1
|
Camon C, Prescott M, Neyt C, Decourt C, Stout MB, Campbell RE, Garratt M. Systemic metabolic benefits of 17α-estradiol are not exclusively mediated by ERα in glutamatergic or GABAergic neurons. GeroScience 2024; 46:6127-6140. [PMID: 38776045 PMCID: PMC11493872 DOI: 10.1007/s11357-024-01192-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 05/02/2024] [Indexed: 10/23/2024] Open
Abstract
17α-Estradiol (17αE2), a less-feminising enantiomer of 17β-estradiol, has been shown to prolong lifespan and improve metabolic health in a sex-specific manner in male, but not in female mice. Recent studies have demonstrated the pivotal role of estrogen receptor α (ERα) in mediating the effects of 17αE2 on metabolic health. However, the specific tissues and/or neuronal signalling pathways that 17αE2 acts through remain to be elucidated. ERα expression in glutamatergic and GABAergic neurons (principal excitatory and inhibitory neurons respectively) in the hypothalamus is essential for estradiol signalling. Therefore, we hypothesised that knocking out ERα from one of these neuronal populations would attenuate the established beneficial metabolic effects of 17αE2 in male mice exposed to a high fat diet. To test this hypothesis we used two established brain specific ERα KO models, targeting either glutamatergic or GABAergic neurons (Vglut2/Vgat-ERαKO). We show that both of these ERα KO models exhibit a strong reduction in ERα expression in the arcuate nucleus of the hypothalamus, a control centre for metabolic regulation. Deletion of ERα from GABAergic neurons significantly diminished the effect of 17αE2 on body weight relative to controls, although these animals still show metabolic benefits with 17αE2 treatment. The response to 17αE2 was unaffected by ERα deletion in glutamatergic neurons. Our results support a benefit of 17αE2 treatment in protection against metabolic dysfunction, but these effects do not depend on exclusive ERα expression in glutamatergic and GABAergic neurons and persist when ERα expression is strongly reduced in the arcuate nucleus of the hypothalamus.
Collapse
Affiliation(s)
- Celine Camon
- Department of Anatomy, University of Otago, Dunedin, New Zealand.
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand.
| | - Mel Prescott
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Christine Neyt
- Department of Anatomy, University of Otago, Dunedin, New Zealand
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - Caroline Decourt
- Department of Anatomy, University of Otago, Dunedin, New Zealand
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Rebecca E Campbell
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Michael Garratt
- Department of Anatomy, University of Otago, Dunedin, New Zealand
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
2
|
McCarthy SF, Bornath DPD, Tucker JAL, Cohen TR, Medeiros PJ, Hazell TJ. Greater lactate accumulation does not alter peripheral concentrations of key appetite-regulating neuropeptides. J Appl Physiol (1985) 2024; 137:1397-1408. [PMID: 39359185 DOI: 10.1152/japplphysiol.00559.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
The potential mechanisms involved in lactate's role in exercise-induced appetite suppression require further examination. We used sodium bicarbonate (NaHCO3) supplementation in a double-blind, placebo-controlled, randomized crossover design to explore lactate's role on neuropeptide Y (NPY), agouti-related peptide (AgRP), and alpha-melanocyte-stimulating hormone (α-MSH) concentrations. Twelve adults (7 males; 24.2 ± 3.4 kg·m-2; 42.18 ± 8.56 mL·kg-1·min-1) completed two identical high-intensity interval training sessions following ingestion of NaHCO3 (BICARB) or sodium chloride (PLACEBO) pre-exercise. Blood lactate, acylated ghrelin, NPY, AgRP, α-MSH, and appetite perceptions were measured pre-exercise, 0-, 30-, 60-, and 90-min postexercise. Free-living energy intake (electronic food diaries) was measured the day before, of, and after each experimental session. In BICARB, blood lactate was greater postexercise (P < 0.002, d > 0.70), though acylated ghrelin was similar (P = 0.075, [Formula: see text] = 0.206) at all time points postexercise (P > 0.034, d < 0.22). NPY (P = 0.006, [Formula: see text] > 0.509) and AgRP (P < 0.001, [Formula: see text] > 0.488) had main effects of time increasing following exercise and returning to baseline, with no differences between sessions (NPY: P = 0.0.192, [Formula: see text] = 0.149; AgRP: P = 0.422, [Formula: see text] = 0.060). α-MSH had no main effect of time (P = 0.573, [Formula: see text] = 0.063) or session (P = 0.269, [Formula: see text] = 0.110). Appetite perceptions were similar during BICARB and PLACEBO (P = 0.007, d = 0.28), increasing in both sessions postexercise (P < 0.088, d > 0.57). Energy intake had a main effect of day (P = 0.025, [Formula: see text] = 0.825), where the experimental session day was greater than the day before (P = 0.010, d = 0.59) with no other differences between days (P > 0.260, d < 0.38). The lower accumulation of lactate than our previous work did not generate exercise-induced appetite suppression as there were no differences in acylated ghrelin, appetite perceptions, or peripheral concentrations of neuropeptides.NEW & NOTEWORTHY Current evidence supports lactate's role in exercise-induced appetite suppression. Here, we demonstrate a smaller degree of lactate accumulation with sodium bicarbonate ingestion and HIIT than our previous work and no subsequent suppression of acylated ghrelin concentrations, subjective appetite perceptions, or peripheral concentrations of neuropeptides. These results suggest either changes in central appetite-regulating neuropeptides are not reflected peripherally or the smaller magnitude of lactate accumulation did not generate exercise-induced appetite suppression as seen previously.
Collapse
Affiliation(s)
- Seth F McCarthy
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, Waterloo, Ontario, Canada
| | - Derek P D Bornath
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, Waterloo, Ontario, Canada
| | - Jessica A L Tucker
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, Waterloo, Ontario, Canada
| | - Tamara R Cohen
- Faculty of Land and Food Systems, University of British Columbia, Vancouver, British Columbia, Canada
| | - Philip J Medeiros
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, Waterloo, Ontario, Canada
| | - Tom J Hazell
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, Waterloo, Ontario, Canada
| |
Collapse
|
3
|
Narukawa M, Saito Y, Kasahara Y, Asakura T, Misaka T. Changes in gene expression due to aging in the hypothalamus of mice. Neuroreport 2024:00001756-990000000-00282. [PMID: 39166393 PMCID: PMC11389885 DOI: 10.1097/wnr.0000000000002092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Aging generally affects food consumption and energy metabolism. Since the feeding center is located in the hypothalamus, it is a major target for understanding the mechanism of age-related changes in eating behavior and metabolism. To obtain insight into the age-related changes in gene expression in the hypothalamus, we investigated genes whose expression changes with age in the hypothalamus. A DNA microanalysis was performed using hypothalamus samples obtained from young (aged 24 weeks) and old male mice (aged 138 weeks). Gene Ontology (GO) analysis was performed using the identified differentially expressed genes. We observed that the expression of 377 probe sets was significantly altered with aging (177 were upregulated and 200 were downregulated in old mice). As a result of the GO analysis of these probe sets, 16 GO terms, including the neuropeptide signaling pathway, were obtained. Intriguingly, although the food intake in old mice was lower than that in young mice, we found that several neuropeptide genes, such as agouti-related neuropeptide (Agrp), neuropeptide Y (Npy), and pro-melanin-concentrating hormone (Pmch), all of which promote food intake, were upregulated in old mice. In conclusion, this suggests that the gene expression pattern in the hypothalamus is regulated to promote food intake.
Collapse
Affiliation(s)
- Masataka Narukawa
- Department of Food and Nutrition, Kyoto Women's University, Kyoto
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yoshikazu Saito
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Research Department, Toyo Institute of Food Technology, Kawanishi, Hyogo
| | - Yoichi Kasahara
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tomiko Asakura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Department of Liberal Arts, The Open University of Japan, Chiba, Chiba, Japan
| | - Takumi Misaka
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
4
|
Mahdavi K, Zendehdel M, Zarei H. The role of central neurotransmitters in appetite regulation of broilers and layers: similarities and differences. Vet Res Commun 2024; 48:1313-1328. [PMID: 38286893 DOI: 10.1007/s11259-024-10312-4] [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: 09/22/2023] [Accepted: 01/18/2024] [Indexed: 01/31/2024]
Abstract
The importance of feeding as a vital physiological function, on the one hand, and the spread of complications induced by its disorder in humans and animals, on the other hand, have led to extensive research on its regulatory factors. Unfortunately, despite many studies focused on appetite, only limited experiments have been conducted on avian, and our knowledge of this species is scant. Considering this, the purpose of this review article is to examine the role of central neurotransmitters in regulating food consumption in broilers and layers and highlight the similarities and differences between these two strains. The methodology of this review study includes a comprehensive search of relevant literature on the topic using appropriate keywords in reliable electronic databases. Based on the findings, the central effect of most neurotransmitters on the feeding of broilers and laying chickens was similar, but in some cases, such as dopamine, ghrelin, nitric oxide, and agouti-related peptide, differences were observed. Also, the lack of conducting a study on the role of some neurotransmitters in one of the bird strains made it impossible to make an exact comparison. Finally, it seems that although there are general similarities in appetite regulatory mechanisms in meat and egg-type chickens, the long-term genetic selection appropriate to breeding goals (meat or egg production) has caused differences in the effect of some neurotransmitters. Undoubtedly, conducting future studies while completing the missing links can lead to a comprehensive understanding of this process and its manipulation according to the breeding purposes of chickens.
Collapse
Affiliation(s)
- Kimia Mahdavi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, 14155-6453, Iran
| | - Morteza Zendehdel
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, 14155-6453, Iran.
| | - Hamed Zarei
- Department of Biology, Faculty of Basic Science, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
5
|
Murthy VL, Mosley JD, Perry AS, Jacobs DR, Tanriverdi K, Zhao S, Sawicki KT, Carnethon M, Wilkins JT, Nayor M, Das S, Abel ED, Freedman JE, Clish CB, Shah RV. Metabolic liability for weight gain in early adulthood. Cell Rep Med 2024; 5:101548. [PMID: 38703763 PMCID: PMC11148768 DOI: 10.1016/j.xcrm.2024.101548] [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: 11/16/2022] [Revised: 03/27/2023] [Accepted: 04/10/2024] [Indexed: 05/06/2024]
Abstract
While weight gain is associated with a host of chronic illnesses, efforts in obesity have relied on single "snapshots" of body mass index (BMI) to guide genetic and molecular discovery. Here, we study >2,000 young adults with metabolomics and proteomics to identify a metabolic liability to weight gain in early adulthood. Using longitudinal regression and penalized regression, we identify a metabolic signature for weight liability, associated with a 2.6% (2.0%-3.2%, p = 7.5 × 10-19) gain in BMI over ≈20 years per SD higher score, after comprehensive adjustment. Identified molecules specified mechanisms of weight gain, including hunger and appetite regulation, energy expenditure, gut microbial metabolism, and host interaction with external exposure. Integration of longitudinal and concurrent measures in regression with Mendelian randomization highlights the complexity of metabolic regulation of weight gain, suggesting caution in interpretation of epidemiologic or genetic effect estimates traditionally used in metabolic research.
