4
|
Yount ST, Wang S, Allen AT, Shapiro LP, Butkovich LM, Gourley SL. A molecularly defined orbitofrontal cortical neuron population controls compulsive-like behavior, but not inflexible choice or habit. Prog Neurobiol 2024; 238:102632. [PMID: 38821345 PMCID: PMC11332912 DOI: 10.1016/j.pneurobio.2024.102632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/11/2024] [Accepted: 05/20/2024] [Indexed: 06/02/2024]
Abstract
Habits are familiar behaviors triggered by cues, not outcome predictability, and are insensitive to changes in the environment. They are adaptive under many circumstances but can be considered antecedent to compulsions and intrusive thoughts that drive persistent, potentially maladaptive behavior. Whether compulsive-like and habit-like behaviors share neural substrates is still being determined. Here, we investigated mice bred to display inflexible reward-seeking behaviors that are insensitive to action consequences. We found that these mice demonstrate habitual response biases and compulsive-like grooming behavior that was reversible by fluoxetine and ketamine. They also suffer dendritic spine attrition on excitatory neurons in the orbitofrontal cortex (OFC). Nevertheless, synaptic melanocortin 4 receptor (MC4R), a factor implicated in compulsive behavior, is preserved, leading to the hypothesis that Mc4r+ OFC neurons may drive aberrant behaviors. Repeated chemogenetic stimulation of Mc4r+ OFC neurons triggered compulsive and not inflexible or habitual response biases in otherwise typical mice. Thus, Mc4r+ neurons within the OFC appear to drive compulsive-like behavior that is dissociable from habitual behavior. Understanding which neuron populations trigger distinct behaviors may advance efforts to mitigate harmful compulsions.
Collapse
Affiliation(s)
- Sophie T Yount
- Graduate Program in Molecular and Systems Pharmacology, USA; Emory National Primate Research Center, USA; Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, USA
| | - Silu Wang
- Emory National Primate Research Center, USA; Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, USA; Graduate Program in Neuroscience, Emory University, Atlanta, GA, USA
| | - Aylet T Allen
- Emory National Primate Research Center, USA; Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, USA
| | - Lauren P Shapiro
- Graduate Program in Molecular and Systems Pharmacology, USA; Emory National Primate Research Center, USA; Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, USA
| | - Laura M Butkovich
- Emory National Primate Research Center, USA; Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, USA
| | - Shannon L Gourley
- Graduate Program in Molecular and Systems Pharmacology, USA; Emory National Primate Research Center, USA; Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, USA; Graduate Program in Neuroscience, Emory University, Atlanta, GA, USA; Children's Healthcare of Atlanta, Atlanta, GA, USA.
| |
Collapse
|
5
|
Wu S, Wang J, Zhang Z, Jin X, Xu Y, Si Y, Liang Y, Ge Y, Zhan H, Peng L, Bi W, Luo D, Li M, Meng B, Guan Q, Zhao J, Gao L, He Z. Shank3 deficiency elicits autistic-like behaviors by activating p38α in hypothalamic AgRP neurons. Mol Autism 2024; 15:14. [PMID: 38570876 PMCID: PMC10993499 DOI: 10.1186/s13229-024-00595-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND SH3 and multiple ankyrin repeat domains protein 3 (SHANK3) monogenic mutations or deficiency leads to excessive stereotypic behavior and impaired sociability, which frequently occur in autism cases. To date, the underlying mechanisms by which Shank3 mutation or deletion causes autism and the part of the brain in which Shank3 mutation leads to the autistic phenotypes are understudied. The hypothalamus is associated with stereotypic behavior and sociability. p38α, a mediator of inflammatory responses in the brain, has been postulated as a potential gene for certain cases of autism occurrence. However, it is unclear whether hypothalamus and p38α are involved in the development of autism caused by Shank3 mutations or deficiency. METHODS Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and immunoblotting were used to assess alternated signaling pathways in the hypothalamus of Shank3 knockout (Shank3-/-) mice. Home-Cage real-time monitoring test was performed to record stereotypic behavior and three-chamber test was used to monitor the sociability of mice. Adeno-associated viruses 9 (AAV9) were used to express p38α in the arcuate nucleus (ARC) or agouti-related peptide (AgRP) neurons. D176A and F327S mutations expressed constitutively active p38α. T180A and Y182F mutations expressed inactive p38α. RESULTS We found that Shank3 controls stereotypic behavior and sociability by regulating p38α activity in AgRP neurons. Phosphorylated p38 level in hypothalamus is significantly enhanced in Shank3-/- mice. Consistently, overexpression of p38α in ARC or AgRP neurons elicits excessive stereotypic behavior and impairs sociability in wild-type (WT) mice. Notably, activated p38α in AgRP neurons increases stereotypic behavior and impairs sociability. Conversely, inactivated p38α in AgRP neurons significantly ameliorates autistic behaviors of Shank3-/- mice. In contrast, activated p38α in pro-opiomelanocortin (POMC) neurons does not affect stereotypic behavior and sociability in mice. LIMITATIONS We demonstrated that SHANK3 regulates the phosphorylated p38 level in the hypothalamus and inactivated p38α in AgRP neurons significantly ameliorates autistic behaviors of Shank3-/- mice. However, we did not clarify the biochemical mechanism of SHANK3 inhibiting p38α in AgRP neurons. CONCLUSIONS These results demonstrate that the Shank3 deficiency caused autistic-like behaviors by activating p38α signaling in AgRP neurons, suggesting that p38α signaling in AgRP neurons is a potential therapeutic target for Shank3 mutant-related autism.
Collapse
Affiliation(s)
- Shanshan Wu
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Jing Wang
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Zicheng Zhang
- School of Modern Posts, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu, 210009, China
| | - Xinchen Jin
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yang Xu
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Youwen Si
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences,East China Normal University, Shanghai, 200062, China
| | - Yixiao Liang
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Yueping Ge
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Huidong Zhan
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Li Peng
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Wenkai Bi
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Dandan Luo
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Mengzhu Li
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Bo Meng
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences,East China Normal University, Shanghai, 200062, China
- Department of Pharmacology and Chemical Biology, Department of Neurology, Emory University, Atlanta, GA, 30322, USA
| | - Qingbo Guan
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Zhao He
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration, and Practice Center, Shandong University, Jinan, Shandong, 250021, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.
- Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, 544 Jingsi Road, Jinan, Shandong, 250021, China.
| |
Collapse
|
6
|
Wright SN, Leger BS, Rosenthal SB, Liu SN, Jia T, Chitre AS, Polesskaya O, Holl K, Gao J, Cheng R, Garcia Martinez A, George A, Gileta AF, Han W, Netzley AH, King CP, Lamparelli A, Martin C, St Pierre CL, Wang T, Bimschleger H, Richards J, Ishiwari K, Chen H, Flagel SB, Meyer P, Robinson TE, Solberg Woods LC, Kreisberg JF, Ideker T, Palmer AA. Genome-wide association studies of human and rat BMI converge on synapse, epigenome, and hormone signaling networks. Cell Rep 2023; 42:112873. [PMID: 37527041 PMCID: PMC10546330 DOI: 10.1016/j.celrep.2023.112873] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 07/05/2023] [Accepted: 07/11/2023] [Indexed: 08/03/2023] Open
Abstract
A vexing observation in genome-wide association studies (GWASs) is that parallel analyses in different species may not identify orthologous genes. Here, we demonstrate that cross-species translation of GWASs can be greatly improved by an analysis of co-localization within molecular networks. Using body mass index (BMI) as an example, we show that the genes associated with BMI in humans lack significant agreement with those identified in rats. However, the networks interconnecting these genes show substantial overlap, highlighting common mechanisms including synaptic signaling, epigenetic modification, and hormonal regulation. Genetic perturbations within these networks cause abnormal BMI phenotypes in mice, too, supporting their broad conservation across mammals. Other mechanisms appear species specific, including carbohydrate biosynthesis (humans) and glycerolipid metabolism (rodents). Finally, network co-localization also identifies cross-species convergence for height/body length. This study advances a general paradigm for determining whether and how phenotypes measured in model species recapitulate human biology.
Collapse
Affiliation(s)
- Sarah N Wright
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Program in Bioinformatics and Systems Biology, University of California San Diego, La Jolla, CA 92093, USA
| | - Brittany S Leger
- Department of Psychiatry, University of California San Diego, La Jolla, CA 93093, USA; Program in Biomedical Sciences, University of California San Diego, La Jolla, CA 93093, USA
| | - Sara Brin Rosenthal
- Center for Computational Biology & Bioinformatics, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sophie N Liu
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Tongqiu Jia
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Apurva S Chitre
- Department of Psychiatry, University of California San Diego, La Jolla, CA 93093, USA
| | - Oksana Polesskaya
- Department of Psychiatry, University of California San Diego, La Jolla, CA 93093, USA
| | - Katie Holl
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jianjun Gao
- Department of Psychiatry, University of California San Diego, La Jolla, CA 93093, USA
| | - Riyan Cheng
- Department of Psychiatry, University of California San Diego, La Jolla, CA 93093, USA
| | - Angel Garcia Martinez
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Anthony George
- Clinical and Research Institute on Addictions, University at Buffalo, Buffalo, NY 14203, USA
| | - Alexander F Gileta
- Department of Psychiatry, University of California San Diego, La Jolla, CA 93093, USA; Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Wenyan Han
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Alesa H Netzley
- Department of Psychiatry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Christopher P King
- Clinical and Research Institute on Addictions, University at Buffalo, Buffalo, NY 14203, USA; Department of Psychology, University at Buffalo, Buffalo, NY 14260, USA
| | | | - Connor Martin
- Clinical and Research Institute on Addictions, University at Buffalo, Buffalo, NY 14203, USA; Department of Psychology, University at Buffalo, Buffalo, NY 14260, USA
| | | | - Tengfei Wang
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Hannah Bimschleger
- Department of Psychiatry, University of California San Diego, La Jolla, CA 93093, USA
| | - Jerry Richards
- Clinical and Research Institute on Addictions, University at Buffalo, Buffalo, NY 14203, USA
| | - Keita Ishiwari
- Clinical and Research Institute on Addictions, University at Buffalo, Buffalo, NY 14203, USA; Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14203, USA
| | - Hao Chen
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Shelly B Flagel
- Department of Psychiatry, University of Michigan, Ann Arbor, MI 48109, USA; Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Paul Meyer
- Department of Psychology, University at Buffalo, Buffalo, NY 14260, USA
| | - Terry E Robinson
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Leah C Solberg Woods
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Jason F Kreisberg
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Trey Ideker
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA 93093, USA; Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
7
|
Shemery AM, Zendlo M, Kowalski J, Gorrell E, Everett S, Wagner JG, Davis AE, Koch LG, Britton SL, Mul JD, Novak CM. Reduced contextually induced muscle thermogenesis in rats with calorie restriction and lower aerobic fitness but not monogenic obesity. Temperature (Austin) 2023; 10:379-393. [PMID: 37554387 PMCID: PMC10405760 DOI: 10.1080/23328940.2023.2171669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/29/2023] Open
Abstract
We have previously identified predator odor as a potent stimulus activating thermogenesis in skeletal muscle in rats. As this may prove relevant for energy balance and weight loss, the current study investigated whether skeletal muscle thermogenesis was altered with negative energy balance, obesity propensity seen in association with low intrinsic aerobic fitness, and monogenic obesity. First, weight loss subsequent to 3 wk of 50% calorie restriction suppressed the muscle thermogenic response to predator odor. Next, we compared rats bred based on artificial selection for intrinsic aerobic fitness - high- and low-capacity runners (HCR, LCR) - that display robust leanness and obesity propensity, respectively. Aerobically fit HCR showed enhanced predator odor-induced muscle thermogenesis relative to the less-fit LCR. This contrasted with the profound monogenic obesity displayed by rats homozygous for a loss of function mutation in Melanocortin 4 receptor (Mc4rK3a,4X/K314X rats), which showed no discernable deficit in thermogenesis. Taken together, these data imply that body size or obesity per se are not associated with deficient muscle thermogenesis. Rather, the physiological phenotype associated with polygenic obesity propensity may encompass pleiotropic mechanisms in the thermogenic pathway. Adaptive thermogenesis associated with weight loss also likely alters muscle thermogenic mechanisms.