Collapse
Affiliation(s)
- Venkatesh L Murthy
- Division of Cardiovascular Medicine, Department of Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Jonathan D Mosley
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Andrew S Perry
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - David R Jacobs
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Kahraman Tanriverdi
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Shilin Zhao
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | | | - Matthew Nayor
- Section of Cardiovascular Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Saumya Das
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jane E Freedman
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Clary B Clish
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Ravi V Shah
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
6
|
Zhao JV, Yao M, Liu Z. Using genetics and proteomics data to identify proteins causally related to COVID-19, healthspan and lifespan: a Mendelian randomization study. Aging (Albany NY) 2024; 16:6384-6416. [PMID: 38575325 PMCID: PMC11042960 DOI: 10.18632/aging.205711] [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: 08/30/2023] [Accepted: 01/24/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND COVID-19 pandemic poses a heavy burden on public health and accounts for substantial mortality and morbidity. Proteins are building blocks of life, but specific proteins causally related to COVID-19, healthspan and lifespan have not been systematically examined. METHODS We conducted a Mendelian randomization study to assess the effects of 1,361 plasma proteins on COVID-19, healthspan and lifespan, using large GWAS of severe COVID-19 (up to 13,769 cases and 1,072,442 controls), COVID-19 hospitalization (32,519 cases and 2,062,805 controls) and SARS-COV2 infection (122,616 cases and 2,475,240 controls), healthspan (n = 300,477) and parental lifespan (~0.8 million of European ancestry). RESULTS We identified 35, 43, and 63 proteins for severe COVID, COVID-19 hospitalization, and SARS-COV2 infection, and 4, 32, and 19 proteins for healthspan, father's attained age, and mother's attained age. In addition to some proteins reported previously, such as SFTPD related to severe COVID-19, we identified novel proteins involved in inflammation and immunity (such as ICAM-2 and ICAM-5 which affect COVID-19 risk, CXCL9, HLA-DRA and LILRB4 for healthspan and lifespan), apoptosis (such as FGFR2 and ERBB4 which affect COVID-19 risk and FOXO3 which affect lifespan) and metabolism (such as PCSK9 which lowers lifespan). We found 2, 2 and 3 proteins shared between COVID-19 and healthspan/lifespan, such as CXADR and LEFTY2, shared between severe COVID-19 and healthspan/lifespan. Three proteins affecting COVID-19 and seven proteins affecting healthspan/lifespan are targeted by existing drugs. CONCLUSIONS Our study provided novel insights into protein targets affecting COVID-19, healthspan and lifespan, with implications for developing new treatment and drug repurposing.
Collapse
Affiliation(s)
- Jie V. Zhao
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| | - Minhao Yao
- Department of Statistics and Actuarial Science, The University of Hong Kong, Hong Kong SAR, China
| | - Zhonghua Liu
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| |
Collapse
|
7
|
Carver JJ, Lau KM, Puckett AE, Didonna A. Autoimmune demyelination alters hypothalamic transcriptome and endocrine function. J Neuroinflammation 2024; 21:12. [PMID: 38178091 PMCID: PMC10768476 DOI: 10.1186/s12974-023-03006-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 12/26/2023] [Indexed: 01/06/2024] Open
Abstract
The hypothalamus is a brain structure that is deputed to maintain organism homeostasis by regulating autonomic function and hormonal production as part of the neuroendocrine system. Dysfunction in hypothalamic activity results in behavioral alterations, depression, metabolic syndromes, fatigue, and infertility. Remarkably, many of these symptoms are associated with multiple sclerosis (MS), a chronic autoimmune disorder of the central nervous system (CNS) characterized by focal demyelination, immune cell infiltration into the brain parenchyma, and neurodegeneration. Furthermore, altered hormonal levels have been documented in MS patients, suggesting the putative involvement of hypothalamic deficits in MS clinical manifestations. Yet, a systematic analysis of hypothalamic function in response to neuroinflammatory stress is still lacking. To fill this gap, here we performed a longitudinal profiling of the hypothalamic transcriptome upon experimental autoimmune encephalomyelitis (EAE)-a murine disease model recapitulating key MS phenotypes at both histopathological and molecular levels. We show that changes in gene expression connected with an anti-inflammatory response start already at pre-onset and persist along EAE progression. Altered levels of hypothalamic neuropeptides were also detected, which possibly underlie homeostatic responses to stress and aberrant feeding behaviors. Last, a thorough investigation of the principal endocrine glands highlighted defects in the main steroidogenic pathways upon disease. Collectively, our findings corroborate the central role of hypothalamic dysfunction in CNS autoimmunity.
Collapse
Affiliation(s)
- Jonathan J Carver
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, 600 Moye Blvd., Greenville, NC, USA
| | - Kristy M Lau
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, 600 Moye Blvd., Greenville, NC, USA
| | - Alexandra E Puckett
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, 600 Moye Blvd., Greenville, NC, USA
| | - Alessandro Didonna
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, 600 Moye Blvd., Greenville, NC, USA.
| |
Collapse
|
8
|
Patel B, Koysombat K, Mills EG, Tsoutsouki J, Comninos AN, Abbara A, Dhillo WS. The Emerging Therapeutic Potential of Kisspeptin and Neurokinin B. Endocr Rev 2024; 45:30-68. [PMID: 37467734 PMCID: PMC10765167 DOI: 10.1210/endrev/bnad023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/13/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023]
Abstract
Kisspeptin (KP) and neurokinin B (NKB) are neuropeptides that govern the reproductive endocrine axis through regulating hypothalamic gonadotropin-releasing hormone (GnRH) neuronal activity and pulsatile GnRH secretion. Their critical role in reproductive health was first identified after inactivating variants in genes encoding for KP or NKB signaling were shown to result in congenital hypogonadotropic hypogonadism and a failure of pubertal development. Over the past 2 decades since their discovery, a wealth of evidence from both basic and translational research has laid the foundation for potential therapeutic applications. Beyond KP's function in the hypothalamus, it is also expressed in the placenta, liver, pancreas, adipose tissue, bone, and limbic regions, giving rise to several avenues of research for use in the diagnosis and treatment of pregnancy, metabolic, liver, bone, and behavioral disorders. The role played by NKB in stimulating the hypothalamic thermoregulatory center to mediate menopausal hot flashes has led to the development of medications that antagonize its action as a novel nonsteroidal therapeutic agent for this indication. Furthermore, the ability of NKB antagonism to partially suppress (but not abolish) the reproductive endocrine axis has supported its potential use for the treatment of various reproductive disorders including polycystic ovary syndrome, uterine fibroids, and endometriosis. This review will provide a comprehensive up-to-date overview of the preclinical and clinical data that have paved the way for the development of diagnostic and therapeutic applications of KP and NKB.
Collapse
Affiliation(s)
- Bijal Patel
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
| | - Kanyada Koysombat
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Edouard G Mills
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Jovanna Tsoutsouki
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
| | - Alexander N Comninos
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Ali Abbara
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Waljit S Dhillo
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| |
Collapse
|
9
|
Sweeney P, Gimenez LE, Hernandez CC, Cone RD. Targeting the central melanocortin system for the treatment of metabolic disorders. Nat Rev Endocrinol 2023; 19:507-519. [PMID: 37365323 DOI: 10.1038/s41574-023-00855-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/18/2023] [Indexed: 06/28/2023]
Abstract
A large body of preclinical and clinical data shows that the central melanocortin system is a promising therapeutic target for treating various metabolic disorders such as obesity and cachexia, as well as anorexia nervosa. Setmelanotide, which functions by engaging the central melanocortin circuitry, was approved by the FDA in 2020 for use in certain forms of syndromic obesity. Furthermore, the FDA approvals in 2019 of two peptide drugs targeting melanocortin receptors for the treatment of generalized hypoactive sexual desire disorder (bremelanotide) and erythropoietic protoporphyria-associated phototoxicity (afamelanotide) demonstrate the safety of this class of peptides. These approvals have also renewed excitement in the development of therapeutics targeting the melanocortin system. Here, we review the anatomy and function of the melanocortin system, discuss progress and challenges in developing melanocortin receptor-based therapeutics, and outline potential metabolic and behavioural disorders that could be addressed using pharmacological agents targeting these receptors.
Collapse
Affiliation(s)
- Patrick Sweeney
- School of Molecular and Cellular Biology, College of Liberal Arts and Sciences, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Luis E Gimenez
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | | | - Roger D Cone
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular and Integrative Physiology, School of Medicine, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular, Cellular, and Developmental Biology, College of Literature Science and the Arts, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
10
|
Tajima Y, Ito K, Yuan Y, Frank MO, Saito Y, Darnell RB. NOVA1 acts on Impact to regulate hypothalamic function and translation in inhibitory neurons. Cell Rep 2023; 42:112050. [PMID: 36716149 PMCID: PMC10382602 DOI: 10.1016/j.celrep.2023.112050] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/12/2022] [Accepted: 01/15/2023] [Indexed: 01/30/2023] Open
Abstract
We describe a patient haploinsufficient for the neuronal RNA binding protein NOVA1 who developed a behavioral motor hyperactivity disorder, suggesting a role of NOVA1 in postnatal motor inhibition. To investigate Nova1's action in adult Gad2+ inhibitory neurons, we generated a conditional Nova1-null mouse (Nova1-cKOGad2-cre). Strikingly, the phenotypes of these mice show many similarities to the NOVA1 haploinsufficient patient and identify a function of Nova1 in the hypothalamus. Molecularly, Nova1 loss in Gad2-positive neurons alters downstream expression of Impact mRNA, along with a subset of RNAs encoding electron transport chain-related factors and ribosomal proteins. NOVA1 stabilizes Impact mRNA by binding its 3' UTR, antagonizing the actions of miR-138 and miR-124. Together, these studies demonstrate actions of NOVA1 in adult hypothalamic neurons, mechanisms by which it functions in translation and metabolism, including through direct binding to Impact mRNA, and illuminate its role in human neurologic disease.
Collapse
Affiliation(s)
- Yoko Tajima
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Keiichi Ito
- Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Yuan Yuan
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Mayu O Frank
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Yuhki Saito
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Robert B Darnell
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA; Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
11
|
Gürbüzer N, Ceyhun HA, Öztürk N, Kasali K. The Relationship Between Eating-Attitudes and Clinical Characteristics, Agouti-Related Peptide, and Other Biochemical Markers in Adult-Attention Deficit Hyperactivity Disorder. J Atten Disord 2023; 27:394-409. [PMID: 36642920 DOI: 10.1177/10870547221149198] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE In our study, we aimed to evaluate eating-attitudes in adult-ADHD, and to examine its relationship with sociodemographic, clinical, AgRP, and biochemical parameters. METHOD The study included 70 adult-patients and 47 healthy-controls. The DIVA2.0, SCID-1 was administered to the participants. Eating-Attitudes Test (EAT), Night-Eating Questionnaire (NEQ), Barratt Impulsivity Scale (BIS-11) were filled by the participants. RESULTS We found that psychological state affect eating-attitudes in adult-ADHD (p = .013), emotional eating is more common, nocturnal chronotype is dominant (p < .001), NES is more frequent (p < .001), waist circumference measurement is higher (p = .030), and lipid profile is deteriorated (p < .001). AgRP levels were significantly lower in patients treated with methylphenidate (p = .021). Those who received methylphenidate treatment had less NES than those who did not. Deterioration in eating-attitudes and symptom severity of night eating in ADHD, it was positively correlated with clinical severity of ADHD and impulsivity. In addition, age and increase in night eating symptoms were predictors of deterioration in eating attitudes in adult-ADHD. We found that impaired eating-attitudes and impulsivity severity were also predictors of NES (p = .006, p = .034). CONCLUSION The necessity of adult-ADHD treatment has been demonstrated by the deterioration in eating-attitudes and cardiometabolic risk dimensions and the underlying mechanisms.
Collapse
|
12
|
Effect of Obesity and High-Density Lipoprotein Concentration on the Pathological Characteristics of Alzheimer's Disease in High-Fat Diet-Fed Mice. Int J Mol Sci 2022; 23:ijms232012296. [PMID: 36293147 PMCID: PMC9603479 DOI: 10.3390/ijms232012296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 12/05/2022] Open
Abstract
The typical pathological features of Alzheimer's disease (AD) are the accumulation of amyloid plaques in the brain and reactivity of glial cells such as astrocytes and microglia. Clinically, the development of AD and obesity are known to be correlated. In this study, we analyzed the changes in AD pathological characteristics in 5XFAD mice after obesity induction through a high-fat diet (HFD). Surprisingly, high-density lipoprotein and apolipoprotein AI (APOA-I) serum levels were increased without low-density lipoprotein alteration in both HFD groups. The reactivity of astrocytes and microglia in the dentate gyrus of the hippocampus and fornix of the hypothalamus in 5XFAD mice was decreased in the transgenic (TG)-HFD high group. Finally, the accumulation of amyloid plaques in the dentate gyrus region of the hippocampus was also significantly decreased in the TG-HFD high group. These results suggest that increased high-density lipoprotein level, especially with increased APOA-I serum level, alleviates the pathological features of AD and could be a new potential therapeutic strategy for AD treatment.
Collapse
|
13
|
Signaling pathways in obesity: mechanisms and therapeutic interventions. Signal Transduct Target Ther 2022; 7:298. [PMID: 36031641 PMCID: PMC9420733 DOI: 10.1038/s41392-022-01149-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/26/2022] [Accepted: 08/08/2022] [Indexed: 12/19/2022] Open
Abstract
Obesity is a complex, chronic disease and global public health challenge. Characterized by excessive fat accumulation in the body, obesity sharply increases the risk of several diseases, such as type 2 diabetes, cardiovascular disease, and nonalcoholic fatty liver disease, and is linked to lower life expectancy. Although lifestyle intervention (diet and exercise) has remarkable effects on weight management, achieving long-term success at weight loss is extremely challenging, and the prevalence of obesity continues to rise worldwide. Over the past decades, the pathophysiology of obesity has been extensively investigated, and an increasing number of signal transduction pathways have been implicated in obesity, making it possible to fight obesity in a more effective and precise way. In this review, we summarize recent advances in the pathogenesis of obesity from both experimental and clinical studies, focusing on signaling pathways and their roles in the regulation of food intake, glucose homeostasis, adipogenesis, thermogenesis, and chronic inflammation. We also discuss the current anti-obesity drugs, as well as weight loss compounds in clinical trials, that target these signals. The evolving knowledge of signaling transduction may shed light on the future direction of obesity research, as we move into a new era of precision medicine.
Collapse
|
14
|
Feng Y, Xu D, Cai X, Xu M, Garbacz WG, Ren S, Jurczak MJ, Yu C, Wang H, Xie W. Gestational Diabetes Sensitizes Mice to Future Metabolic Syndrome That Can Be Relieved by Activating CAR. Endocrinology 2022; 163:6582264. [PMID: 35524740 DOI: 10.1210/endocr/bqac061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Indexed: 11/19/2022]
Abstract
Diabetes and related metabolic syndrome are common metabolic disorders. Gestational diabetes mellitus (GDM) is rather prevalent in the clinic. Although most GDM resolves after therapeutic intervention and/or after delivery, the long-term health effect of GDM remains to be better understood. The constitutive androstane receptor (CAR), initially characterized as a xenobiotic receptor, was more recently proposed to be a therapeutic target for obesity and type 2 diabetes mellitus (T2DM). In this study, high-fat diet (HFD) feeding was used to induce GDM. Upon delivery, GDM mice were returned to chow diet until the metabolic parameters were normalized. Parous non-GDM control females or metabolically normalized GDM females were then subjected to HFD feeding to induce nongestational obesity and T2DM. Our results showed that GDM sensitized mice to metabolic abnormalities induced by a second hit of HFD. Treatment with the CAR agonist 1,4-bis [2-(3,5 dichloropyridyloxy)] benzene efficiently attenuated GDM-sensitized and HFD-induced obesity and T2DM, including decreased body weight, improved insulin sensitivity, inhibition of hyperglycemia and hepatic steatosis, increased oxygen consumption, and decreased adipocyte hypertrophy. In conclusion, our results have established GDM as a key risk factor for the future development of metabolic disease. We also propose that CAR is a therapeutic target for the management of metabolic disease sensitized by GDM.
Collapse
Affiliation(s)
- Ye Feng
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, 15261 USA
- Department of Endocrinology and Metabolic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China
| | - Dan Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, 15261 USA
- Department of pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Xinran Cai
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Meishu Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Wojciech G Garbacz
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Songrong Ren
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Michael J Jurczak
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Chaohui Yu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Hui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, 15261 USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
15
|
Mardones L, Parra-Valencia E, Petermann-Rocha F, Martínez-Sanguinetti MA, Leiva-Ordoñez AM, Lasserre-Laso N, Martorell M, Ulloa N, Sanhueza E, Pérez-Bravo F, Celis-Morales C, Villagrán M. The rs483145 polymorphism of MC4R gene is not associated with obesity in the Chilean population: Results of GENADIO study. ENDOCRINOL DIAB NUTR 2022; 69:254-261. [PMID: 35570141 DOI: 10.1016/j.endien.2022.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/04/2021] [Accepted: 06/05/2021] [Indexed: 06/15/2023]
Abstract
INTRODUCTION The melanocortin receptor 4 (MC4R) participates in the control of appetite at the level of the central nervous system, through the leptin-melanocortin pathway. An association between different polymorphisms of the MC4R gene and obesity has been reported. However, there are few studies of the rs483145 single nucleotide polymorphism (SNP) of this gene. OBJECTIVE To investigate its prevalence and association with adiposity markers in Chilean adults. METHODS The prevalence of SNP rs483145, of the MC4R gene, was determined in 259 participants of the GENADIO study (genes, environment, diabetes and obesity) by means of real-time polymerase chain reaction (PCR). The association between the risk allele of MC4R (A) and adiposity markers (body weight, body mass index, fat mass percentage, hip circumference, waist circumference, waist-to-hip ratio) was performed by linear regression analysis and adjusted for confusion variables (socio-demographic and physic activity) using three statistical models. RESULTS It was determined that the prevalence of the risk allele (A) of the SNP rs483145 of the MC4R gene is 24.5% in the Chilean adult population included in this study, without finding an association with any of the adiposity markers studied, both in adjusted and unadjusted models. CONCLUSION The presence of the risk allele of SNP rs483145 of the MC4R gene is not associated with adiposity markers in the Chilean adult population studied. New studies with a bigger sample size will be necessary to confirm these results.
Collapse
Affiliation(s)
- Lorena Mardones
- Laboratorio de Investigación en Ciencias Biomédicas, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Esteban Parra-Valencia
- Departamento de Ciencias Clínicas y Pre-Clínicas, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Fanny Petermann-Rocha
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK; Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | | | - Ana María Leiva-Ordoñez
- Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Nicole Lasserre-Laso
- Escuela de Nutrición y Dietética, Facultad de Salud, Universidad Santo Tomas, Región Metropolitana, Chile
| | - Miquel Martorell
- Centro de Vida Saludable, Universidad de Concepción, Concepción, Chile; Departamento de Nutrición y Dietética, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Natalia Ulloa
- Centro de Vida Saludable, Universidad de Concepción, Concepción, Chile; Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Eduardo Sanhueza
- Laboratorio de Investigación en Ciencias Biomédicas, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Francisco Pérez-Bravo
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
| | - Carlos Celis-Morales
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK; Centro de Investigación en Fisiología del Ejercicio (CIFE), Universidad Mayor, Santiago, Chile; Laboratorio de Rendimiento Humano, Grupo de Estudio en Educación, Actividad Física y Salud (GEEAFyS), Universidad Católica del Maule, Talca, Chile
| | - Marcelo Villagrán
- Laboratorio de Investigación en Ciencias Biomédicas, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile.
| |
Collapse
|
16
|
Effect of disrupted episodic memory on food consumption: no impact of neuronal loss of Endophilin A1 on food intake and energy balance. J Genet Genomics 2022; 49:329-337. [DOI: 10.1016/j.jgg.2022.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/04/2022] [Accepted: 01/20/2022] [Indexed: 11/19/2022]
|
17
|
Zhang J, Chen Y, Yan L, Zhang X, Zheng X, Qi J, Yang F, Li J. EphA3 deficiency in hypothalamus promotes high fat diet-induced obesity in mice. J Biomed Res 2022; 37:179-193. [PMID: 37013864 DOI: 10.7555/jbr.36.20220168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Erythropoietin-producing hepatocellular carcinoma A3 (EphA3) is a member of the largest subfamily of tyrosine kinase receptors-Eph receptors. Previous studies have shown that EphA3 is associated with tissue development. Recently, we have found that the expression of EphA3 is elevated in the hypothalamus of mice with diet-induced obesity (DIO). However, the role of EphA3 in hypothalamic-controlled energy metabolism remains unclear. In the current study, we demonstrated that the deletion of EphA3 in the hypothalamus by CRISPR/Cas9-mediated gene editing promotes obesity in male mice with high-fat diet feeding rather than those with normal chow diet feeding. Moreover, the deletion of hypothalamic EphA3 promotes high-fat DIO by increasing food intake and reducing energy expenditure. Knockdown of EphA3 leads to smaller intracellular vesicles in GT1-7 cells. The current study reveals that hypothalamic EphA3 plays important roles in promoting DIO.