Collapse
Affiliation(s)
| | - Meredith Zendlo
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Jesse Kowalski
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Erin Gorrell
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Scott Everett
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Jacob G. Wagner
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Ashley E. Davis
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Lauren G. Koch
- Department of Physiology and Pharmacology, the University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Steven L. Britton
- Department of Anesthesiology, and Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Joram D. Mul
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Colleen M. Novak
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| |
Collapse
|
8
|
Micioni Di Bonaventura E, Botticelli L, Del Bello F, Giorgioni G, Piergentili A, Quaglia W, Romano A, Gaetani S, Micioni Di Bonaventura MV, Cifani C. Investigating the role of the central melanocortin system in stress and stress-related disorders. Pharmacol Res 2022; 185:106521. [DOI: 10.1016/j.phrs.2022.106521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 11/30/2022]
|
9
|
Thurston L, Hunjan T, Mills EG, Wall MB, Ertl N, Phylactou M, Muzi B, Patel B, Alexander EC, Suladze S, Modi M, Eng PC, Bassett PA, Abbara A, Goldmeier D, Comninos AN, Dhillo WS. Melanocortin 4 receptor agonism enhances sexual brain processing in women with hypoactive sexual desire disorder. J Clin Invest 2022; 132:152341. [PMID: 36189794 PMCID: PMC9525110 DOI: 10.1172/jci152341] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Hypoactive sexual desire disorder (HSDD) is characterized by a persistent deficiency of sexual fantasies and desire for sexual activity, causing marked distress and interpersonal difficulty. It is the most prevalent female sexual health problem globally, affecting approximately 10% of women, but has limited treatment options. Melanocortin 4 receptor (MC4R) agonists have emerged as a promising therapy for women with HSDD, through unknown mechanisms. Studying the pathways involved is crucial for our understanding of normal and abnormal sexual behavior. METHODS Using psychometric, functional neuroimaging, and hormonal analyses, we conducted a randomized, double-blinded, placebo-controlled, crossover clinical study to assess the effects of MC4R agonism compared with placebo on sexual brain processing in 31 premenopausal heterosexual women with HSDD. RESULTS MC4R agonism significantly increased sexual desire for up to 24 hours after administration compared with placebo. During functional neuroimaging, MC4R agonism enhanced cerebellar and supplementary motor area activity and deactivated the secondary somatosensory cortex, specifically in response to visual erotic stimuli, compared with placebo. In addition, MC4R agonism enhanced functional connectivity between the amygdala and the insula during visual erotic stimuli compared with placebo. CONCLUSION These data suggest that MC4R agonism enhanced sexual brain processing by reducing self-consciousness, increasing sexual imagery, and sensitizing women with HSDD to erotic stimuli. These findings provide mechanistic insight into the action of MC4R agonism in sexual behavior and are relevant to the ongoing development of HSDD therapies and MC4R agonist development more widely. TRIAL REGISTRATION ClinicalTrials.gov NCT04179734. FUNDING This is an investigator-sponsored study funded by AMAG Pharmaceuticals Inc., the Medical Research Council (MRC) (MR/T006242/1), and the National Institute for Health Research (NIHR) (CS-2018-18-ST2-002 and RP-2014-05-001).
Collapse
Affiliation(s)
- Layla Thurston
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Tia Hunjan
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Edouard G Mills
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Matthew B Wall
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom.,Invicro, a Konica Minolta Company, London, United Kingdom
| | - Natalie Ertl
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom.,Invicro, a Konica Minolta Company, London, United Kingdom
| | - Maria Phylactou
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Beatrice Muzi
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Bijal Patel
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Emma C Alexander
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Sofiya Suladze
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Manish Modi
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Pei C Eng
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | | | - Ali Abbara
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - David Goldmeier
- Jane Wadsworth Sexual Function Clinic, St. Mary's Hospital and
| | - Alexander N Comninos
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom.,Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom.,Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
10
|
Genes Predisposing to Erectile Dysfunction and Management: A Review. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2022. [DOI: 10.2478/sjecr-2021-0080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Background: The burden of erectile dysfunction (ED) is rising worldwide due to unresponsiveness of some affected individuals to existing drugs and treatment strategies. Fortunately, improvement in biological techniques has led to the understanding that some cases of the disorder may have a genetic etiology, which, when fully understood, may lead to improved treatment.
Objective: This review articulated established ED candidate genes and pathophysiology to assist researchers and medical practitioners to formulate effective drugs and treatment procedures.
Methods: The Google search engine was used to retrieve relevant information on the topic from reputable academic databases, including PubMed, Medline, Google Scholar, Scopus, and SpringerLink.
Results: The search discovered 10 ED candidate genes, which are SIM1, SLC6A4, 5-HTTLPR, TGFB1, DAT1, MC4R, NOS3, GNB3, AR, and MTHFR. Polymorphisms or mutations in these genes may disrupt erectile activities of the hypothalamus, neurotransmitters such as dopamine, serotonin, and nitric oxide as well as relaxation of penile tissues. Clinical presentations of ED include loss of erection, weak vaginal penetration, premature ejaculation, and anejaculation. Each gene has a distinct mechanism, which, if targeted in the affected may reverse the disorder or reduce the effects.
Conclusion: Some cases of ED are genetic, which, when fully understood, may give an insight into new treatment procedures or improve on the current ones. Medical practitioners are advised to formulate treatment procedures that target the affected gene (s) in individuals.
Collapse
|
11
|
Matsumura S, Miyakita M, Miyamori H, Kyo S, Shima D, Yokokawa T, Ishikawa F, Sasaki T, Jinno T, Tanaka J, Goto T, Momma K, Ishihara K, Berdeaux R, Inoue K. Stimulation of G s signaling in MC4R cells by DREADD increases energy expenditure, suppresses food intake, and increases locomotor activity in mice. Am J Physiol Endocrinol Metab 2022; 322:E436-E445. [PMID: 35344393 DOI: 10.1152/ajpendo.00439.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The melanocortin 4 receptor (MC4R) plays an important role in the regulation of appetite and energy expenditure in humans and rodents. Impairment of MC4R signaling causes severe obesity. MC4R mainly couples to the G-protein Gs. Ligand binding to MC4R activates adenylyl cyclase resulting in increased intracellular cAMP levels. cAMP acts as a secondary messenger, regulating various cellular processes. MC4R can also couple with Gq and other signaling pathways. Therefore, the contribution of MC4R/Gs signaling to energy metabolism and appetite remains unclear. To study the effect of Gs signaling activation in MC4R cells on whole body energy metabolism and appetite, we generated a novel mouse strain that expresses a Gs-coupled designer receptors exclusively activated by designer drugs [Gs-DREADD (GsD)] selectively in MC4R-expressing cells (GsD-MC4R mice). Chemogenetic activation of the GsD by a designer drug [deschloroclozapine (DCZ); 0.01∼0.1 mg/kg body wt] in MC4R-expressing cells significantly increased oxygen consumption and locomotor activity. In addition, GsD activation significantly reduced the respiratory exchange ratio, promoting fatty acid oxidation, but did not affect core (rectal) temperature. A low dose of DCZ (0.01 mg/kg body wt) did not suppress food intake, but a high dose of DCZ (0.1 mg/kg body wt) suppressed food intake in MC4R-GsD mice, although either DCZ dose (0.01 or 0.1 mg/kg body wt) did not affect food intake in the control mice. In conclusion, the current study demonstrated that the stimulation of Gs signaling in MC4R-expressing cells increases energy expenditure and locomotor activity and suppresses appetite.NEW & NOTEWORTHY We report that Gs signaling in melanocortin 4 receptor (MC4R)-expressing cells regulates energy expenditure, appetite, and locomotor activity. These findings shed light on the mechanism underlying the regulation of energy metabolism and locomotor activity by MC4R/cAMP signaling.
Collapse
Affiliation(s)
- Shigenobu Matsumura
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
- Department of Clinical Nutrition, Graduate School of Comprehensive Rehabilitation, Osaka Prefecture University, Osaka, Japan
| | - Motoki Miyakita
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Haruka Miyamori
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Satomi Kyo
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| | - Daisuke Shima
- Department of Food Sciences and Human Nutrition, Faculty of Agriculture, Ryukoku University, Shiga, Japan
| | - Takumi Yokokawa
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Fuka Ishikawa
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Tsutomu Sasaki
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tomoki Jinno
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Jin Tanaka
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Goto
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Keiko Momma
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| | - Kengo Ishihara
- Department of Food Sciences and Human Nutrition, Faculty of Agriculture, Ryukoku University, Shiga, Japan
| | - Rebecca Berdeaux
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Kazuo Inoue
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| |
Collapse
|
12
|
Ren J, Wu NN, Wang S, Sowers JR, Zhang Y. Obesity cardiomyopathy: evidence, mechanisms, and therapeutic implications. Physiol Rev 2021; 101:1745-1807. [PMID: 33949876 PMCID: PMC8422427 DOI: 10.1152/physrev.00030.2020] [Citation(s) in RCA: 207] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The prevalence of heart failure is on the rise and imposes a major health threat, in part, due to the rapidly increased prevalence of overweight and obesity. To this point, epidemiological, clinical, and experimental evidence supports the existence of a unique disease entity termed “obesity cardiomyopathy,” which develops independent of hypertension, coronary heart disease, and other heart diseases. Our contemporary review evaluates the evidence for this pathological condition, examines putative responsible mechanisms, and discusses therapeutic options for this disorder. Clinical findings have consolidated the presence of left ventricular dysfunction in obesity. Experimental investigations have uncovered pathophysiological changes in myocardial structure and function in genetically predisposed and diet-induced obesity. Indeed, contemporary evidence consolidates a wide array of cellular and molecular mechanisms underlying the etiology of obesity cardiomyopathy including adipose tissue dysfunction, systemic inflammation, metabolic disturbances (insulin resistance, abnormal glucose transport, spillover of free fatty acids, lipotoxicity, and amino acid derangement), altered intracellular especially mitochondrial Ca2+ homeostasis, oxidative stress, autophagy/mitophagy defect, myocardial fibrosis, dampened coronary flow reserve, coronary microvascular disease (microangiopathy), and endothelial impairment. Given the important role of obesity in the increased risk of heart failure, especially that with preserved systolic function and the recent rises in COVID-19-associated cardiovascular mortality, this review should provide compelling evidence for the presence of obesity cardiomyopathy, independent of various comorbid conditions, underlying mechanisms, and offer new insights into potential therapeutic approaches (pharmacological and lifestyle modification) for the clinical management of obesity cardiomyopathy.
Collapse
Affiliation(s)
- Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China.,Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Ne N Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| | - Shuyi Wang
- School of Medicine, Shanghai University, Shanghai, China.,University of Wyoming College of Health Sciences, Laramie, Wyoming
| | - James R Sowers
- Dalton Cardiovascular Research Center, Diabetes and Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Yingmei Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| |
Collapse
|
13
|
Cellular and Molecular Players in the Interplay between Adipose Tissue and Breast Cancer. Int J Mol Sci 2021; 22:ijms22031359. [PMID: 33572982 PMCID: PMC7866411 DOI: 10.3390/ijms22031359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
The incidence and severity of obesity are rising in most of the world. In addition to metabolic disorders, obesity is associated with an increase in the incidence and severity of a variety of types of cancer, including breast cancer (BC). The bidirectional interaction between BC and adipose cells has been deeply investigated, although the molecular and cellular players involved in these mechanisms are far from being fully elucidated. Here, we review the current knowledge on these interactions and describe how preclinical research might be used to clarify the effects of obesity over BC progression and morbidity, with particular attention paid to promising therapeutic interventions.
Collapse
|
14
|
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: 5.4] [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
|
15
|
Szpirer C. Rat models of human diseases and related phenotypes: a systematic inventory of the causative genes. J Biomed Sci 2020; 27:84. [PMID: 32741357 PMCID: PMC7395987 DOI: 10.1186/s12929-020-00673-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/09/2020] [Indexed: 12/13/2022] Open
Abstract
The laboratory rat has been used for a long time as the model of choice in several biomedical disciplines. Numerous inbred strains have been isolated, displaying a wide range of phenotypes and providing many models of human traits and diseases. Rat genome mapping and genomics was considerably developed in the last decades. The availability of these resources has stimulated numerous studies aimed at discovering causal disease genes by positional identification. Numerous rat genes have now been identified that underlie monogenic or complex diseases and remarkably, these results have been translated to the human in a significant proportion of cases, leading to the identification of novel human disease susceptibility genes, helping in studying the mechanisms underlying the pathological abnormalities and also suggesting new therapeutic approaches. In addition, reverse genetic tools have been developed. Several genome-editing methods were introduced to generate targeted mutations in genes the function of which could be clarified in this manner [generally these are knockout mutations]. Furthermore, even when the human gene causing a disease had been identified without resorting to a rat model, mutated rat strains (in particular KO strains) were created to analyze the gene function and the disease pathogenesis. Today, over 350 rat genes have been identified as underlying diseases or playing a key role in critical biological processes that are altered in diseases, thereby providing a rich resource of disease models. This article is an update of the progress made in this research and provides the reader with an inventory of these disease genes, a significant number of which have similar effects in rat and humans.