Collapse
Affiliation(s)
- Jubiao Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yang Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Lihong Yan
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xin Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiaoyan Zheng
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Junxia Qi
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Fen Yang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Juxue Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, China
| |
Collapse
|
18
|
Šket R, Kotnik P, Bizjan BJ, Kocen V, Mlinarič M, Tesovnik T, Debeljak M, Battelino T, Kovač J. Heterozygous Genetic Variants in Autosomal Recessive Genes of the Leptin-Melanocortin Signalling Pathway Are Associated With the Development of Childhood Obesity. Front Endocrinol (Lausanne) 2022; 13:832911. [PMID: 35574020 PMCID: PMC9105721 DOI: 10.3389/fendo.2022.832911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/23/2022] [Indexed: 11/29/2022] Open
Abstract
Monogenic obesity is a severe, genetically determined disorder that affects up to 1/1000 newborns. Recent reports on potential new therapeutics and innovative clinical approaches have highlighted the need for early identification of individuals with rare genetic variants that can alter the functioning of the leptin-melanocortin signalling pathway, in order to speed up clinical intervention and reduce the risk of chronic complications. Therefore, next-generation DNA sequencing of central genes in the leptin-melanocortin pathway was performed in 1508 children and adolescents with and without obesity, aged 2-19 years. The recruited cohort comprised approximately 5% of the national paediatric population with obesity. The model-estimated effect size of rare variants in the leptin-melanocortin signalling pathway on longitudinal weight gain between carriers and non-carriers was derived. In total, 21 (1.4%) participants had known disease-causing heterozygous variants (DCVs) in the genes under investigation, and 62 (4.1%) participants were carriers of rare variants of unknown clinical significance (VUS). The estimated frequency of potential genetic variants associated with obesity (including rare VUS) ranged between 1/150 (VUS and DCV) and 1/850 (DCV) and differed significantly between participants with and without obesity. On average, the variants identified would result in approximately 7.6 kg (7.0-12.9 kg at the 95th percentile of body weight) (girls) and 8.4 kg (8.2-14.4 kg) (boys) of additional weight gain in carriers at age 18 years compared with subjects without obesity. In conclusion, children with a genetic predisposition to obesity can be promptly identified and may account for more than 6% of obesity cases. Early identification of genetic variants in the LEPR, PCSK1, POMC, MC3R and MC4R genes could reduce the societal burden and improve the clinical management of early severe childhood obesity and its implementation should be further investigated.
Collapse
Affiliation(s)
- Robert Šket
- Clinical Institute of Special Laboratory Diagnostics, University Children’s Hospital, University Medical Center Ljubljana (UMC), Ljubljana, Slovenia
| | - Primož Kotnik
- Department of Pediatrics Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Center Ljubljana (UMC), Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Barbara Jenko Bizjan
- Clinical Institute of Special Laboratory Diagnostics, University Children’s Hospital, University Medical Center Ljubljana (UMC), Ljubljana, Slovenia
| | - Valentina Kocen
- Clinical Institute of Special Laboratory Diagnostics, University Children’s Hospital, University Medical Center Ljubljana (UMC), Ljubljana, Slovenia
| | - Matej Mlinarič
- Department of Pediatrics Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Center Ljubljana (UMC), Ljubljana, Slovenia
| | - Tine Tesovnik
- Clinical Institute of Special Laboratory Diagnostics, University Children’s Hospital, University Medical Center Ljubljana (UMC), Ljubljana, Slovenia
| | - Maruša Debeljak
- Clinical Institute of Special Laboratory Diagnostics, University Children’s Hospital, University Medical Center Ljubljana (UMC), Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tadej Battelino
- Department of Pediatrics Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Center Ljubljana (UMC), Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jernej Kovač
- Clinical Institute of Special Laboratory Diagnostics, University Children’s Hospital, University Medical Center Ljubljana (UMC), Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Jernej Kovač,
| |
Collapse
|
19
|
Mardones L, Parra-Valencia E, Petermann-Rocha F, Martínez-Sanguinetti MA, Leiva-Ordoñez AM, Lasserre-Laso N, Martorell M, Ulloa N, Sanhueza E, Pérez-Bravo F, Celis-Morales C, Villagrán M. The rs483145 polymorphism of MC4R gene is not associated with obesity in the Chilean population: Results of GENADIO study. ENDOCRINOL DIAB NUTR 2021; 69:S2530-0164(21)00156-7. [PMID: 34340956 DOI: 10.1016/j.endinu.2021.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/04/2021] [Accepted: 06/05/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION The melanocortin receptor 4 (MC4R) participates in the control of appetite at the level of the central nervous system, through the leptin-melanocortin pathway. An association between different polymorphisms of the MC4R gene and obesity has been reported. However, there are few studies of the rs483145 single nucleotide polymorphism (SNP) of this gene. OBJECTIVE To investigate its prevalence and association with adiposity markers in Chilean adults. METHODS The prevalence of SNP rs483145, of the MC4R gene, was determined in 259 participants of the GENADIO study (genes, environment, diabetes and obesity) by means of real-time polymerase chain reaction (PCR). The association between the risk allele of MC4R (A) and adiposity markers (body weight, body mass index, fat mass percentage, hip circumference, waist circumference, waist-to-hip ratio) was performed by linear regression analysis and adjusted for confusion variables (socio-demographic and physic activity) using three statistical models. RESULTS It was determined that the prevalence of the risk allele (A) of the SNP rs483145 of the MC4R gene is 24.5% in the Chilean adult population included in this study, without finding an association with any of the adiposity markers studied, both in adjusted and unadjusted models. CONCLUSION The presence of the risk allele of SNP rs483145 of the MC4R gene is not associated with adiposity markers in the Chilean adult population studied. New studies with a bigger sample size will be necessary to confirm these results.
Collapse
Affiliation(s)
- Lorena Mardones
- Laboratorio de Investigación en Ciencias Biomédicas, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Esteban Parra-Valencia
- Departamento de Ciencias Clínicas y Pre-Clínicas, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Fanny Petermann-Rocha
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK; Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | | | - Ana María Leiva-Ordoñez
- Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Nicole Lasserre-Laso
- Escuela de Nutrición y Dietética, Facultad de Salud, Universidad Santo Tomas, Región Metropolitana, Chile
| | - Miquel Martorell
- Centro de Vida Saludable, Universidad de Concepción, Concepción, Chile; Departamento de Nutrición y Dietética, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Natalia Ulloa
- Centro de Vida Saludable, Universidad de Concepción, Concepción, Chile; Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Eduardo Sanhueza
- Laboratorio de Investigación en Ciencias Biomédicas, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Francisco Pérez-Bravo
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
| | - Carlos Celis-Morales
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK; Centro de Investigación en Fisiología del Ejercicio (CIFE), Universidad Mayor, Santiago, Chile; Laboratorio de Rendimiento Humano, Grupo de Estudio en Educación, Actividad Física y Salud (GEEAFyS), Universidad Católica del Maule, Talca, Chile
| | - Marcelo Villagrán
- Laboratorio de Investigación en Ciencias Biomédicas, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile.
| |
Collapse
|
20
|
The linkage of cell cycle and DNA replication with growth difference in female Chinese tongue sole (Cynoglossus semilaevis): Analysis from transcriptomic study and WGCNA. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 39:100833. [PMID: 33848768 DOI: 10.1016/j.cbd.2021.100833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/19/2021] [Accepted: 04/02/2021] [Indexed: 11/21/2022]
Abstract
In addition to the typical sexual size dimorphism, considerable size differences within the female population of the Chinese tongue sole (Cynoglossus semilaevis) have become a further bottleneck of the improvement of sole aquaculture. To identify the internal mechanism, transcriptomic analysis and weighted gene co-expression network analysis (WGCNA) were employed simultaneously. Transcriptomic analyses of brain, pituitary gland, liver, gonad, and muscle tissues from two female groups with size differences identified 109, 698, 1325, 2299, and 2141 differentially expressed genes (DEGs), respectively. The results of these enrichment analyses suggest that the up-regulation of neuroactive ligand-receptor interaction, cell cycle, DNA replication, and MAPK signaling pathway in the group with larger females may be involved in the regulation of the observed growth differences. WGCNA of DEGs showed that cell cycle and DNA replication might be crucial pathways for accelerating cell growth in the groups with larger females. Finally, a series of hub genes including 6-phosphofructokinase type C (pfkp), ribosome biogenesis protein (wdr12), bleomycin hydrolase (blmh), and semaphorin-3A (sema3a) were recognized by the illustrated network map of modules. The linkage of cell cycle, DNA replication, and hub genes in the growth regulation of C. semilaevis provides further information for a better understanding of growth differences in fish.
Collapse
|
21
|
Alhabeeb H, AlFaiz A, Kutbi E, AlShahrani D, Alsuhail A, AlRajhi S, Alotaibi N, Alotaibi K, AlAmri S, Alghamdi S, AlJohani N. Gut Hormones in Health and Obesity: The Upcoming Role of Short Chain Fatty Acids. Nutrients 2021; 13:nu13020481. [PMID: 33572661 PMCID: PMC7911102 DOI: 10.3390/nu13020481] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 12/21/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
We are currently facing an obesity pandemic, with worldwide obesity rates having tripled since 1975. Obesity is one of the main risk factors for the development of non-communicable diseases, which are now the leading cause of death worldwide. This calls for urgent action towards understanding the underlying mechanisms behind the development of obesity as well as developing more effective treatments and interventions. Appetite is carefully regulated in humans via the interaction between the central nervous system and peripheral hormones. This involves a delicate balance in external stimuli, circulating satiating and appetite stimulating hormones, and correct functioning of neuronal signals. Any changes in this equilibrium can lead to an imbalance in energy intake versus expenditure, which often leads to overeating, and potentially weight gain resulting in overweight or obesity. Several lines of research have shown imbalances in gut hormones are found in those who are overweight or obese, which may be contributing to their condition. Therefore, this review examines the evidence for targeting gut hormones in the treatment of obesity by discussing how their dysregulation influences food intake, the potential possibility of altering the circulating levels of these hormones for treating obesity, as well as the role of short chain fatty acids and protein as novel treatments.
Collapse
Affiliation(s)
- Habeeb Alhabeeb
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
- Correspondence:
| | - Ali AlFaiz
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Emad Kutbi
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Dayel AlShahrani
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Abdullah Alsuhail
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Saleh AlRajhi
- Family Medicine, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia;
| | - Nemer Alotaibi
- College of Medicine, Shaqra University, Shaqra 11961, Saudi Arabia; (N.A.); (K.A.)
| | - Khalid Alotaibi
- College of Medicine, Shaqra University, Shaqra 11961, Saudi Arabia; (N.A.); (K.A.)
| | - Saad AlAmri
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Saleh Alghamdi
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Naji AlJohani
- Obesity, Endocrine, and Metabolism Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia;
| |
Collapse
|
22
|
van Loenen MR, Geenen B, Arnoldussen IAC, Kiliaan AJ. Ghrelin as a prominent endocrine factor in stress-induced obesity. Nutr Neurosci 2020; 25:1413-1424. [PMID: 33373270 DOI: 10.1080/1028415x.2020.1863740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Objectives: Ghrelin acts on a variety of central- and peripheral organs causing an orexigenic effect, conclusively followed by increased caloric intake. Recent studies have indicated that ghrelin's function as an orexigenic agent does not entirely reflect the full functional properties of the peptide. Specifically, ghrelin regulates stress-hormone synthesis and secretion therewith affecting the stress-axis. The role of stress in the development of obesity has been extensively studied. However, the orexigenic and underlying stress-regulatory effect of ghrelin has not yet been further considered in the development of stress-induced obesity.Methods: Therefore, this review aims to accentuate the potential of ghrelin as a factor in the pathological development of stress-induced obesity.Results: In this review we discuss (1) the ghrelin-mediated intracellular cascades and elucidate the overall bioactivation of the peptide, and (2) the mechanisms of ghrelin signalling and regulation within the central nervous system and the gastro-intestinal system.Discussion: These biological processes will be ultimately discussed in relation to the pathogenesis of stress-induced obesity.
Collapse
Affiliation(s)
- Mark R van Loenen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Preclinical Imaging Center PRIME, Nijmegen, Netherlands
| | - Bram Geenen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Preclinical Imaging Center PRIME, Nijmegen, Netherlands
| | - Ilse A C Arnoldussen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Preclinical Imaging Center PRIME, Nijmegen, Netherlands
| | - Amanda J Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Preclinical Imaging Center PRIME, Nijmegen, Netherlands
| |
Collapse
|
23
|
Schellekens H, Torres-Fuentes C, van de Wouw M, Long-Smith CM, Mitchell A, Strain C, Berding K, Bastiaanssen TFS, Rea K, Golubeva AV, Arboleya S, Verpaalen M, Pusceddu MM, Murphy A, Fouhy F, Murphy K, Ross P, Roy BL, Stanton C, Dinan TG, Cryan JF. Bifidobacterium longum counters the effects of obesity: Partial successful translation from rodent to human. EBioMedicine 2020; 63:103176. [PMID: 33349590 PMCID: PMC7838052 DOI: 10.1016/j.ebiom.2020.103176] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/13/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023] Open
Abstract
Background The human gut microbiota has emerged as a key factor in the development of obesity. Certain probiotic strains have shown anti-obesity effects. The objective of this study was to investigate whether Bifidobacterium longum APC1472 has anti-obesity effects in high-fat diet (HFD)-induced obese mice and whether B. longum APC1472 supplementation reduces body-mass index (BMI) in healthy overweight/obese individuals as the primary outcome. B. longum APC1472 effects on waist-to-hip ratio (W/H ratio) and on obesity-associated plasma biomarkers were analysed as secondary outcomes. Methods B. longum APC1472 was administered to HFD-fed C57BL/6 mice in drinking water for 16 weeks. In the human intervention trial, participants received B. longum APC1472 or placebo supplementation for 12 weeks, during which primary and secondary outcomes were measured at the beginning and end of the intervention. Findings B. longum APC1472 supplementation was associated with decreased bodyweight, fat depots accumulation and increased glucose tolerance in HFD-fed mice. While, in healthy overweight/obese adults, the supplementation of B. longum APC1472 strain did not change primary outcomes of BMI (0.03, 95% CI [-0.4, 0.3]) or W/H ratio (0.003, 95% CI [-0.01, 0.01]), a positive effect on the secondary outcome of fasting blood glucose levels was found (-0.299, 95% CI [-0.44, -0.09]). Interpretation This study shows a positive translational effect of B. longum APC1472 on fasting blood glucose from a preclinical mouse model of obesity to a human intervention study in otherwise healthy overweight and obese individuals. This highlights the promising potential of B. longum APC1472 to be developed as a valuable supplement in reducing specific markers of obesity. Funding This research was funded in part by Science Foundation Ireland in the form of a Research Centre grant (SFI/12/RC/2273) to APC Microbiome Ireland and by a research grant from Cremo S.A.
Collapse
Affiliation(s)
- Harriët Schellekens
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | | | | | | | - Avery Mitchell
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Conall Strain
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Kirsten Berding
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Thomaz F S Bastiaanssen
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Kieran Rea
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Anna V Golubeva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Silvia Arboleya
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Mathieu Verpaalen
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | | - Amy Murphy
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Fiona Fouhy
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Kiera Murphy
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland; College of Science Engineering & Food Science, University College Cork, Cork, Ireland
| | | | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Dept of Psychiatry and Behavioural Neuroscience, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
24
|
Madelaine R, Ngo KJ, Skariah G, Mourrain P. Genetic deciphering of the antagonistic activities of the melanin-concentrating hormone and melanocortin pathways in skin pigmentation. PLoS Genet 2020; 16:e1009244. [PMID: 33301440 PMCID: PMC7755275 DOI: 10.1371/journal.pgen.1009244] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/22/2020] [Accepted: 10/30/2020] [Indexed: 01/18/2023] Open
Abstract
The genetic origin of human skin pigmentation remains an open question in biology. Several skin disorders and diseases originate from mutations in conserved pigmentation genes, including albinism, vitiligo, and melanoma. Teleosts possess the capacity to modify their pigmentation to adapt to their environmental background to avoid predators. This background adaptation occurs through melanosome aggregation (white background) or dispersion (black background) in melanocytes. These mechanisms are largely regulated by melanin-concentrating hormone (MCH) and α-melanocyte–stimulating hormone (α-MSH), two hypothalamic neuropeptides also involved in mammalian skin pigmentation. Despite evidence that the exogenous application of MCH peptides induces melanosome aggregation, it is not known if the MCH system is physiologically responsible for background adaptation. In zebrafish, we identify that MCH neurons target the pituitary gland-blood vessel portal and that endogenous MCH peptide expression regulates melanin concentration for background adaptation. We demonstrate that this effect is mediated by MCH receptor 2 (Mchr2) but not Mchr1a/b. mchr2 knock-out fish cannot adapt to a white background, providing the first genetic demonstration that MCH signaling is physiologically required to control skin pigmentation. mchr2 phenotype can be rescued in adult fish by knocking-out pomc, the gene coding for the precursor of α-MSH, demonstrating the relevance of the antagonistic activity between MCH and α-MSH in the control of melanosome organization. Interestingly, MCH receptor is also expressed in human melanocytes, thus a similar antagonistic activity regulating skin pigmentation may be conserved during evolution, and the dysregulation of these pathways is significant to our understanding of human skin disorders and cancers. Melanocytes produce melanin, a natural skin pigment, for body coloration which helps to protect and camouflage an organism and to attract mates. Melanocytes are ubiquitous pigment cells in vertebrates and the genes underlying their development are well conserved, making fishes that possess the ability to modify their pigmentation, biologically relevant and successful models for human skin disorders. Many human skin diseases including albinism, vitiligo, and melanoma are derived from mutations in conserved pigmentation genes. However, much of the conserved molecular mechanisms behind these diseases and human pigmentation remain unknown. For instance, melanin concentrating hormone (MCH) was originally identified as a peptide that when injected, could make fish paler by promoting melanin aggregation but no mutants demonstrating an endogenous function for MCH in pigmentation have been reported. Here, we use zebrafish mutants of MCH and the MCH receptor to determine their specific genetic function in pigmentation. Additionally, we demonstrate that MCH has an antagonistic pigmentation function to the melanocortin system, where MCH expression promotes lighter pigmentation and melanocortin activity promotes darkening. Thus, we find that the balance between the MCH and melanocortin system activities are likely required for skin pigmentation and dysregulation of these pathways could underlie adverse human skin conditions.
Collapse
Affiliation(s)
- Romain Madelaine
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California, United States of America
| | - Keri J. Ngo
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California, United States of America
- Department of Developmental Biology, Stanford University, Stanford, California, United States of America
| | - Gemini Skariah
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California, United States of America
| | - Philippe Mourrain
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California, United States of America
- INSERM 1024, Ecole Normale Supérieure, Paris, France
- * E-mail:
| |
Collapse
|
25
|
Abstract
Since the discovery of functionally competent, energy-consuming brown adipose tissue (BAT) in adult humans, much effort has been devoted to exploring this tissue as a means for increasing energy expenditure to counteract obesity. However, despite promising effects on metabolic rate and insulin sensitivity, no convincing evidence for weight-loss effects of cold-activated human BAT exists to date. Indeed, increasing energy expenditure would naturally induce compensatory feedback mechanisms to defend body weight. Interestingly, BAT is regulated by multiple interactions with the hypothalamus from regions overlapping with centers for feeding behavior and metabolic control. Therefore, in the further exploration of BAT as a potential source of novel drug targets, we discuss the hypothalamic orchestration of BAT activity and the relatively unexplored BAT feedback mechanisms on neuronal regulation. With a holistic view on hypothalamic-BAT interactions, we aim to raise ideas and provide a new perspective on this circuit and highlight its clinical relevance.
Collapse
Affiliation(s)
- Jo B Henningsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark;
| |
Collapse
|
26
|
Yang Y, Xu Y. The central melanocortin system and human obesity. J Mol Cell Biol 2020; 12:785-797. [PMID: 32976556 PMCID: PMC7816681 DOI: 10.1093/jmcb/mjaa048] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/04/2020] [Accepted: 08/14/2020] [Indexed: 12/13/2022] Open
Abstract
The prevalence of obesity and the associated comorbidities highlight the importance of understanding the regulation of energy homeostasis. The central melanocortin system plays a critical role in controlling body weight balance. Melanocortin neurons sense and integrate the neuronal and hormonal signals, and then send regulatory projections, releasing anorexigenic or orexigenic melanocortin neuropeptides, to downstream neurons to regulate the food intake and energy expenditure. This review summarizes the latest progress in our understanding of the role of the melanocortin pathway in energy homeostasis. We also review the advances in the identification of human genetic variants that cause obesity via mechanisms that affect the central melanocortin system, which have provided rational targets for treatment of genetically susceptible patients.