Collapse
Affiliation(s)
- Claude Szpirer
- Université Libre de Bruxelles, B-6041, Gosselies, Belgium.
- , Waterloo, Belgium.
| |
Collapse
|
16
|
Long-term effects of pro-opiomelanocortin methylation induced in food-restricted dams on metabolic phenotypes in male rat offspring. Obstet Gynecol Sci 2020; 63:239-250. [PMID: 32489968 PMCID: PMC7231940 DOI: 10.5468/ogs.2020.63.3.239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 09/09/2019] [Accepted: 09/24/2019] [Indexed: 11/08/2022] Open
Abstract
Objective Maternal malnutrition affects the growth and metabolic health of the offspring. Little is known about the long-term effect on metabolic indices of epigenetic changes in the brain caused by maternal diet. Thus, we explored the effect of maternal food restriction during pregnancy on metabolic profiles of the offspring, by evaluating the DNA methylation of hypothalamic appetite regulators at 3 weeks of age. Methods Sprague-Dawley rats were divided into 2 groups: a control group and a group with a 50% food-restricted (FR) diet during pregnancy. Methylation and expression of appetite regulator genes were measured in 3-week-old offspring using pyrosequencing, real-time polymerase chain reaction, and western blotting analyses. We analyzed the relationship between DNA methylation and metabolic profiles by Pearson's correlation analysis. Results The expression of pro-opiomelanocortin (POMC) decreased, whereas DNA methylation significantly increased in male offspring of the FR dams, compared to the male offspring of control dams. Hypermethylation of POMC was positively correlated with the levels of high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol in 3-week-old male offspring. In addition, there were significant positive correlations between hypermethylation of POMC and the levels of triglycerides, HDL-C, and leptin in 6-month-old male offspring. Conclusion Our findings suggest that maternal food restriction during pregnancy influences the expression of hypothalamic appetite regulators via epigenetic changes, leading to the development of metabolic disorders in the offspring.
Collapse
|
17
|
Goudarzi M, Nahavandi A, Mehrabi S, Eslami M, Shahbazi A, Barati M. Valproic acid administration exerts protective effects against stress-related anhedonia in rats. J Chem Neuroanat 2020; 105:101768. [DOI: 10.1016/j.jchemneu.2020.101768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/12/2020] [Accepted: 02/12/2020] [Indexed: 12/22/2022]
|
18
|
Suleiman JB, Mohamed M, Bakar ABA. A systematic review on different models of inducing obesity in animals: Advantages and limitations. J Adv Vet Anim Res 2020; 7:103-114. [PMID: 32219116 PMCID: PMC7096124 DOI: 10.5455/javar.2020.g399] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/20/2022] Open
Abstract
Several animals have been in the limelight of basic research associated with metabolic diseases like obesity. Obesity can be considered as a significant public health concern in the world. It raises the chances for a variety of disease conditions that includes diabetes, hypertension, liver disease, and cancers, which, in turn, decreases the overall lifespan of adult men and women. The World Health Organization has considered obesity as a global epidemic. Researchers have made several attempts to classify human obesity, but none have been successful. Animal obesity can be classified based on their etiology; however, till now, no animal model of obesity can replicate models of the human condition, they have only provided clues into the causes, aftermaths, and preventive remedy to human adiposity. Over the years, there are varieties of animal models used to induce obesity. Some of them include monogenic, polygenic, surgical, seasonal, and other models of obesity. Apart from the advantages of these models, most of them are accompanied by limitations. The primary purpose of this review is, therefore, to highlight the several models with their advantages and limitations. By knowing the benefits and limitations of animal models of obesity, researchers may be at liberty to select the appropriate one for the study of obesity.
Collapse
Affiliation(s)
- Joseph Bagi Suleiman
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Malaysia
| | - Mahaneem Mohamed
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Malaysia
| | - Ainul Bahiyah Abu Bakar
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Malaysia
| |
Collapse
|
19
|
Abstract
Many animal models that are currently used in appetite and obesity research share at least some main features of human obesity and its comorbidities. Hence, even though no animal model replicates all aspects of "common" human obesity, animal models are imperative in studying the control of energy balance and reasons for its imbalance that may eventually lead to overt obesity. The most frequently used animal models are small rodents that may be based on mutations or manipulations of individual or several genes and on the exposure to obesogenic diets or other manipulations that predispose the animals to gaining or maintaining excessive weight. Characteristics include hyperphagia or changes in energy metabolism and at least in some models the frequent comorbidities of obesity, like hyperglycemia, insulin resistance, or diabetes-like syndromes. Some of the most frequently used animal models of obesity research involve animals with monogenic mutations of the leptin pathway which in fact are useful to study specific mechanistic aspects of eating controls, but typically do not recapitulate "common" obesity in the human population. Hence, this review will mention advantages and disadvantages of respective animal models in order to build a basis for the most appropriate use in biomedical research.
Collapse
Affiliation(s)
- Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland.
| |
Collapse
|
20
|
Spradley FT, Palei AC, Anderson CD, Granger JP. Melanocortin-4 Receptor Deficiency Attenuates Placental Ischemia-Induced Hypertension in Pregnant Rats. Hypertension 2019; 73:162-170. [PMID: 30571561 DOI: 10.1161/hypertensionaha.118.12028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Preeclampsia is a pregnancy-specific disorder of new-onset hypertension linked to placental ischemia. While obesity is a major risk factor for preeclampsia, not all obese pregnant women develop pregnancy-induced hypertension or preeclampsia. Previously, we reported that placental ischemia-induced hypertension is dependent upon intact signaling of the sympathetic nervous system. Moreover, in various models of obesity, blockade of MC4R (melanocortin-4 receptor) signaling protects against the development of hypertension via suppression of the sympathetic nervous system. Less is known about this pathway during obese pregnancy. Although blockade of MC4R may lead to increased body weight during pregnancy, we tested the hypothesis that placental ischemia-induced hypertension is attenuated in obese MC4R-deficient pregnant rats. On gestational day 14, MC4R wild-type or heterozygous-deficient (MC4R-def) rats were subjected to chronic placental ischemia via the reduced uterine perfusion pressure procedure or Sham surgery then examined on gestational day 19. In Sham MC4R-def versus Sham wild-type pregnant rats, there was increased body weight, fat mass, and circulating leptin levels but they had similar fetus weights. Reduced uterine perfusion pressure reduced fetus weights in both strains. Reduced uterine perfusion pressure increased blood pressure in wild-type rats but this response was significantly attenuated in MC4R-def rats, although blood pressure was elevated in Sham MC4R-def over Sham wild-type. These data indicate that while obese MC4R-def pregnant rats have higher blood pressure during pregnancy, placental ischemia-induced hypertension is attenuated in obese MC4R-def pregnant rats. Thus, obese women with abnormal MC4R signaling may be less susceptible to the development of placental ischemia-induced hypertension.
Collapse
Affiliation(s)
- Frank T Spradley
- From the Department of Surgery (F.T.S., A.C.P., C.D.A.), The University of Mississippi Medical Center, Jackson.,Department of Physiology and Biophysics (F.T.S., J.P.G.), The University of Mississippi Medical Center, Jackson
| | - Ana C Palei
- From the Department of Surgery (F.T.S., A.C.P., C.D.A.), The University of Mississippi Medical Center, Jackson
| | - Christopher D Anderson
- From the Department of Surgery (F.T.S., A.C.P., C.D.A.), The University of Mississippi Medical Center, Jackson
| | - Joey P Granger
- Department of Physiology and Biophysics (F.T.S., J.P.G.), The University of Mississippi Medical Center, Jackson
| |
Collapse
|
21
|
Chaffin ATB, Fang Y, Larson KR, Mul JD, Ryan KK. Sex-dependent effects of MC4R genotype on HPA axis tone: implications for stress-associated cardiometabolic disease. Stress 2019; 22:571-580. [PMID: 31184537 PMCID: PMC6690797 DOI: 10.1080/10253890.2019.1610742] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/18/2019] [Indexed: 12/15/2022] Open
Abstract
The melanocortin-4 receptor (MC4R) facilitates hypothalamic-pituitary-adrenocortical (HPA) axis responses to acute stress in male rodents and is a well known to regulator of energy balance. Mutations in the MC4R is the most common monogenic cause of obesity in humans and has been associated with sex-specific effects, but whether stress regulation by the MC4R is sex-dependent, and whether the MC4R facilitates HPA responses to chronic stress, is unknown. We hypothesized that MC4R-signaling contributes to HPA axis dysregulation and metabolic pathophysiology following chronic stress exposure. We measured changes in energy balance, HPA axis tone, and vascular remodeling during chronic variable stress (CVS) in male and female rats with MC4R loss-of-function. Rats were placed into three groups (n = 9-18/genotype/sex) and half of each group was subjected to CVS for 30 days or were non-stressed littermate controls. All rats underwent an acute restraint stress challenge on Day 30. Rats were euthanized on Day 31, adrenals collected for weight, and descending aortas fixed for morphological indices of vascular pathophysiology. We observed a marked interaction between Mc4r genotype and sex for basal HPA axis tone and acute stress responsivity. MC4R loss-of-function blunted both endpoints in males but exaggerated them in females. Contrary to our hypothesis, Mc4r genotype had no effect on either HPA axis responses or metabolic responses to chronic stress. Heightened stress reactivity of females with MC4R mutations suggests a possible mechanism for the sex-dependent effects associated with this mutation in humans and highlights how stress may differentially regulate metabolism in males and females. Lay summary The hypothalamic melanocortin system is an important regulator of energy balance and stress responses. Here, we report a sex-difference in the stress reactivity of rats with a mutation in this system. Our findings highlight how stress may regulate metabolism differently in males and females and may provide insight into sex-differences associated with this mutation in humans.
Collapse
Affiliation(s)
- Aki T-B Chaffin
- a Department of Neurobiology, Physiology and Behavior, University of California , Davis , CA , USA
| | - Yanbin Fang
- a Department of Neurobiology, Physiology and Behavior, University of California , Davis , CA , USA
| | - Karlton R Larson
- a Department of Neurobiology, Physiology and Behavior, University of California , Davis , CA , USA
| | - Joram D Mul
- b Amsterdam UMC, University of Amsterdam , The Netherlands
- c Metabolism and Reward Group, Netherlands Institute for Neuroscience , Amsterdam , The Netherlands
| | - Karen K Ryan
- a Department of Neurobiology, Physiology and Behavior, University of California , Davis , CA , USA
| |
Collapse
|
22
|
Duque-Díaz E, Alvarez-Ojeda O, Coveñas R. Enkephalins and ACTH in the mammalian nervous system. VITAMINS AND HORMONES 2019; 111:147-193. [PMID: 31421699 DOI: 10.1016/bs.vh.2019.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The pentapeptides methionine-enkephalin and leucine-enkephalin belong to the opioid family of peptides, and the non-opiate peptide adrenocorticotropin hormone (ACTH) to the melanocortin peptide family. Enkephalins/ACTH are derived from pro-enkephalin, pro-dynorphin or pro-opiomelanocortin precursors and, via opioid and melanocortin receptors, are responsible for many biological activities. Enkephalins exhibit the highest affinity for the δ receptor, followed by the μ and κ receptors, whereas ACTH binds to the five subtypes of melanocortin receptor, and is the only member of the melanocortin family of peptides that binds to the melanocortin-receptor 2 (ACTH receptor). Enkephalins/ACTH and their receptors exhibit a widespread anatomical distribution. Enkephalins are involved in analgesia, angiogenesis, blood pressure, embryonic development, emotional behavior, feeding, hypoxia, limbic system modulation, neuroprotection, peristalsis, and wound repair; as well as in hepatoprotective, motor, neuroendocrine and respiratory mechanisms. ACTH plays a role in acetylcholine release, aggressive behavior, blood pressure, bone maintenance, hyperalgesia, feeding, fever, grooming, learning, lipolysis, memory, nerve injury repair, neuroprotection, sexual behavior, sleep, social behavior, tissue growth and stimulates the synthesis and secretion of glucocorticoids. Enkephalins/ACTH are also involved in many pathologies. Enkephalins are implicated in alcoholism, cancer, colitis, depression, heart failure, Huntington's disease, influenza A virus infection, ischemia, multiple sclerosis, and stress. ACTH plays a role in Addison's disease, alcoholism, cancer, Cushing's disease, dermatitis, encephalitis, epilepsy, Graves' disease, Guillain-Barré syndrome, multiple sclerosis, podocytopathies, and stress. In this review, we provide an updated description of the enkephalinergic and ACTH systems.