Collapse
Affiliation(s)
- Yongjie Yang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
27
|
Lis M, Stańczykiewicz B, Liśkiewicz P, Misiak B. Impaired hormonal regulation of appetite in schizophrenia: A narrative review dissecting intrinsic mechanisms and the effects of antipsychotics. Psychoneuroendocrinology 2020; 119:104744. [PMID: 32534330 DOI: 10.1016/j.psyneuen.2020.104744] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/25/2020] [Accepted: 05/30/2020] [Indexed: 12/14/2022]
Abstract
Cardiometabolic diseases are the main contributor of reduced life expectancy in patients with schizophrenia. It is now widely accepted that antipsychotic treatment plays an important role in the development of obesity and its consequences. However, some intrinsic mechanisms need to be taken into consideration. One of these mechanisms might be related to impaired hormonal regulation of appetite in this group of patients. In this narrative review, we aimed to dissect impairments of appetite-regulating hormones attributable to intrinsic mechanisms and those related to medication effects. Early hormonal alterations that might be associated with intrinsic mechanisms include low levels of leptin and glucagon-like peptide-1 (GLP-1) together with elevated insulin levels in first-episode psychosis (FEP) patients. However, evidence regarding low GLP-1 levels in FEP patients is based on one large study. In turn, multiple-episode schizophrenia patients show elevated levels of insulin, leptin and orexin A together with decreased levels of adiponectin. In addition, patients receiving olanzapine may present with low ghrelin levels. Post mortem studies have also demonstrated reduced number of neuropeptide Y neurons in the prefrontal cortex of patients with schizophrenia. Treatment with certain second-generation antipsychotics may also point to these alterations. Although our understanding of hormonal regulation of appetite in schizophrenia has largely been improved, several limitations and directions for future studies need to be addressed. This is of particular importance since several novel pharmacological interventions for obesity and diabetes have already been developed and translation of these developments to the treatment of cardiometabolic comorbidities in schizophrenia patients is needed.
Collapse
Affiliation(s)
- Michał Lis
- Clinical Department of Internal Diseases, Endocrinology and Diabetology, The Central Clinical Hospital of the Ministry of the Interior in Warsaw, Wołoska 137 Street, 02-507 Warsaw, Poland
| | - Bartłomiej Stańczykiewicz
- Department of Nervous System Diseases, Wroclaw Medical University, Bartla 5 Street, 51-618, Wroclaw, Poland
| | - Paweł Liśkiewicz
- Department of Psychiatry, Pomeranian Medical University, Broniewskiego 26 Street, 71-460, Szczecin, Poland
| | - Błażej Misiak
- Department of Genetics, Wroclaw Medical University, Marcinkowskiego 1 Street, 50-368 Wroclaw, Poland.
| |
Collapse
|
28
|
Crosstalk of Brain and Bone-Clinical Observations and Their Molecular Bases. Int J Mol Sci 2020; 21:ijms21144946. [PMID: 32668736 PMCID: PMC7404044 DOI: 10.3390/ijms21144946] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/06/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023] Open
Abstract
As brain and bone disorders represent major health issues worldwide, substantial clinical investigations demonstrated a bidirectional crosstalk on several levels, mechanistically linking both apparently unrelated organs. While multiple stress, mood and neurodegenerative brain disorders are associated with osteoporosis, rare genetic skeletal diseases display impaired brain development and function. Along with brain and bone pathologies, particularly trauma events highlight the strong interaction of both organs. This review summarizes clinical and experimental observations reported for the crosstalk of brain and bone, followed by a detailed overview of their molecular bases. While brain-derived molecules affecting bone include central regulators, transmitters of the sympathetic, parasympathetic and sensory nervous system, bone-derived mediators altering brain function are released from bone cells and the bone marrow. Although the main pathways of the brain-bone crosstalk remain ‘efferent’, signaling from brain to bone, this review emphasizes the emergence of bone as a crucial ‘afferent’ regulator of cerebral development, function and pathophysiology. Therefore, unraveling the physiological and pathological bases of brain-bone interactions revealed promising pharmacologic targets and novel treatment strategies promoting concurrent brain and bone recovery.
Collapse
|
29
|
Sutton AK, Krashes MJ. Integrating Hunger with Rival Motivations. Trends Endocrinol Metab 2020; 31:495-507. [PMID: 32387196 DOI: 10.1016/j.tem.2020.04.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/09/2020] [Accepted: 04/13/2020] [Indexed: 12/11/2022]
Abstract
Motivated behaviors have fascinated neuroscientists and ethologists for decades due to their necessity for organism survival. Motivations guide behavioral choice through an intricate synthesis of internal state detection, external stimulus exposure, and learned associations. One critical motivation, hunger, provides an accessible example for understanding purposeful behavior. Neuroscientists commonly focus research efforts on neural circuits underlying individual motivations, sacrificing ethological relevance for tight experimental control. This restrictive focus deprives the field of a more nuanced understanding of the unified nervous system in weighing multiple motivations simultaneously and choosing, moment-to-moment, optimal behaviors for survival. Here, we explore the reciprocal interplay between hunger, encoded via hypothalamic neurons marked by the expression of Agouti-related peptide, and alternative need-based motivational systems.
Collapse
Affiliation(s)
- Amy K Sutton
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael J Krashes
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD 20892, USA; National Institute on Drug Abuse (NIDA), National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
30
|
Koerperich ZM, Ericson MD, Freeman KT, Speth RC, Pogozheva ID, Mosberg HI, Haskell-Luevano C. Incorporation of Agouti-Related Protein (AgRP) Human Single Nucleotide Polymorphisms (SNPs) in the AgRP-Derived Macrocyclic Scaffold c[Pro-Arg-Phe-Phe-Asn-Ala-Phe-dPro] Decreases Melanocortin-4 Receptor Antagonist Potency and Results in the Discovery of Melanocortin-5 Receptor Antagonists. J Med Chem 2020; 63:2194-2208. [PMID: 31845801 DOI: 10.1021/acs.jmedchem.9b00860] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
While the melanocortin receptors (MCRs) are known to be involved in numerous biological pathways, the potential roles of the MC5R have not been clearly elucidated in humans. Agouti-related protein (AgRP), an MC3R/MC4R antagonist and MC4R inverse agonist, contains an exposed β-hairpin loop composed of six residues (Arg-Phe-Phe-Asn-Ala-Phe) that is imperative for binding and function. Within this active loop of AgRP, four human missense polymorphisms were deposited into the NIH Variation Viewer database. These polymorphisms, Arg111Cys, Arg111His, Phe112Tyr, and Ala115Val (AgRP full-length numbering), were incorporated into the peptide macrocycles c[Pro1-Arg2-Phe3-Phe4-Xaa5-Ala6-Phe7-dPro8], where Xaa was Dap5 or Asn5, to explore the functional effects of these naturally occurring substitutions in a simplified AgRP scaffold. All peptides lowered potency at least 10-fold in a cAMP accumulation assay compared to the parent sequences at the MC4Rs. Compounds MDE 6-82-3c, ZMK 2-82, MDE 6-82-1c, ZMK 2-85, and ZMK 2-112 are also the first AgRP-based chemotypes that antagonize the MC5R.
Collapse
Affiliation(s)
- Zoe M Koerperich
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mark D Ericson
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Katie T Freeman
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Robert C Speth
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida 33328-2018, United States.,College of Medicine, Georgetown University, Washington, D.C. 20057, United States
| | - Irina D Pogozheva
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Henry I Mosberg
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Carrie Haskell-Luevano
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
31
|
Mehta R, Lam-Chung CE, Hinojosa-Amaya JM, Roldán-Sarmiento P, Guillen-Placencia MF, Villanueva-Rodriguez G, Juarez-Leon OA, Leon-Domínguez J, Grajales-Gómez M, Ventura-Gallegos JL, León-Suárez A, Gómez-Pérez FJ, Cuevas-Ramos D. High Molecular Weight ACTH-Precursor Presence in a Metastatic Pancreatic Neuroendocrine Tumor Causing Severe Ectopic Cushing's Syndrome: A Case Report. Front Endocrinol (Lausanne) 2020; 11:557. [PMID: 32903471 PMCID: PMC7438413 DOI: 10.3389/fendo.2020.00557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/07/2020] [Indexed: 12/17/2022] Open
Abstract
Ectopic ACTH-secretion causing Cushing's syndrome is unusual and its diagnosis is frequently challenging. The presence of high-molecular-weight precursors throughout pro-opiomelanocortin (POMC) translation by these tumors is often not reported. We present the case of a 49-year-old woman with a 3-month history of proximal muscular weakness, skin pigmentation, and weight loss. Upon initial evaluation, she had a full moon face, hirsutism, and a buffalo hump. Laboratory workup showed hyperglycemia, hypokalemia and metabolic alkalosis. ACTH, plasma cortisol, and urinary free cortisol levels were quite elevated. Serum cortisol levels were not suppressed on dexamethasone suppression testing. An octreo-SPECT scan showed enhanced nucleotide uptake in the liver and pancreas. Transendoscopic ultrasound-guided biopsy confirmed the diagnosis of a pancreatic ACTH-secreting neuroendocrine tumor (NET). Surgical excision of both pancreatic and liver lesions was carried out. Western blot analysis of the tumor and metastases revealed the presence of a high-molecular-weight precursor possibly POMC (at 30 kDa) but not ACTH (normally 4.5 kDa). ACTH-precursor secretion is more frequent in ectopic ACTH-secreting tumors compared with other causes of Cushing's syndrome. Hence, the measurement of such ACTH precursors warrants further evaluation, especially in the context of ACTH-dependent hypercortisolism.
Collapse
Affiliation(s)
- Roopa Mehta
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - César Ernesto Lam-Chung
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José Miguel Hinojosa-Amaya
- Pituitary Clinic, Endocrinology Division, Department of Medicine, Hospital Universitario “Dr. José E. González” UANL. Francisco I. Monterrey, Monterrey, Mexico
| | - Paola Roldán-Sarmiento
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Maria Fernanda Guillen-Placencia
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Gerladine Villanueva-Rodriguez
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Oscar Alfredo Juarez-Leon
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jefsi Leon-Domínguez
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Mariana Grajales-Gómez
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jose Luis Ventura-Gallegos
- Biochemistry Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Mexico City, Mexico
| | - Andrés León-Suárez
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Francisco J. Gómez-Pérez
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Daniel Cuevas-Ramos
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- *Correspondence: Daniel Cuevas-Ramos
| |
Collapse
|
32
|
Mittal P, Jaiswal SK, Vijay N, Saxena R, Sharma VK. Comparative analysis of corrected tiger genome provides clues to its neuronal evolution. Sci Rep 2019; 9:18459. [PMID: 31804567 PMCID: PMC6895189 DOI: 10.1038/s41598-019-54838-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 11/14/2019] [Indexed: 01/01/2023] Open
Abstract
The availability of completed and draft genome assemblies of tiger, leopard, and other felids provides an opportunity to gain comparative insights on their unique evolutionary adaptations. However, genome-wide comparative analyses are susceptible to errors in genome sequences and thus require accurate genome assemblies for reliable evolutionary insights. In this study, while analyzing the tiger genome, we found almost one million erroneous substitutions in the coding and non-coding region of the genome affecting 4,472 genes, hence, biasing the current understanding of tiger evolution. Moreover, these errors produced several misleading observations in previous studies. Thus, to gain insights into the tiger evolution, we corrected the erroneous bases in the genome assembly and gene set of tiger using ‘SeqBug’ approach developed in this study. We sequenced the first Bengal tiger genome and transcriptome from India to validate these corrections. A comprehensive evolutionary analysis was performed using 10,920 orthologs from nine mammalian species including the corrected gene sets of tiger and leopard and using five different methods at three hierarchical levels, i.e. felids, Panthera, and tiger. The unique genetic changes in tiger revealed that the genes showing signatures of adaptation in tiger were enriched in development and neuronal functioning. Specifically, the genes belonging to the Notch signalling pathway, which is among the most conserved pathways involved in embryonic and neuronal development, were found to have significantly diverged in tiger in comparison to the other mammals. Our findings suggest the role of adaptive evolution in neuronal functions and development processes, which correlates well with the presence of exceptional traits such as sensory perception, strong neuro-muscular coordination, and hypercarnivorous behaviour in tiger.