Collapse
Affiliation(s)
- Ewing Duque-Díaz
- Universidad de Santander UDES, Laboratory of Neurosciences, School of Medicine, Bucaramanga, Colombia.
| | - Olga Alvarez-Ojeda
- Universidad Industrial de Santander, Department of Pathology, School of Medicine, Bucaramanga, Colombia
| | - Rafael Coveñas
- Laboratory of Neuroanatomy of the Peptidergic Systems, Institute of Neurosciences of Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| |
Collapse
|
23
|
Minakova E, Lang J, Medel-Matus JS, Gould GG, Reynolds A, Shin D, Mazarati A, Sankar R. Melanotan-II reverses autistic features in a maternal immune activation mouse model of autism. PLoS One 2019; 14:e0210389. [PMID: 30629642 PMCID: PMC6328175 DOI: 10.1371/journal.pone.0210389] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/21/2018] [Indexed: 12/19/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by impaired social interactions, difficulty with communication, and repetitive behavior patterns. In humans affected by ASD, there is a male pre-disposition towards the condition with a male to female ratio of 4:1. In part due to the complex etiology of ASD including genetic and environmental interplay, there are currently no available medical therapies to improve the social deficits of ASD. Studies in rodent models and humans have shown promising therapeutic effects of oxytocin in modulating social adaptation. One pharmacological approach to stimulating oxytocinergic activity is the melanocortin receptor 4 agonist Melanotan-II (MT-II). Notably the effects of oxytocin on environmental rodent autism models has not been investigated to date. We used a maternal immune activation (MIA) mouse model of autism to assess the therapeutic potential of MT-II on autism-like features in adult male mice. The male MIA mice exhibited autism-like features including impaired social behavioral metrics, diminished vocal communication, and increased repetitive behaviors. Continuous administration of MT-II to male MIA mice over a seven-day course resulted in rescue of social behavioral metrics. Normal background C57 male mice treated with MT-II showed no significant alteration in social behavioral metrics. Additionally, there was no change in anxiety-like or repetitive behaviors following MT-II treatment of normal C57 mice, though there was significant weight loss following subacute treatment. These data demonstrate MT-II as an effective agent for improving autism-like behavioral deficits in the adult male MIA mouse model of autism.
Collapse
Affiliation(s)
- Elena Minakova
- Department of Pediatrics, Division of Neonatology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Jordan Lang
- Department of Internal Medicine, Huntington Memorial Hospital, Pasadena, California, United States of America
| | - Jesus-Servando Medel-Matus
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Georgianna G. Gould
- University of Texas Health Science Center at San Antonio, Department of Cellular and Integrative Physiology, San Antonio, Texas, United States of America
| | - Ashley Reynolds
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Don Shin
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Andrey Mazarati
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Children's Discovery and Innovation Institute at UCLA, Los Angeles, California, United States of America
| | - Raman Sankar
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Children's Discovery and Innovation Institute at UCLA, Los Angeles, California, United States of America
| |
Collapse
|
24
|
Zhang M, Chen Y, Xu H, Yang L, Yuan F, Li L, Xu Y, Chen Y, Zhang C, Lin G. Melanocortin Receptor 4 Signaling Regulates Vertebrate Limb Regeneration. Dev Cell 2018; 46:397-409.e5. [PMID: 30130530 PMCID: PMC6107305 DOI: 10.1016/j.devcel.2018.07.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/28/2018] [Accepted: 07/21/2018] [Indexed: 11/16/2022]
Abstract
Melanocortin 4 receptor (Mc4r) plays a crucial role in the central control of energy homeostasis, but its role in peripheral organs has not been fully explored. We have investigated the roles of hypothalamus-mediated energy metabolism during Xenopus limb regeneration. We report that hypothalamus injury inhibits Xenopus tadpole limb regeneration. By loss-of-function and gain-of-function studies, we show that Mc4r signaling is required for limb regeneration in regeneration-competent tadpoles and stimulates limb regeneration in later-stage regeneration-defective tadpoles. It regulates limb regeneration through modulating energy homeostasis and ROS production. Even more interestingly, our results demonstrate that Mc4r signaling is regulated by innervation and α-MSH substitutes for the effect of nerves in limb regeneration. Mc4r signaling is also required for mouse digit regeneration. Thus, our findings link vertebrate limb regeneration with Mc4r-mediated energy homeostasis and provide a new avenue for understanding Mc4r signaling in the peripheral organs.
Collapse
Affiliation(s)
- Mengshi Zhang
- Research Center for Translational Medicine, Translational Medical Center for Stem Cell Therapy, and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Youwei Chen
- Research Center for Translational Medicine, Translational Medical Center for Stem Cell Therapy, and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Hanqian Xu
- Research Center for Translational Medicine, Translational Medical Center for Stem Cell Therapy, and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China; Stem Cell Institute, Department of Genetics Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Li Yang
- Research Center for Translational Medicine, Translational Medical Center for Stem Cell Therapy, and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Feng Yuan
- Research Center for Translational Medicine, Translational Medical Center for Stem Cell Therapy, and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Lei Li
- Research Center for Translational Medicine, Translational Medical Center for Stem Cell Therapy, and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Ying Xu
- Research Center for Translational Medicine, Translational Medical Center for Stem Cell Therapy, and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Ying Chen
- Stem Cell Institute, Department of Genetics Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Chao Zhang
- Research Center for Translational Medicine, Translational Medical Center for Stem Cell Therapy, and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China.
| | - Gufa Lin
- Research Center for Translational Medicine, Translational Medical Center for Stem Cell Therapy, and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China; Stem Cell Institute, Department of Genetics Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
25
|
Moore BS, Mirshahi T. Genetic variants help define the role of the MC4R C-terminus in signaling and cell surface stability. Sci Rep 2018; 8:10397. [PMID: 29991773 PMCID: PMC6039487 DOI: 10.1038/s41598-018-28758-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 06/26/2018] [Indexed: 11/09/2022] Open
Abstract
Screening 92,445 subjects in the Geisinger-Regeneron DiscovEHR cohort, we identified 5 patients heterozygous for nonsense mutations causing early terminations at Glu307 or Leu328 on the C-terminus of melanocortin 4 receptor (MC4R). Two Q307Ter carriers are severely obese (BMI > 40), while one is overweight (BMI > 25). One L328Ter carrier is overweight and the other is lean. Pedigree analysis for two Q307Ter carriers shows segregation of the variant with higher BMI. Functionally, MC4R(Q307Ter) eliminated receptor surface expression and signaling, while MC4R(L328Ter) functioned like the wild-type receptor. MC4R(Q307Ter) is therefore a loss of function (LOF) variant and the region between the two truncation sites identified in our patients is critical to MC4R function. Truncating MC4R at various C-terminal positions between these two variant sites, we find that cysteine318 sits at a critical junction for receptor trafficking and function. We show that MC4R is lipid modified at cysteine318 and cysteine319. Therefore, truncation early in the MC4R C-terminus results in haploinsufficiency in humans while truncation after the first lipid-modification site is well tolerated. MC4R haploinsufficiency clearly segregates with higher BMI; however, severe obesity is not fully penetrant even in MC4R LOF carriers, suggesting critical roles for environmental and lifestyle factors in MC4R monogenic obesity.
Collapse
Affiliation(s)
- Bryn S Moore
- Molecular and Functional Genomics, Weis Center for Research Geisinger Clinic, Danville, PA, United States
| | - Tooraj Mirshahi
- Molecular and Functional Genomics, Weis Center for Research Geisinger Clinic, Danville, PA, United States. .,Geisinger Obesity Institute, Geisinger Clinic, Danville, PA, United States.
| |
Collapse
|
26
|
Differential body weight, blood pressure and placental inflammatory responses to normal versus high-fat diet in melanocortin-4 receptor-deficient pregnant rats. J Hypertens 2017; 34:1998-2007. [PMID: 27467764 DOI: 10.1097/hjh.0000000000001059] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Although obesity increases the risk for hypertensive disorders of pregnancy, the mechanisms remain unclear. Neural melanocortin-4 receptor (MC4R) deficiency causes hyperphagia and obesity. Effects of MC4R deficiency on body weight, blood pressure (BP) and placental inflammatory responses to high-fat diet (HFD) are unknown. We tested two hypotheses: MC4R deficiency results in higher body weight, BP and placental inflammation under normal-fat diet (NFD) conditions and HFD exaggerates these responses in MC4R-deficient pregnant rats. METHODS MC4R and MC4R rats were maintained on NFD (13% kcal fat) or HFD (40% kcal fat) for ∼15 weeks, then measurements made on gestational day 19. RESULTS MC4R pregnant rats had greater body mass and total body fat and visceral adipose tissue weights along with greater circulating total cholesterol (TC) and leptin levels than MC4R rats regardless of diet. On NFD, circulating adiponectin levels were lower and placental TNFα levels and BP (conscious with carotid catheter) were higher in these heavier rats. Circulating adiponectin levels were lower and placental TNFα levels and BP were higher in MC4R rats compared with NFD controls. These parameters were not affected by HFD in the already heavier and hypertensive MC4R pregnant rats. CONCLUSION Obesity in MC4R deficiency and HFD in MC4R rats result in higher BP and placental inflammation during pregnancy. However, HFD did not exaggerate these responses in already obese MC4R pregnant rats. These data suggest that obesity and HFD are independently related to hypertension and placental inflammation in pregnancy.
Collapse
|
27
|
Lede V, Meusel A, Garten A, Popkova Y, Penke M, Franke C, Ricken A, Schulz A, Kiess W, Huster D, Schöneberg T, Schiller J. Altered hepatic lipid metabolism in mice lacking both the melanocortin type 4 receptor and low density lipoprotein receptor. PLoS One 2017; 12:e0172000. [PMID: 28207798 PMCID: PMC5313158 DOI: 10.1371/journal.pone.0172000] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 01/30/2017] [Indexed: 01/01/2023] Open
Abstract
Obesity is often associated with dyslipidemia and hepatosteatosis. A number of animal models of non-alcoholic fatty liver disease (NAFLD) are established but they significantly differ in the molecular and biochemical changes depending on the genetic modification and diet used. Mice deficient for melanocortin type 4 receptor (Mc4rmut) develop hyperphagia, obesity, and subsequently NAFLD already under regular chow and resemble more closely the energy supply-driven obesity found in humans. This animal model was used to assess the molecular and biochemical consequences of hyperphagia-induced obesity on hepatic lipid metabolism. We analyzed transcriptome changes in Mc4rmut mice by RNA sequencing and used high resolution 1H magic angle spinning NMR spectroscopy and MALDI-TOF mass spectrometry to assess changes in the lipid composition. On the transcriptomic level we found significant changes in components of the triacylglycerol metabolism, unsaturated fatty acids biosynthesis, peroxisome proliferator-activated receptor signaling pathways, and lipid transport and storage compared to the wild-type. These findings were supported by increases in triacylglycerol, monounsaturated fatty acid, and arachidonic acid levels. The transcriptome signatures significantly differ from those of other NAFLD mouse models supporting the concept of hepatic subphenotypes depending on the genetic background and diet. Comparative analyses of our data with previous studies allowed for the identification of common changes and genotype-specific components and pathways involved in obesity-associated NAFLD.