Collapse
Affiliation(s)
- Parul Mittal
- Metaomics and Systems Biology Group, Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India
| | - Shubham K Jaiswal
- Metaomics and Systems Biology Group, Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India
| | - Nagarjun Vijay
- Computational Evolutionary Genomics Lab, Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India
| | - Rituja Saxena
- Metaomics and Systems Biology Group, Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India
| | - Vineet K Sharma
- Metaomics and Systems Biology Group, Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India.
| |
Collapse
|
33
|
Wang J, Li Y, Luo P, Chen Y, Xi Q, Wu H, Zhao W, Shu G, Wang S, Gao P, Zhu X, Zhang Y, Jiang Q, Wang L. Oral supplementation with ginseng polysaccharide promotes food intake in mice. Brain Behav 2019; 9:e01340. [PMID: 31392839 PMCID: PMC6749478 DOI: 10.1002/brb3.1340] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Ginseng polysaccharide (GPS, same as Panax polysaccharide) is a kind of polysaccharide extracted from ginseng. It has been reported that GPS has the ability to activate innate immunity, regulates blood sugar balance, and improves antioxidant capacity, but the effect on feeding behavior and its mechanism remains unclear. METHOD To investigate the possible effect of GPS on feeding behavior of animals, mice were supplied with GPS in water, and food intake, hedonic feeding behavior, anxiety-like behavior, expression of appetite-regulation peptides in the central nervous system and glucose-related hormone levels in the serum of mice were measured. RESULTS Ginseng polysaccharide significantly increased the average daily food intake in mice and promoted hedonic eating behavior. Meanwhile, the levels of serum glucose and glucagon were significantly reduced by GPS, and GPS promoted hypothalamic neuropeptide Y expression, inhibited proopiomelanocortin (POMC) expression, and reduced dopamine D1 receptor (DRD1) levels in the midbrain. We also found that the anxiety level of mice was significantly lower after GPS intake. In conclusion, oral supplementation with GPS promoted food intake in mice, most likely through the regulation of circulating glucose levels.
Collapse
Affiliation(s)
- Jiawen Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Yongxiang Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Pei Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Yuhuang Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Hanyu Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Weijie Zhao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Songbo Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Ping Gao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaotong Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Lina Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
34
|
Rodríguez-Pardo C, Segura A, Zamorano-León JJ, Martínez-Santos C, Martínez D, Collado-Yurrita L, Giner M, García-García JM, Rodríguez-Pardo JM, López-Farre A. Decision tree learning to predict overweight/obesity based on body mass index and gene polymporphisms. Gene 2019; 699:88-93. [DOI: 10.1016/j.gene.2019.03.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 12/26/2022]
|
35
|
Sirohi S, Skripnikova E, Davis JF. Vertical Sleeve Gastrectomy Attenuates Hedonic Feeding Without Impacting Alcohol Drinking in Rats. Obesity (Silver Spring) 2019; 27:603-611. [PMID: 30740914 PMCID: PMC6430654 DOI: 10.1002/oby.22415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/13/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Roux-en-Y gastric bypass surgery and vertical sleeve gastrectomy (VSG) are the most commonly performed bariatric procedures. Whereas studies report new-onset alcohol misuse following Roux-en-Y gastric bypass, the impact of VSG on alcohol intake is less clear. Hedonic feeding, alcohol drinking, and hypothalamic obesity-related gene expression following VSG were evaluated. METHODS Male Long-Evans rats underwent VSG or sham surgery. To evaluate hedonic feeding, rats received a high-fat diet following behavioral satiation on chow. Alcohol (5%-10% v/v) drinking was assessed in a two-bottle choice paradigm. Finally, polymerase chain reaction array evaluated gene expression. RESULTS VSG induced moderate but significant weight loss. Sham rats significantly escalated high-fat diet intake following behavioral satiation, an effect significantly reduced in VSG rats. A moderate decrease in alcohol intake was observed in VSG rats at low (5%) alcohol concentration. However, overall, no significant between-group differences were evident. Key hypothalamic orexigenic transcripts linked to stimulation of food and alcohol intake were significantly decreased in VSG rats. CONCLUSIONS VSG attenuated hedonic feeding without impacting alcohol drinking, an effect potentially mediated by alterations in genetic information flow within the hypothalamus. Importantly, these data highlight VSG as an effective bariatric procedure with a potentially reduced risk of developing alcohol use disorder.
Collapse
Affiliation(s)
- Sunil Sirohi
- Laboratory of Endocrine and Neuropsychiatric Disorders, Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA
- Corresponding Authors: Jon F. Davis, PhD, Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, 1815 Ferdinand’s Lane, Pullman, WA, 99164, Tel (Office): 509-335-8163, , Sunil Sirohi, PhD, Laboratory of Endocrine and Neuropsychiatric Disorders, Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, 1 Drexel Dr., New Orleans, LA 70125, Tel (Office): 504-520-5471; (lab) 504-520-5332, ;
| | - Elena Skripnikova
- Laboratory of Endocrine and Neuropsychiatric Disorders, Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA
| | - Jon F. Davis
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA
- Corresponding Authors: Jon F. Davis, PhD, Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, 1815 Ferdinand’s Lane, Pullman, WA, 99164, Tel (Office): 509-335-8163, , Sunil Sirohi, PhD, Laboratory of Endocrine and Neuropsychiatric Disorders, Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, 1 Drexel Dr., New Orleans, LA 70125, Tel (Office): 504-520-5471; (lab) 504-520-5332, ;
| |
Collapse
|
36
|
Özkorumak Karagüzel E, Kural BV, Tiryaki A, Keleş Altun İ, Özer SY, Civil Arslan F. Blood levels of agouti-related peptide (AgRP), obestatin, corticosteroid-binding globulin (CBG), and cortisol in patients with bipolar disorder (BD): a case–control study. PSYCHIAT CLIN PSYCH 2018. [DOI: 10.1080/24750573.2018.1487649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Affiliation(s)
| | - Birgül Vanizor Kural
- Department of Medical Biochemistry, Karadeniz Technical University, Faculty of Medicine, Trabzon, Turkey
| | - Ahmet Tiryaki
- Department of Psychiatry, İstanbul Aydın University, Faculty of Medicine, İstanbul, Turkey
| | | | - Serap Yaman Özer
- Department of Medical Biochemistry, Karadeniz Technical University, Faculty of Medicine, Trabzon, Turkey
| | - Filiz Civil Arslan
- Department of Psychiatry, Karadeniz Technical University, Faculty of Medicine, Trabzon, Turkey
| |
Collapse
|
37
|
Ericson MD, Haskell-Luevano C. A Review of Single-Nucleotide Polymorphisms in Orexigenic Neuropeptides Targeting G Protein-Coupled Receptors. ACS Chem Neurosci 2018; 9:1235-1246. [PMID: 29714060 DOI: 10.1021/acschemneuro.8b00151] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Many physiological pathways are involved in appetite, food intake, and the maintenance of energy homeostasis. In particular, neuropeptides within the central nervous system have been demonstrated to be critical signaling molecules for modulating appetite. Both anorexigenic (appetite-decreasing) and orexigenic (appetite-stimulating) neuropeptides have been described. The biological effects of these neuropeptides can be observed following central administration in animal models. This review focuses on single nucleotide polymorphisms (SNPs) in six orexigenic neuropeptides: agouti-related protein (AGRP), galanin, melanin concentrating hormone (MCH), neuropeptide Y (NPY), orexin A, and orexin B. Following a brief summary of the neuropeptides and their orexigenic activities, reports associating SNPs within the orexigenic neuropeptides to energy homeostasis, food intake, obesity, and BMI in humans are reviewed. Additionally, the NIH tool Variation Viewer was utilized to identify missense SNPs within the mature, biologically active neuropeptide sequences. For SNPs found through Variation Viewer, a concise discussion on relevant pharmacological structure-activity relationship studies for select SNPs is included. This review is meant to update reported orexigenic neuropeptide SNPs and demonstrate the potential utility of genomic sequence databases for finding SNPs that may result in altered receptor signaling for neuropeptide pathways associated with appetite.
Collapse
Affiliation(s)
- Mark D. Ericson
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Carrie Haskell-Luevano
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
38
|
Parlak N, Görgülü Y, Köse Çinar R, Sönmez MB, Parlak E. Serum agouti-related protein (AgRP) levels in bipolar disorder: Could AgRP be a state marker for mania? Psychiatry Res 2018; 260:36-40. [PMID: 29172096 DOI: 10.1016/j.psychres.2017.11.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 08/21/2017] [Accepted: 11/04/2017] [Indexed: 11/28/2022]
Abstract
Orexigenic and anorexigenic peptides, especially agouti-related protein (AgRP) and leptin, play important roles in the regulation of energy homeostasis in bipolar disorder. AgRP regulates energy metabolism by increasing appetite and decreasing energy expenditure. The resting energy expenditures of patients with manic bipolar disorder are higher than those of controls. Due to the effects of AgRP on energy expenditure and the increased physical activity of manic patients, we hypothesised that serum AgRP levels may be lower in manic patients than in euthymic patients and controls. There was a total of 112 participants, including 47 patients in the manic group, 35 patients in the euthymic group and 30 healthy controls. For this study, serum AgRP, leptin, cholesterol, and cortisol levels were measured and compared between the groups. The serum AgRP, leptin, and cholesterol levels were significantly different between the groups. The serum AgRP levels of manic group were significantly lower than those of euthymic and control groups. The lower serum AgRP levels of manic patients could be indicators of impaired energy homeostasis during manic episodes. Since the serum AgRP levels of manic patients are lower than those of euthymic patients and controls, AgRP could be a state marker for manic episodes.