Collapse
MESH Headings
- Animals
- Diet, High-Fat/adverse effects
- Disease Models, Animal
- Female
- Gene Expression Profiling
- High-Throughput Nucleotide Sequencing
- Hypercholesterolemia/etiology
- Hypercholesterolemia/metabolism
- Hypercholesterolemia/pathology
- Lipid Metabolism
- Lipogenesis/genetics
- Liver/metabolism
- Liver/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mutation/genetics
- Non-alcoholic Fatty Liver Disease/etiology
- Non-alcoholic Fatty Liver Disease/metabolism
- Non-alcoholic Fatty Liver Disease/pathology
- Obesity/complications
- Receptor, Melanocortin, Type 4/deficiency
- Receptor, Melanocortin, Type 4/genetics
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
Collapse
Affiliation(s)
- Vera Lede
- Molecular Biochemistry, Rudolf-Schönheimer-Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| | - Andrej Meusel
- Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
| | - Antje Garten
- Hospital for Children & Adolescents, Department of Women and Child Health, Center for Pediatric Research Leipzig, University of Leipzig, Leipzig, Germany
| | - Yulia Popkova
- Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
| | - Melanie Penke
- Hospital for Children & Adolescents, Department of Women and Child Health, Center for Pediatric Research Leipzig, University of Leipzig, Leipzig, Germany
| | | | - Albert Ricken
- Institute of Anatomy, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Angela Schulz
- Molecular Biochemistry, Rudolf-Schönheimer-Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| | - Wieland Kiess
- Hospital for Children & Adolescents, Department of Women and Child Health, Center for Pediatric Research Leipzig, University of Leipzig, Leipzig, Germany
| | - Daniel Huster
- Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
| | - Torsten Schöneberg
- Molecular Biochemistry, Rudolf-Schönheimer-Institute of Biochemistry, University of Leipzig, Leipzig, Germany
- * E-mail: (JS); (TS)
| | - Jürgen Schiller
- Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
- * E-mail: (JS); (TS)
| |
Collapse
|
28
|
Elfers CT, Roth CL. Robust Reductions of Excess Weight and Hyperphagia by Beloranib in Rat Models of Genetic and Hypothalamic Obesity. Endocrinology 2017; 158:41-55. [PMID: 27849360 DOI: 10.1210/en.2016-1665] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 11/15/2016] [Indexed: 02/08/2023]
Abstract
Hypothalamic lesions or deficient melanocortin (MC) signaling via MC4 receptor (MC4r) mutations often lead to hyperphagia and severe treatment-resistant obesity. We tested the methionine aminopeptidase 2-inhibitor beloranib (ZGN-440) in 2 male rat models of obesity, one modeling hypothalamic obesity with a combined medial hypothalamic lesion (CMHL) and the other modeling a monogenic form of obesity with MC4r mutations (MC4r knockout [MC4rKO]). In CMHL rats (age 3 months), postsurgery excess weight gain was significantly inhibited (ZGN-440, 0.2 ± 0.7 g/d; vehicle, 3.8 ± 0.6 g/d; P < 0.001) during 12 days of ZGN-440 treatment (0.1 mg/kg daily subcutaneously) together with a 30% reduction of daily food intake vs vehicle injection. In addition, ZGN-440 treatment improved glucose tolerance and reduced plasma insulin, and circulating levels of α-melanocyte stimulating hormone were increased. Serum lipid levels did not differ significantly in ZGN-440-treated vs vehicle-treated rats. Similar results were found in MC4rKO rats: ZGN-440 treatment (14-21 d) was associated with significant reductions of body weight gain (MC4rKO, -1.7 ± 0.6 vs 2.8 ± 0.4 g/d; lean wild-type controls, -0.7 ± 0.2 vs 1.7 ± 0.7 g/d; ZGN-440 vs vehicle, respectively), reduction of food intake (MC4rKO, -28%; lean controls, -7.5%), and insulin resistance, whereas circulating levels of interleukin-1β did not change. In both obesity models, body temperature and locomotor activity were not affected by ZGN-440 treatment. In conclusion, the robust reduction of body weight in response to ZGN-440 observed in rats with severe obesity is related to a strong reduction of food intake that is likely related to changes in the central regulation of feeding.
Collapse
Affiliation(s)
- Clinton T Elfers
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101; and
| | - Christian L Roth
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101; and
- Division of Endocrinology, Department of Pediatrics, University of Washington, Seattle, Washington 98105
| |
Collapse
|
29
|
Almundarij TI, Smyers ME, Spriggs A, Heemstra LA, Beltz L, Dyne E, Ridenour C, Novak CM. Physical Activity, Energy Expenditure, and Defense of Body Weight in Melanocortin 4 Receptor-Deficient Male Rats. Sci Rep 2016; 6:37435. [PMID: 27886210 PMCID: PMC5122857 DOI: 10.1038/srep37435] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/28/2016] [Indexed: 01/28/2023] Open
Abstract
Melanocortin 4 receptor (MC4R) variants contribute to human obesity, and rats lacking functional MC4R (Mc4rK314X/K314X) are obese. We investigated the hypothesis that low energy expenditure (EE) and physical activity contribute to this obese phenotype in male rats, and determined whether lack of functional MC4R conferred protection from weight loss during 50% calorie restriction. Though Mc4rK314X/K314X rats showed low brown adipose Ucp1 expression and were less physically active than rats heterozygous for the mutation (Mc4r+/K314X) or wild-type (Mc4r+/+) rats, we found no evidence of lowered EE in Mc4rK314X/K314X rats once body weight was taken into account using covariance. Mc4rK314X/K314X rats had a significantly higher respiratory exchange ratio. Compared to Mc4r+/+ rats, Mc4rK314X/K314X and Mc4r+/K314X rats lost less lean mass during calorie restriction, and less body mass when baseline weight was accounted for. Limited regional overexpression of Mc3r was found in the hypothalamus. Although lower physical activity levels in rats with nonfunctional MC4R did not result in lower total EE during free-fed conditions, rats lacking one or two functional copies of Mc4r showed conservation of mass, particularly lean mass, during energy restriction. This suggests that variants affecting MC4R function may contribute to individual differences in the metabolic response to food restriction.
Collapse
Affiliation(s)
- Tariq I Almundarij
- College of Agriculture and Veterinary Medicine, Al-Qassim University, Buraydah, Al-Qassim Province, Saudi Arabia.,Department of Biological Sciences, Kent State University, Kent, OH, 44242, US
| | - Mark E Smyers
- School of Biomedical Sciences, Kent State University, Kent, OH, 44242, US
| | - Addison Spriggs
- Department of Biological Sciences, Kent State University, Kent, OH, 44242, US
| | - Lydia A Heemstra
- Department of Biological Sciences, Kent State University, Kent, OH, 44242, US
| | - Lisa Beltz
- Department of Natural Sciences, Malone University, Canton, OH, 44709, US
| | - Eric Dyne
- School of Biomedical Sciences, Kent State University, Kent, OH, 44242, US.,Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, US
| | - Caitlyn Ridenour
- Department of Natural Sciences, Malone University, Canton, OH, 44709, US
| | - Colleen M Novak
- Department of Biological Sciences, Kent State University, Kent, OH, 44242, US.,School of Biomedical Sciences, Kent State University, Kent, OH, 44242, US
| |
Collapse
|
30
|
You P, Hu H, Chen Y, Zhao Y, Yang Y, Wang T, Xing R, Shao Y, Zhang W, Li D, Chen H, Liu M. Effects of Melanocortin 3 and 4 Receptor Deficiency on Energy Homeostasis in Rats. Sci Rep 2016; 6:34938. [PMID: 27713523 PMCID: PMC5054679 DOI: 10.1038/srep34938] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/20/2016] [Indexed: 01/08/2023] Open
Abstract
Melanocortin-3 and 4 receptors (MC3R and MC4R) can regulate energy homeostasis, but their respective roles especially the functions of MC3R need more exploration. Here Mc3r and Mc4r single and double knockout (DKO) rats were generated using CRISPR-Cas9 system. Metabolic phenotypes were examined and data were compared systematically. Mc3r KO rats displayed hypophagia and decreased body weight, while Mc4r KO and DKO exhibited hyperphagia and increased body weight. All three mutants showed increased white adipose tissue mass and adipocyte size. Interestingly, although Mc3r KO did not show a significant elevation in lipids as seen in Mc4r KO, DKO displayed even higher lipid levels than Mc4r KO. DKO also showed more severe glucose intolerance and hyperglycaemia than Mc4r KO. These data demonstrated MC3R deficiency caused a reduction of food intake and body weight, whereas at the same time exhibited additive effects on top of MC4R deficiency on lipid and glucose metabolism. This is the first phenotypic analysis and systematic comparison of Mc3r KO, Mc4r KO and DKO rats on a homogenous genetic background. These mutant rats will be important in defining the complicated signalling pathways of MC3R and MC4R. Both Mc4r KO and DKO are good models for obesity and diabetes research.
Collapse
Affiliation(s)
- Panpan You
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Handan Hu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Yuting Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Yongliang Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Yiqing Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Tongtong Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Roumei Xing
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Yanjiao Shao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Wen Zhang
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Huaqing Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China.,Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas 77030, USA
| |
Collapse
|
31
|
van der Klaauw AA, Keogh JM, Henning E, Stephenson C, Kelway S, Trowse VM, Subramanian N, O'Rahilly S, Fletcher PC, Farooqi IS. Divergent effects of central melanocortin signalling on fat and sucrose preference in humans. Nat Commun 2016; 7:13055. [PMID: 27701398 PMCID: PMC5059464 DOI: 10.1038/ncomms13055] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 08/31/2016] [Indexed: 01/13/2023] Open
Abstract
Melanocortin-4-receptor (MC4R)-expressing neurons modulate food intake and preference in rodents but their role in human food preference is unknown. Here we show that compared with lean and weight-matched controls, MC4R deficient individuals exhibited a markedly increased preference for high fat, but a significantly reduced preference for high sucrose food. These effects mirror those in Mc4r null rodents and provide evidence for a central molecular circuit influencing human macronutrient preference. Hypothalamic melanocortin-4-receptors (MC4R) regulate food preference in rodents, but their role in humans is unclear. Here, the authors perform food preference and liking tests in humans with MC4R mutations and find that they prefer fatty food more, but sweet food less, than people without MC4R mutations.
Collapse
Affiliation(s)
- Agatha A van der Klaauw
- Clinical Biochemistry, University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Julia M Keogh
- Clinical Biochemistry, University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Elana Henning
- Clinical Biochemistry, University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Cheryl Stephenson
- Clinical Biochemistry, University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Sarah Kelway
- Clinical Biochemistry, University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Victoria M Trowse
- Clinical Biochemistry, University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Naresh Subramanian
- Clinical Biochemistry, University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK.,Department of Psychiatry, University of Cambridge and the Cambridgeshire and Peterborough NHS Foundation Trust Cambridge, Cambridge CB2 0SZ, UK
| | - Stephen O'Rahilly
- Clinical Biochemistry, University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Paul C Fletcher
- Clinical Biochemistry, University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK.,Department of Psychiatry, University of Cambridge and the Cambridgeshire and Peterborough NHS Foundation Trust Cambridge, Cambridge CB2 0SZ, UK
| | - I Sadaf Farooqi
- Clinical Biochemistry, University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| |
Collapse
|
32
|
Tan HY, Steyn FJ, Huang L, Cowley M, Veldhuis JD, Chen C. Hyperphagia in male melanocortin 4 receptor deficient mice promotes growth independently of growth hormone. J Physiol 2016; 594:7309-7326. [PMID: 27558671 DOI: 10.1113/jp272770] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/22/2016] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS Loss of function of the melanocortin 4 receptor (MC4R) results in hyperphagia, obesity and increased growth. Despite knowing that MC4Rs control food intake, we are yet to understand why defects in the function of the MC4R receptor contribute to rapid linear growth. We show that hyperphagia following germline loss of MC4R in male mice promotes growth while suppressing the growth hormone-insulin-like growth factor-1 (GH-IGF-1) axis. We propose that hyperinsulinaemia promotes growth while suppressing the GH-IGF-1 axis. It is argued that physiological responses essential to maintain energy flux override conventional mechanisms of pubertal growth to promote the storage of excess energy while ensuring growth. ABSTRACT Defects in melanocortin-4-receptor (MC4R) signalling result in hyperphagia, obesity and increased growth. Clinical observations suggest that loss of MC4R function may enhance growth hormone (GH)-mediated growth, although this remains untested. Using male mice with germline loss of the MC4R, we assessed pulsatile GH release and insulin-like growth factor-1 (IGF-1) production and/or release relative to pubertal growth. We demonstrate early-onset suppression of GH release in rapidly growing MC4R deficient (MC4RKO) mice, confirming that increased linear growth in MC4RKO mice does not occur in response to enhanced activation of the GH-IGF-1 axis. The progressive suppression of GH release in MC4RKO mice occurred alongside increased adiposity and the progressive worsening of hyperphagia-associated hyperinsulinaemia. We next prevented hyperphagia in MC4RKO mice through restricting calorie intake in these mice to match that of wild-type (WT) littermates. Pair feeding of MC4RKO mice did not prevent increased adiposity, but attenuated hyperinsulinaemia, recovered GH release, and normalized linear growth rate to that seen in pair-fed WT littermate controls. We conclude that the suppression of GH release in MC4RKO mice occurs independently of increased adipose mass, and is a consequence of hyperphagia-associated hyperinsulinaemia. It is proposed that physiological responses essential to maintain energy flux (hyperinsulinaemia and the suppression of GH release) override conventional mechanisms of pubertal growth to promote the storage of excess energy while ensuring growth. Implications of these findings are likely to extend beyond individuals with defects in MC4R signalling, encompassing physiological changes central to mechanisms of growth and energy homeostasis universal to hyperphagia-associated childhood-onset obesity.
Collapse
Affiliation(s)
- H Y Tan
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - F J Steyn
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia.,The University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia
| | - L Huang
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - M Cowley
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - J D Veldhuis
- Department of Medicine, Endocrine Research Unit, Mayo School of Graduate Medical Education, Clinical Translational Science Center, Mayo Clinic, Rochester, MN, USA
| | - C Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
33
|
Gavini CK, Jones WC, Novak CM. Ventromedial hypothalamic melanocortin receptor activation: regulation of activity energy expenditure and skeletal muscle thermogenesis. J Physiol 2016; 594:5285-301. [PMID: 27126579 PMCID: PMC5023712 DOI: 10.1113/jp272352] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 04/25/2016] [Indexed: 01/13/2023] Open
Abstract
KEY POINTS The ventromedial hypothalamus (VMH) and the central melanocortin system both play vital roles in regulating energy balance by modulating energy intake and utilization. Recent evidence suggests that activation of the VMH alters skeletal muscle metabolism. We show that intra-VMH melanocortin receptor activation increases energy expenditure and physical activity, switches fuel utilization to fats, and lowers work efficiency such that excess calories are dissipated by skeletal muscle as heat. We also show that intra-VMH melanocortin receptor activation increases sympathetic nervous system outflow to skeletal muscle. Intra-VMH melanocortin receptor activation also induced significant changes in the expression of mediators of energy expenditure in muscle. These results support the role of melanocortin receptors in the VMH in the modulation of skeletal muscle metabolism. ABSTRACT The ventromedial hypothalamus (VMH) and the brain melanocortin system both play vital roles in increasing energy expenditure (EE) and physical activity, decreasing appetite and modulating sympathetic nervous system (SNS) outflow. Because of recent evidence showing that VMH activation modulates skeletal muscle metabolism, we propose the existence of an axis between the VMH and skeletal muscle, modulated by brain melanocortins, modelled on the brain control of brown adipose tissue. Activation of melanocortin receptors in the VMH of rats using a non-specific agonist melanotan II (MTII), compared to vehicle, increased oxygen consumption and EE and decreased the respiratory exchange ratio. Intra-VMH MTII enhanced activity-related EE even when activity levels were held constant. MTII treatment increased gastrocnemius muscle heat dissipation during controlled activity, as well as in the home cage. Compared to vehicle-treated rats, rats with intra-VMH melanocortin receptor activation had higher skeletal muscle norepinephrine turnover, indicating an increased SNS drive to muscle. Lastly, intra-VMH MTII induced mRNA expression of muscle energetic mediators, whereas short-term changes at the protein level were primarily limited to phosphorylation events. These results support the hypothesis that melanocortin peptides act in the VMH to increase EE by lowering the economy of activity via the enhanced expression of mediators of EE in the periphery including skeletal muscle. The data are consistent with the role of melanocortins in the VMH in the modulation of skeletal muscle metabolism.
Collapse
MESH Headings
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/physiology
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/physiology
- Animals
- Energy Metabolism
- Hypothalamus/physiology
- Liver/drug effects
- Liver/metabolism
- Liver/physiology
- Male
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiology
- Norepinephrine/metabolism
- Peptides, Cyclic/pharmacology
- Physical Conditioning, Animal
- Rats, Sprague-Dawley
- Receptors, Melanocortin/agonists
- Receptors, Melanocortin/physiology
- Thermogenesis
- alpha-MSH/analogs & derivatives
- alpha-MSH/pharmacology
Collapse
Affiliation(s)
- Chaitanya K Gavini
- School of Biomedical Sciences, Kent State University, Kent, OH, USA.
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA.
| | - William C Jones
- Department of Exercise Science/Physiology, College of Education, Health, and Human Services, Kent State University, Kent, OH, USA
| | - Colleen M Novak
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| |
Collapse
|
34
|
Spradley FT, Sasser JM, Musall JB, Sullivan JC, Granger JP. Nitric oxide synthase-mediated blood pressure regulation in obese melanocortin-4 receptor-deficient pregnant rats. Am J Physiol Regul Integr Comp Physiol 2016; 311:R851-R857. [PMID: 27534879 DOI: 10.1152/ajpregu.00285.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 08/14/2016] [Indexed: 11/22/2022]
Abstract
Although obesity increases the risk for hypertension in pregnancy, the mechanisms responsible are unknown. Increased nitric oxide (NO) production results in vasodilation and reduced blood pressure during normal pregnancy in lean rats; however, the role of NO is less clear during obese pregnancies. We examined the impact of obesity on NO synthase (NOS)-mediated regulation of blood pressure during pregnancy by testing the hypothesis that NOS activity, expression, and regulation of vascular tone and blood pressure are reduced in obese pregnant rats. At gestational day 19, melanocortin-4 receptor (MC4R)-deficient obese rats (MC4R) had greater body weight and fat mass with elevated blood pressure and circulating sFlt-1 levels compared with MC4R pregnant rats. MC4R pregnant rats also had less circulating cGMP levels and reduced total NOS enzymatic activity and expression in mesenteric arteries. Despite decreased biochemical measures of NO/NOS in MC4R rats, NOS inhibition enhanced vasoconstriction only in mesenteric arteries from MC4R rats, suggesting greater NOS-mediated tone. To examine the role of NOS on blood pressure regulation in obese pregnant rats, MC4R and MC4R pregnant rats were administered the nonselective NOS inhibitor NG-nitro-l-arginine methyl ester (l-NAME, 100 mg/l) from gestational day 14 to 19 in drinking water. The degree by which l-NAME raised blood pressure was similar between obese and lean pregnant rats. Although MC4R obese pregnant rats had elevated blood pressure associated with reduced total NOS activity and expression, they had enhanced NOS-mediated attenuation of vasoconstriction, with no evidence of alterations in NOS-mediated regulation of blood pressure.
Collapse
Affiliation(s)
- Frank T Spradley
- Department of Surgery, The University of Mississippi Medical Center, Jackson, Mississippi; .,Department of Physiology and Biophysics, The University of Mississippi Medical Center, Jackson, Mississippi.,Cardiovascular-Renal Research Center, The University of Mississippi Medical Center, Jackson, Mississippi.,Women's Health Research Center, The University of Mississippi Medical Center, Jackson, Mississippi; and
| | - Jennifer M Sasser
- Department of Pharmacology and Toxicology, The University of Mississippi Medical Center, Jackson, Mississippi.,Cardiovascular-Renal Research Center, The University of Mississippi Medical Center, Jackson, Mississippi.,Women's Health Research Center, The University of Mississippi Medical Center, Jackson, Mississippi; and
| | | | | | - Joey P Granger
- Department of Physiology and Biophysics, The University of Mississippi Medical Center, Jackson, Mississippi.,Cardiovascular-Renal Research Center, The University of Mississippi Medical Center, Jackson, Mississippi.,Women's Health Research Center, The University of Mississippi Medical Center, Jackson, Mississippi; and
| |
Collapse
|
35
|
Melanocortin 3 Receptor Signaling in Midbrain Dopamine Neurons Increases the Motivation for Food Reward. Neuropsychopharmacology 2016; 41:2241-51. [PMID: 26852738 PMCID: PMC4946052 DOI: 10.1038/npp.2016.19] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/31/2016] [Accepted: 02/01/2016] [Indexed: 01/21/2023]
Abstract
The central melanocortin (MC) system mediates its effects on food intake via MC3 (MC3R) and MC4 receptors (MC4R). Although the role of MC4R in meal size determination, satiation, food preference, and motivation is well established, the involvement of MC3R in the modulation of food intake has been less explored. Here, we investigated the role of MC3R on the incentive motivation for food, which is a crucial component of feeding behavior. Dopaminergic neurons within the ventral tegmental area (VTA) have a crucial role in the motivation for food. We here report that MC3Rs are expressed on VTA dopaminergic neurons and that pro-opiomelanocortinergic (POMC) neurons in the arcuate nucleus of the hypothalamus (Arc) innervate these VTA dopaminergic neurons. Our findings show that intracerebroventricular or intra-VTA infusion of the selective MC3R agonist γMSH increases responding for sucrose under a progressive ratio schedule of reinforcement, but not free sucrose consumption in rats. Furthermore, ex vivo electrophysiological recordings show increased VTA dopaminergic neuronal activity upon γMSH application. Consistent with a dopamine-mediated effect of γMSH, the increased motivation for sucrose after intra-VTA infusion of γMSH was blocked by pretreatment with the dopamine receptor antagonist α-flupenthixol. Taken together, we demonstrate an Arc POMC projection onto VTA dopaminergic neurons that modulates motivation for palatable food via activation of MC3R signaling.
Collapse
|
36
|
Cigarroa I, Lalanza JF, Caimari A, del Bas JM, Capdevila L, Arola L, Escorihuela RM. Treadmill Intervention Attenuates the Cafeteria Diet-Induced Impairment of Stress-Coping Strategies in Young Adult Female Rats. PLoS One 2016; 11:e0153687. [PMID: 27099927 PMCID: PMC4839746 DOI: 10.1371/journal.pone.0153687] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/03/2016] [Indexed: 12/12/2022] Open
Abstract
The current prevalence of diet-induced overweight and obesity in adolescents and adults is continuously growing. Although the detrimental biochemical and metabolic consequences of obesity are widely studied, its impact on stress-coping behavior and its interaction with specific exercise doses (in terms of intensity, duration and frequency) need further investigation. To this aim, we fed adolescent rats either an obesogenic diet (cafeteria diet, CAF) or standard chow (ST). Each group was subdivided into four subgroups according to the type of treadmill intervention as follows: a sedentary group receiving no manipulation; a control group exposed to a stationary treadmill; a low-intensity treadmill group trained at 12 m/min; and a higher intensity treadmill group trained at 17 m/min. Both the diet and treadmill interventions started at weaning and lasted for 8 weeks. Subjects were tested for anxiety-like behavior in the open field test and for coping strategies in the two-way active avoidance paradigm at week 7 and were sacrificed at week 8 for biometric and metabolic characterization. CAF feeding increased the weight gain, relative retroperitoneal white adipose tissue (RWAT %), and plasma levels of glucose, insulin, triglycerides and leptin and decreased the insulin sensitivity. Treadmill intervention partially reversed the RWAT% and triglyceride alterations; at higher intensity, it decreased the leptin levels of CAF-fed animals. CAF feeding decreased the motor activity and impaired the performance in a two-way active avoidance assessment. Treadmill intervention reduced defecation in the shuttle box, suggesting diminished anxiety. CAF feeding combined with treadmill training at 17 m/min increased the time spent in the center of the open field and more importantly, partially reversed the two-way active avoidance deficit. In conclusion, this study demonstrates that at doses that decreased anxiety-like behavior, treadmill exercise partially improved the coping strategy in terms of active avoidance behavior in the CAF-fed animals. This effect was not observed at lower doses of treadmill training.
Collapse
Affiliation(s)
- Igor Cigarroa
- Institut de Neurociències, Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona, Barcelona, Spain
- Carrera de Kinesiología, Facultad de Salud, Universidad Santo Tomás, Los Ángeles, región del Bio-Bio, Chile
| | - Jaume F. Lalanza
- Institut de Neurociències, Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Antoni Caimari
- Grup de Recerca en Nutrició i Salut (GRNS). Centre Tecnològic de Nutrició i Salut (CTNS), TECNIO, CEICS, Reus, Spain
| | - Josep M. del Bas
- Grup de Recerca en Nutrició i Salut (GRNS). Centre Tecnològic de Nutrició i Salut (CTNS), TECNIO, CEICS, Reus, Spain
| | - Lluís Capdevila
- Laboratori de Psicologia de l’Esport, Departament de Psicologia Bàsica, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lluís Arola
- Departament de Bioquímica i Biotecnologia, Nutrigenomics Research Group, Universitat Rovira i Virgili, Tarragona, Spain
- Centre Tecnològic de Nutrició i Salut (CTNS), TECNIO, CEICS, Reus, Spain
| | - Rosa M. Escorihuela
- Institut de Neurociències, Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona, Barcelona, Spain
- * E-mail:
| |
Collapse
|
37
|
Heinrich G, Russo L, Castaneda TR, Pfeiffer V, Ghadieh HE, Ghanem SS, Wu J, Faulkner LD, Ergün S, McInerney MF, Hill JW, Najjar SM. Leptin Resistance Contributes to Obesity in Mice with Null Mutation of Carcinoembryonic Antigen-related Cell Adhesion Molecule 1. J Biol Chem 2016; 291:11124-32. [PMID: 27002145 DOI: 10.1074/jbc.m116.716431] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Indexed: 01/28/2023] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) promotes hepatic insulin clearance. Consistently, mice with null mutation of Ceacam1 (Cc1(-/-)) exhibit impaired insulin clearance with increased lipid production in liver and redistribution to white adipose tissue, leading to visceral obesity at 2 months of age. When the mutation is propagated on the C57/BL6J genetic background, total fat mass rises significantly with age, and glucose intolerance and systemic insulin resistance develop at 6 months of age. This study was carried out to determine the mechanisms underlying the marked increase in total fat mass in 6-month-old mutants. Indirect calorimetry analysis showed that Cc1(-/-) mice develop hyperphagia and a significant reduction in physical activity, in particular in the early hours of the dark cycle, during which energy expenditure is only slightly lower than in wild-type mice. They also exhibit increased triglyceride accumulation in skeletal muscle, due in part to incomplete fatty acid β-oxidation. Mechanistically, hypothalamic leptin signaling is reduced, as demonstrated by blunted STAT3 phosphorylation in coronal sections in response to an intracerebral ventricular injection of leptin. Hypothalamic fatty-acid synthase activity is also elevated in the mutants. Together, the data show that the increase in total fat mass in Cc1(-/-) mice is mainly attributed to hyperphagia and reduced spontaneous physical activity. Although the contribution of the loss of CEACAM1 from anorexigenic proopiomelanocortin neurons in the arcuate nucleus is unclear, leptin resistance and elevated hypothalamic fatty-acid synthase activity could underlie altered energy balance in these mice.
Collapse
Affiliation(s)
| | - Lucia Russo
- From the Center for Diabetes and Endocrine Research and
| | - Tamara R Castaneda
- From the Center for Diabetes and Endocrine Research and Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, Toledo, Ohio 43614
| | - Verena Pfeiffer
- the Institut für Anatomie und Zellbiologie, Universität Würzburg, 97070 Würzburg, Germany, and
| | | | | | - Jieshen Wu
- From the Center for Diabetes and Endocrine Research and
| | | | - Süleyman Ergün
- the Institut für Anatomie und Zellbiologie, Universität Würzburg, 97070 Würzburg, Germany, and
| | - Marcia F McInerney
- From the Center for Diabetes and Endocrine Research and Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, Toledo, Ohio 43614
| | | | - Sonia M Najjar
- From the Center for Diabetes and Endocrine Research and the Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701
| |
Collapse
|
38
|
van den Heuvel JK, Eggels L, van Rozen AJ, Fliers E, Kalsbeek A, Adan RAH, la Fleur SE. Inhibitory Effect of the Melanocortin Receptor Agonist Melanotan-II (MTII) on Feeding Depends on Dietary Fat Content and not Obesity in Rats on Free-Choice Diets. Front Behav Neurosci 2015; 9:358. [PMID: 26733840 PMCID: PMC4689860 DOI: 10.3389/fnbeh.2015.00358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 12/09/2015] [Indexed: 02/02/2023] Open
Abstract
Introduction: Conflicting data exist on sensitivity changes of the melanocortin system during diet-induced obesity. We hypothesized that melanocortin sensitivity depends on diet composition, in particular on the fat content rather than the level of obesity. The aim of this study was to determine the influence of diet composition on feeding responses to a melanocortin receptor agonist, using free-choice diets that differ in food components. Methods: Male Wistar rats were subjected to a chow (CHOW) diet or a free-choice (fc) diet of either chow, saturated fat and liquid sugar (fcHFHS), chow and saturated fat (fcHF), or chow and liquid sugar (fcHS) for 4 weeks. Melanocortin sensitivity was tested by measuring food intake following administration of the melanocortin 3/4 receptor agonist melanotan II (MTII) or vehicle in the lateral ventricle. In a separate experiment, proopiomelanocortin (POMC) and agouti-related protein (AgRP) mRNA levels were determined in the arcuate nucleus with in situ hybridization in rats subjected to the free-choice diets for 4 weeks. Results: Rats on the fcHFHS diet for 4 weeks show increased caloric intake and body weight gain compared to rats on the CHOW, fcHS and fcHF diet. Caloric intake and body weight gain was comparable between rats on the fcHF, fcHS, and CHOW diet. After 4 weeks diet, POMC and AgRP mRNA levels were not different between diet groups. MTII inhibited caloric intake to a larger extent in rats on the fcHF diet compared to rats on the CHOW, fcHFHS or fcHS diet. Moreover, the fat component was the most inhibited by MTII, and the sugar component the least. Conclusion: Rats on the fcHF diet show stronger food intake inhibition to the melanocortin receptor agonist MTII than rats on the CHOW, fcHS, and fcHFHS diet, which is independent of caloric intake and body weight gain. Our data point toward an important role for diet composition, particularly the dietary fat content, and not obesity in the sensitivity of the melanocortin system.
Collapse
Affiliation(s)
- José K van den Heuvel
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam Amsterdam, Netherlands
| | - Leslie Eggels
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam Amsterdam, Netherlands
| | - Andrea J van Rozen
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, Utrecht University Medical Centre Utrecht, Netherlands
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam Amsterdam, Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Academic Medical Center, University of AmsterdamAmsterdam, Netherlands; Hypothalamic Integration Mechanisms, Netherlands Institute for NeuroscienceAmsterdam, Netherlands
| | - Roger A H Adan
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, Utrecht University Medical Centre Utrecht, Netherlands
| | - Susanne E la Fleur
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam Amsterdam, Netherlands
| |
Collapse
|
39
|
Kim M, Song SB, Cha YS. Effects of black adzuki bean (Vigna angularis, Geomguseul) extract on body composition and hypothalamic neuropeptide expression in rats fed a high-fat diet. Food Nutr Res 2015; 59:27719. [PMID: 26493717 PMCID: PMC4615815 DOI: 10.3402/fnr.v59.27719] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 10/05/2015] [Accepted: 10/05/2015] [Indexed: 01/18/2023] Open
Abstract
Background Obesity is often considered to result from either excessive food intake or insufficient physical activity. Adzuki beans have been evaluated as potential remedies for various health conditions, and recent studies have reported their effects on the regulation of lipid metabolism, but it remains to be determined whether they may be effective in overcoming obesity by regulating appetite and satiety. Objective This study investigated the effect of black adzuki bean (BAB) extract on body composition and hypothalamic neuropeptide expression in Sprague Dawley rats (Rattus norvegicus) fed a high-fat diet. Design The rats were fed for 8 weeks with a control diet containing 10 kcal% from fat (CD), a high-fat diet containing 60 kcal% from fat (HD), or a high-fat diet with 1% or 2% freeze-dried ethanolic extract powder of BAB (BAB-1 and BAB-2). Results The body weights and epididymal fat weights were significantly reduced and the serum lipid profiles were improved in the group fed the diet containing BAB compared to the HD group. The expression of AGRP mRNA significantly decreased in the BAB groups, and treatment with BAB-2 resulted in a marked induction of the mRNA expression of POMC and CART, which are anorexigenic neuropeptides that suppress food intake. Furthermore, mRNA expression levels of ObRb, a gene related to leptin sensitivity in the hypothalamus, were significantly higher in the BAB groups than in the HD group. Conclusions These results suggest that supplementation with BAB has a significant effect on body weight via regulation of hypothalamic neuropeptides.
Collapse
Affiliation(s)
- Mina Kim
- Department of Food Science and Human Nutrition, BK21 PLUS Program, Chonbuk National University, Jeonju, Republic of Korea
| | - Seok-Bo Song
- Department of Functional Crops, National Institute of Crop Science, Rural Development Administration, Miryang, Republic of Korea
| | - Youn-Soo Cha
- Department of Food Science and Human Nutrition, BK21 PLUS Program, Chonbuk National University, Jeonju, Republic of Korea;
| |
Collapse
|
40
|
Shukla C, Koch LG, Britton SL, Cai M, Hruby VJ, Bednarek M, Novak CM. Contribution of regional brain melanocortin receptor subtypes to elevated activity energy expenditure in lean, active rats. Neuroscience 2015; 310:252-67. [PMID: 26404873 DOI: 10.1016/j.neuroscience.2015.09.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/10/2015] [Accepted: 09/11/2015] [Indexed: 12/13/2022]
Abstract
Physical activity and non-exercise activity thermogenesis (NEAT) are crucial factors accounting for individual differences in body weight, interacting with genetic predisposition. In the brain, a number of neuroendocrine intermediates regulate food intake and energy expenditure (EE); this includes the brain melanocortin (MC) system, consisting of MC peptides as well as their receptors (MCR). MC3R and MC4R have emerged as critical modulators of EE and food intake. To determine how variance in MC signaling may underlie individual differences in physical activity levels, we examined behavioral response to MC receptor agonists and antagonists in rats that show high and low levels of physical activity and NEAT, that is, high- and low-capacity runners (HCR, LCR), developed by artificial selection for differential intrinsic aerobic running capacity. Focusing on the hypothalamus, we identified brain region-specific elevations in expression of MCR 3, 4, and also MC5R, in the highly active, lean HCR relative to the less active and obesity-prone LCR. Further, the differences in activity and associated EE as a result of MCR activation or suppression using specific agonists and antagonists were similarly region-specific and directly corresponded to the differential MCR expression patterns. The agonists and antagonists investigated here did not significantly impact food intake at the doses used, suggesting that the differential pattern of receptor expression may by more meaningful to physical activity than to other aspects of energy balance regulation. Thus, MCR-mediated physical activity may be a key neural mechanism in distinguishing the lean phenotype and a target for enhancing physical activity and NEAT.
Collapse
Affiliation(s)
- C Shukla
- Department of Biological Sciences, Kent State University, Kent, OH, United States; Harvard Medical School - VA Boston Healthcare System, Boston, MA, United States.
| | - L G Koch
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, United States; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - S L Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, United States; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - M Cai
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, United States
| | - V J Hruby
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, United States
| | - M Bednarek
- MedImmune Limited, Cambridge, United Kingdom
| | - C M Novak
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| |
Collapse
|
41
|
Roseberry AG, Stuhrman K, Dunigan AI. Regulation of the mesocorticolimbic and mesostriatal dopamine systems by α-melanocyte stimulating hormone and agouti-related protein. Neurosci Biobehav Rev 2015; 56:15-25. [DOI: 10.1016/j.neubiorev.2015.06.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 06/15/2015] [Accepted: 06/21/2015] [Indexed: 11/24/2022]
|
42
|
Obici S, Magrisso IJ, Ghazarian AS, Shirazian A, Miller JR, Loyd CM, Begg DP, Krawczewski Carhuatanta KA, Haas MK, Davis JF, Woods SC, Sandoval DA, Seeley RJ, Goodyear LJ, Pothos EN, Mul JD. Moderate voluntary exercise attenuates the metabolic syndrome in melanocortin-4 receptor-deficient rats showing central dopaminergic dysregulation. Mol Metab 2015; 4:692-705. [PMID: 26500841 PMCID: PMC4588435 DOI: 10.1016/j.molmet.2015.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 07/07/2015] [Accepted: 07/09/2015] [Indexed: 01/12/2023] Open
Abstract
Objective Melanocortin-4 receptors (MC4Rs) are highly expressed by dopamine-secreting neurons of the mesolimbic tract, but their functional role has not been fully resolved. Voluntary wheel running (VWR) induces adaptations in the mesolimbic dopamine system and has a myriad of long-term beneficial effects on health. In the present experiments we asked whether MC4R function regulates the effects of VWR, and whether VWR ameliorates MC4R-associated symptoms of the metabolic syndrome. Methods Electrically evoked dopamine release was measured in slice preparations from sedentary wild-type and MC4R-deficient Mc4rK314X (HOM) rats. VWR was assessed in wild-type and HOM rats, and in MC4R-deficient loxTBMc4r mice, wild-type mice body weight-matched to loxTBMc4r mice, and wild-type mice with intracerebroventricular administration of the MC4R antagonist SHU9119. Mesolimbic dopamine system function (gene/protein expression) and metabolic parameters were examined in wheel-running and sedentary wild-type and HOM rats. Results Sedentary obese HOM rats had increased electrically evoked dopamine release in several ventral tegmental area (VTA) projection sites compared to wild-type controls. MC4R loss-of-function decreased VWR, and this was partially independent of body weight. HOM wheel-runners had attenuated markers of intracellular D1-type dopamine receptor signaling despite increased dopamine flux in the VTA. VWR increased and decreased ΔFosB levels in the nucleus accumbens (NAc) of wild-type and HOM runners, respectively. VWR improved metabolic parameters in wild-type wheel-runners. Finally, moderate voluntary exercise corrected many aspects of the metabolic syndrome in HOM runners. Conclusions Central dopamine dysregulation during VWR reinforces the link between MC4R function and molecular and behavioral responding to rewards. The data also suggest that exercise can be a successful lifestyle intervention in MC4R-haploinsufficient individuals despite reduced positive reinforcement during exercise training. MC4R-deficiency causes metabolic syndrome. Loss of MC4R signaling decreases voluntary wheel running (VWR). Despite moderate amounts of VWR, MC4R-associated metabolic syndrome is severely attenuated. MC4R-deficiency is associated with mesolimbic dopamine dysregulation during VWR.
Collapse
Affiliation(s)
- Silvana Obici
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH, USA
| | - I Jack Magrisso
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH, USA
| | - Armen S Ghazarian
- Programs in Pharmacology and Experimental Therapeutics and Neuroscience, Sackler School of Graduate Biomedical Sciences and Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
| | - Alireza Shirazian
- Programs in Pharmacology and Experimental Therapeutics and Neuroscience, Sackler School of Graduate Biomedical Sciences and Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
| | - Jonas R Miller
- Programs in Pharmacology and Experimental Therapeutics and Neuroscience, Sackler School of Graduate Biomedical Sciences and Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
| | - Christine M Loyd
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH, USA
| | - Denovan P Begg
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH, USA ; School of Psychology, UNSW Australia, Sydney, NSW, Australia
| | | | - Michael K Haas
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH, USA
| | - Jon F Davis
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH, USA
| | - Stephen C Woods
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH, USA
| | - Darleen A Sandoval
- North Campus Research Complex, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Randy J Seeley
- North Campus Research Complex, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Emmanuel N Pothos
- Programs in Pharmacology and Experimental Therapeutics and Neuroscience, Sackler School of Graduate Biomedical Sciences and Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
| | - Joram D Mul
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH, USA ; Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
43
|
Wang Y, Wang C, Zhang J, Meng C, Zhang X, Wang Z, Fang Y, Mao D, Cao S. Three novel MC4R SNPs associated with growth traits in Hu sheep and East Friesian×Hu crossbred sheep. Small Rumin Res 2015. [DOI: 10.1016/j.smallrumres.2015.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
44
|
Khalilitehrani A, Qorbani M, Hosseini S, Pishva H. The association of MC4R rs17782313 polymorphism with dietary intake in Iranian adults. Gene 2015; 563:125-9. [PMID: 25769385 DOI: 10.1016/j.gene.2015.03.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 03/07/2015] [Accepted: 03/09/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND Malfunction in the energy homeostasis system is a major cause of developing obesity. Melanocortin-4 receptor (MC4R) plays a crucial role in this system as a key receptor. Although MC4R gene as an obesity candidate gene is associated with higher BMI, only few attempts have been carried out to understand the mechanism underlying body-weight regulation. OBJECTIVE The aim of this study is to investigate the association between variant rs17782313 near MC4R gene and both dietary energy and macronutrient intakes. METHODS An Iranian population, 400 adults aged over 22years were selected from the Iranian Multicenter Osteoporosis Study (IMOS). Genotyping for the near MC4R rs17782313 was performed by PCR-RFLP. Weight and height were measured. Dietary intake and physical activity were assessed by using validated questionnaires. Analysis was carried out in two groups with regard to BMI. Multiple linear regression models adjusted for covariates were used to examine the association between rs17782313 and dietary intake. RESULTS MC4R rs17782313 was associated with high energy intake (P<0.001), and low carbohydrate and protein intakes (P<0.001 and P<0.01 respectively). In addition, the significant association between variant rs17782313 and fat intake disappeared after adjusting for energy. CONCLUSIONS The rs17782313 variant contributes to the variety of dietary energy and energy-dense macronutrient intakes. Moreover, a novel association was suggested between this polymorphism and dietary fat intake.
Collapse
Affiliation(s)
- Azadeh Khalilitehrani
- Department of Cellular-Molecular Nutrition, School of Nutrition Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Qorbani
- School of Medicine, Community Medicine Department, Alborz University of Medical Sciences, Karaj, Iran
| | - Saeed Hosseini
- Endocrinology and Metabolism Research Center (EMRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamideh Pishva
- Department of Cellular-Molecular Nutrition, School of Nutrition Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
45
|
Melanocortin-4 receptor modulators for the treatment of obesity: a patent analysis (2008–2014). Pharm Pat Anal 2015; 4:95-107. [DOI: 10.4155/ppa.15.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The central melanocortin system and particularly the melanocortin-4 receptor (MC4R) subtype, plays an important role in the regulation of body weight. The discovery of orally active MC4R agonists suitable for evaluation in human clinical trials as weight loss agents has attracted considerable interest over the past decade, but has proved challenging, in part because of cardiovascular and behavioral side effects. Currently, the only MC4R agonist in clinical trials is a peptide identified as RM-493. To avoid some of the undesirable side effects associated with MC4R activation, new pharmacological approaches for modulating the MC system have been investigated. In this article, we provide a review of the MC4R patent landscape from 2008 to 2014 and analyze the physicochemical properties of compounds described herein.
Collapse
|
46
|
Central vagal afferent endings mediate reduction of food intake by melanocortin-3/4 receptor agonist. J Neurosci 2014; 34:12636-45. [PMID: 25232103 DOI: 10.1523/jneurosci.1121-14.2014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Injection of the melanocortin-3/4 receptor agonist melanotan-II (MTII) into the nucleus of the solitary tract (NTS) produces rapid and sustained reduction of food intake. Melanocortin-4 receptors (MC4Rs) are expressed by vagal afferent endings in the NTS, but it is not known whether these endings participate in MTII-induced reduction of food intake. In experiments described here, we evaluated the contribution of central vagal afferent endings in MTII-induced reduction of food intake. Examination of rat hindbrain sections revealed that neuronal processes expressing immunoreactivity for the endogenous MC4R agonist α-melanoctyte-stimulating hormone course parallel and wrap around anterogradely labeled vagal afferent endings in the NTS and thus are aptly positioned to activate vagal afferent MC4Rs. Furthermore, MTII and endogenous MC4R agonists increased protein kinase A (PKA)-catalyzed phosphorylation of synapsin I in vagal afferent endings, an effect known to increase synaptic strength by enhancing neurotransmitter release in other neural systems. Hindbrain injection of a PKA inhibitor, KT5720, significantly attenuated MTII-induced reduction of food intake and the increase in synapsin I phosphorylation. Finally, unilateral nodose ganglion removal, resulting in degeneration of vagal afferent endings in the ipsilateral NTS, abolished MTII-induced synapsin I phosphorylation ipsilateral to nodose ganglion removal. Moreover, reduction of food intake following MTII injection into the NTS ipsilateral to nodose ganglion removal was significantly attenuated, whereas the response to MTII was not diminished when injected into the contralateral NTS. Altogether, our results suggest that reduction of food intake following hindbrain MC4R activation is mediated by central vagal afferent endings.
Collapse
|
47
|
Abstract
Obesity and its related comorbidities can be detrimental for the affected individual and challenge public health systems worldwide. Currently, the only available treatment options leading to clinically significant and maintained body weight loss and reduction in obesity-related morbidity and mortality are based on surgical interventions. Apart from the 'gold standard' Roux-en-Y gastric bypass (RYGB), the vertical sleeve gastrectomy and gastric banding are two frequently performed procedures. This review will discuss animal experiments designed to understand the underlying mechanisms of body weight loss after bariatric surgery. While caloric malabsorption and mechanical restriction are no major factors in this respect, alterations in gut hormone levels are invariably found after RYGB. However, their causal role in RYGB effects on eating and body weight has recently been challenged. Other potential factors contributing to the RYGB effects include increased bile acid concentrations and an altered composition of gut microbiota. RYGB is further associated with remarkable changes in the preference for different dietary components such as a decrease in the preference for high fat or sugar; it is important to note that the contribution of altered food preferences to the RYGB effects on body weight is not clear.
Collapse
Affiliation(s)
- Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
48
|
|
49
|
Ryan KK, Mul JD, Clemmensen C, Egan AE, Begg DP, Halcomb K, Seeley RJ, Herman JP, Ulrich-Lai YM. Loss of melanocortin-4 receptor function attenuates HPA responses to psychological stress. Psychoneuroendocrinology 2014; 42:98-105. [PMID: 24636506 PMCID: PMC4120841 DOI: 10.1016/j.psyneuen.2014.01.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/12/2014] [Accepted: 01/13/2014] [Indexed: 12/27/2022]
Abstract
The melanocortin 4 receptor (MC4R), well-known for its role in the regulation of energy balance, is widely expressed in stress-regulatory brain regions, including the paraventricular nucleus of the hypothalamus (PVH) and the medial amygdala (MeA). In agreement with this, MC4R has been implicated in hypothalamic-pituitary-adrenocortical axis (HPA) regulation. The present work investigated the role of chronic Mc4r function to modulate basal HPA axis tone and to facilitate acute HPA responses to psychological stress, using a novel rat model with Mc4r loss-of-function. In this study, adult male rats were placed into 3 groups (n=15/group) according to genotype [wild-type (WT); heterozygous mutant (HET); and homozygous mutant (HOM)]. Basal (pre-stress) plasma adrenocorticotropic hormone (ACTH) and corticosterone were measured in the AM and PM, and the HPA axis response to restraint was assessed in the AM. Rats were perfused at 2h after restraint to assess the effect of loss of MC4R on stress-induced c-Fos immunolabeling in stress-regulatory brain regions. We find that basal (non-stress) AM and PM plasma ACTH and corticosterone showed a normal diurnal rhythm that was not altered according to genotype. Consistent with this, adrenal and thymus weights were unaffected by genotype. However, the plasma ACTH and corticosterone responses to restraint were significantly reduced by loss of MC4R function. Likewise, stress-induced c-Fos immunolabeling in both PVH and MeA was significantly reduced by loss of Mc4r function. These results support the hypothesis that endogenous MC4R signaling contributes to the HPA axis response to stress. Because MC4R plays a critical role in the regulation of energy balance, the present work suggests that it may also serve as an important communication link between brain metabolic and stress systems.
Collapse
Affiliation(s)
- Karen K. Ryan
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati, Cincinnati OH, USA
| | - Joram D. Mul
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati, Cincinnati OH, USA
| | - Christoffer Clemmensen
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati, Cincinnati OH, USA,Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Ann E. Egan
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati OH, USA
| | - Denovan P. Begg
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati OH, USA
| | - Kristen Halcomb
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati OH, USA
| | - Randy J. Seeley
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati, Cincinnati OH, USA
| | - James P. Herman
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati OH, USA
| | - Yvonne M. Ulrich-Lai
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati OH, USA
| |
Collapse
|
50
|
Abstract
PURPOSE OF THE REVIEW Although rodent models provide insight into the mechanisms underlying type 2 diabetes mellitus (T2DM), they are limited in their translatability to humans. The nonhuman primate (NHP) shares important metabolic similarities with the human, making it an ideal model for the investigation of type 2 diabetes and use in preclinical trials. This review highlights the key contributions in the field over the last year using the NHP model. RECENT FINDINGS The NHP has not only provided novel insight into the normal and pathological processes that occur within the islet, but has also allowed for the preclinical testing of novel pharmaceutical targets for obesity and T2DM. Particularly, administration of fibroblast growth factor-21 in the NHP resulted in weight loss and improvements in metabolic health, supporting rodent studies and recent clinical trials. In addition, the NHP was used to demonstrate that a novel melanocortin-4 receptor agonist did not cause adverse cardiovascular effects. Finally, this model has been used to provide evidence that glucagon-like peptide-1-based therapies do not induce pancreatitis in the healthy NHP. SUMMARY The insight gained from studies using the NHP model has allowed for a better understanding of the processes driving T2DM and has promoted the development of well tolerated and effective treatments.
Collapse
Affiliation(s)
- Lynley D Pound
- aDivision of Diabetes, Obesity, & Metabolism bDivision of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | | | | |
Collapse
|