Collapse
Affiliation(s)
- Naci Parlak
- Department of Psychiatry, Izzet Baysal Mental Health and Disease Training and Research Hospital, Bolu 14030, Turkey.
| | - Yasemin Görgülü
- Department of Psychiatry, Trakya University Faculty of Medicine, Edirne 22030, Turkey.
| | - Rugül Köse Çinar
- Department of Psychiatry, Trakya University Faculty of Medicine, Edirne 22030, Turkey.
| | - Mehmet Bülent Sönmez
- Department of Psychiatry, Trakya University Faculty of Medicine, Edirne 22030, Turkey.
| | - Ebru Parlak
- Department of Psychiatry, Izzet Baysal Mental Health and Disease Training and Research Hospital, Bolu 14030, Turkey.
| |
Collapse
|
39
|
Abstract
Multiple biological, behavioural and genetic determinants or correlates of obesity have been identified to date. Genome-wide association studies (GWAS) have contributed to the identification of more than 100 obesity-associated genetic variants, but their roles in causal processes leading to obesity remain largely unknown. Most variants are likely to have tissue-specific regulatory roles through joint contributions to biological pathways and networks, through changes in gene expression that influence quantitative traits, or through the regulation of the epigenome. The recent availability of large-scale functional genomics resources provides an opportunity to re-examine obesity GWAS data to begin elucidating the function of genetic variants. Interrogation of knockout mouse phenotype resources provides a further avenue to test for evidence of convergence between genetic variation and biological or behavioural determinants of obesity.
Collapse
|
40
|
AgRP Neurons Can Increase Food Intake during Conditions of Appetite Suppression and Inhibit Anorexigenic Parabrachial Neurons. J Neurosci 2017; 37:8678-8687. [PMID: 28821663 DOI: 10.1523/jneurosci.0798-17.2017] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/11/2017] [Accepted: 08/01/2017] [Indexed: 01/13/2023] Open
Abstract
To maintain energy homeostasis, orexigenic (appetite-inducing) and anorexigenic (appetite suppressing) brain systems functionally interact to regulate food intake. Within the hypothalamus, neurons that express agouti-related protein (AgRP) sense orexigenic factors and orchestrate an increase in food-seeking behavior. In contrast, calcitonin gene-related peptide (CGRP)-expressing neurons in the parabrachial nucleus (PBN) suppress feeding. PBN CGRP neurons become active in response to anorexigenic hormones released following a meal, including amylin, secreted by the pancreas, and cholecystokinin (CCK), secreted by the small intestine. Additionally, exogenous compounds, such as lithium chloride (LiCl), a salt that creates gastric discomfort, and lipopolysaccharide (LPS), a bacterial cell wall component that induces inflammation, exert appetite-suppressing effects and activate PBN CGRP neurons. The effects of increasing the homeostatic drive to eat on feeding behavior during appetite suppressing conditions are unknown. Here, we show in mice that food deprivation or optogenetic activation of AgRP neurons induces feeding to overcome the appetite suppressing effects of amylin, CCK, and LiCl, but not LPS. AgRP neuron photostimulation can also increase feeding during chemogenetic-mediated stimulation of PBN CGRP neurons. AgRP neuron stimulation reduces Fos expression in PBN CGRP neurons across all conditions. Finally, stimulation of projections from AgRP neurons to the PBN increases feeding following administration of amylin, CCK, and LiCl, but not LPS. These results demonstrate that AgRP neurons are sufficient to increase feeding during noninflammatory-based appetite suppression and to decrease activity in anorexigenic PBN CGRP neurons, thereby increasing food intake during homeostatic need.SIGNIFICANCE STATEMENT The motivation to eat depends on the relative balance of activity in distinct brain regions that induce or suppress appetite. An abnormal amount of activity in neurons that induce appetite can cause obesity, whereas an abnormal amount of activity in neurons that suppress appetite can cause malnutrition and a severe reduction in body weight. The purpose of this study was to determine whether a population of neurons known to induce appetite ("AgRP neurons") could induce food intake to overcome appetite-suppression following administration of various appetite-suppressing compounds. We found that stimulating AgRP neurons could overcome various forms of appetite suppression and decrease neural activity in a separate population of appetite-suppressing neurons, providing new insights into how the brain regulates food intake.
Collapse
|
41
|
Jiang DN, Li JT, Tao YX, Chen HP, Deng SP, Zhu CH, Li GL. Effects of melanocortin-4 receptor agonists and antagonists on expression of genes related to reproduction in spotted scat, Scatophagus argus. J Comp Physiol B 2017; 187:603-612. [PMID: 28197776 DOI: 10.1007/s00360-017-1062-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 01/04/2017] [Accepted: 01/20/2017] [Indexed: 11/26/2022]
Abstract
Melanocortin-4 receptor (Mc4r) function related to reproduction in fish has not been extensively investigated. Here, we report on gene expression changes by real-time PCR following treatment with Mc4r agonists and antagonists in the spotted scat (Scatophagus argus). Using in vitro incubated hypothalamus, the Mc4r nonselective agonist NDP-MSH ([Nle4, D-Phe7]-α-melanocyte stimulating hormone; 10-6 M) and selective agonist THIQ (N-[(3R)-1, 2, 3, 4-Tetrahydroisoquinolinium-3-ylcarbonyl]- (1R)-1-(4-chlorobenzyl)-2-[4-cyclohexyl-4-(1H-1,2,4-triazol-1-ylmethyl) piperidin-1-yl]-2-oxoethylamine; 10-7 M) significantly increased the expression of gnrh (Gonadotropin releasing hormone), while the Mc4r nonselective antagonist SHU9119 (Ac-Nle-[Asp-His-DPhe/DNal(2')-Arg-Trp-Lys]-NH2; 10-6 M) and selective antagonist Ipsen 5i (compound 5i synthesized in Ipsen Research Laboratories; 10-6 M) significantly inhibited gnrh expression after 3 h of incubation. In incubated pituitary tissue, NDP-MSH and THIQ significantly increased the expression of fshb (Follicle-stimulating hormone beta subunit) and lhb (Luteinizing hormone beta subunit), while SHU9119 and Ipsen 5i significantly decreased fshb and lhb expression after 3 h of incubation. During the in vivo experiment, THIQ (1 mg/kg bw) significantly increased gnrh expression in hypothalamic tissue, as well as the fshb and lhb expression in pituitary tissue 12 h after abdominal injection. Furthermore, Ipsen 5i (1 mg/kg bw) significantly inhibited gnrh expression in hypothalamic tissue, as well as fshb and lhb gene expression in pituitary tissue 12 h after abdominal injection. In summary, Mc4r singling appears to stimulate gnrh expression in the hypothalamus, thereby modulating the synthesis of Fsh and Lh in the pituitary. In addition, Mc4r also appears to directly regulate fshb and lhb levels in the pituitary in spotted scat. Our study suggests that Mc4r, through the hypothalamus and pituitary, participates in reproductive regulation in fish.
Collapse
Affiliation(s)
- Dong-Neng Jiang
- Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Jian-Tao Li
- Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, 36849, USA
| | - Hua-Pu Chen
- Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Si-Ping Deng
- Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Chun-Hua Zhu
- Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Guang-Li Li
- Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China.
| |
Collapse
|
42
|
Li JT, Yang Z, Chen HP, Zhu CH, Deng SP, Li GL, Tao YX. Molecular cloning, tissue distribution, and pharmacological characterization of melanocortin-4 receptor in spotted scat, Scatophagus argus. Gen Comp Endocrinol 2016; 230-231:143-52. [PMID: 27080551 DOI: 10.1016/j.ygcen.2016.04.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 03/26/2016] [Accepted: 04/09/2016] [Indexed: 11/22/2022]
Abstract
Melanocortin-4 receptor (MC4R) plays an important role in the regulation of food intake and energy expenditure in mammals. The functions of the MC4R in fish have not been investigated extensively. We herein reported on the cloning, tissue distribution, and pharmacological characterization of spotted scat (Scatophagus argus) MC4R (SAMC4R). It consisted of a 984bp open reading frame predicted to encode a protein of 327 amino acids. Sequence analysis revealed that SAMC4R was highly homologous (>80%) at amino acid levels to several teleost MC4Rs. Phylogenetic analyses showed that SAMC4R was closely related to piscine MC4R. Using RT-PCR, we showed that in addition to brain, pituitary, and gonads, mc4r mRNA was also widely expressed in peripheral tissues of spotted scat in sexually divergent pattern. With human MC4R (hMC4R) as a control, several agonists including α-melanocyte stimulating hormone (α-MSH), [Nle(4), D-Phe(7)]-α-MSH (NDP-MSH), adrenocorticotropic hormone (ACTH) and THIQ (N-[(3R)-1,2,3,4-tetrahydroisoquinolinium3-ylcarbonyl]-(1R)-1-(4-chlorobenzyl)-2-[4-cyclohexyl-4-(1H-1,2,4-triazol-1-ylmethyl)piperidin-1-yl]-2-oxoethylamine), were used to investigate the binding and signaling properties of SAMC4R. The results showed that SAMC4R bound NDP-MSH with the highest affinity followed by ACTH (1-24) and α-MSH. Similar ranking was also found for hMC4R, although SAMC4R had two to five-fold higher affinities for these ligands. THIQ did not displace NDP-MSH from SAMC4R, different from hMC4R. α-MSH, NDP-MSH, and ACTH (1-24) were identified as potent agonists to stimulate cAMP generation followed by THIQ in SAMC4R. The availability of SAMC4R and its pharmacological characteristics will facilitate the investigation of its function in regulating diverse physiological processes in spotted scat.
Collapse
Affiliation(s)
- Jian-Tao Li
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zhao Yang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Hua-Pu Chen
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Guangdong Ocean University, Zhanjiang 524088, China
| | - Chun-Hua Zhu
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Guangdong Ocean University, Zhanjiang 524088, China
| | - Si-Ping Deng
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Guangdong Ocean University, Zhanjiang 524088, China
| | - Guang-Li Li
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Guangdong Ocean University, Zhanjiang 524088, China.
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
43
|
Regulation of arcuate genes by developmental exposures to endocrine-disrupting compounds in female rats. Reprod Toxicol 2016; 62:18-26. [PMID: 27103539 DOI: 10.1016/j.reprotox.2016.04.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/11/2016] [Accepted: 04/15/2016] [Indexed: 01/09/2023]
Abstract
Developmental exposure to endocrine-disrupting compounds (EDCs) alters reproduction and energy homeostasis, both of which are regulated by the arcuate nucleus (ARC). Little is known about the effects of EDC on ARC gene expression. In Experiment #1, pregnant dams were treated with either two doses of bisphenol A (BPA) or oil from embryonic day (E)18-21. Neonates were injected from postnatal day (PND)0-7. Vaginal opening, body weights, and ARC gene expression were measured. Chrm3 (muscarinic receptor 3) and Adipor1 (adiponectin receptor 1) were decreased by BPA. Bdnf (brain-derived neurotropic factor), Igf1 (insulin-like growth factor 1), Htr2c (5-hydroxytryptamine receptor), and Cck2r (cholescystokinin 2 receptor) were impacted. In Experiment #2, females were exposed to BPA, diethylstilbestrol (DES), di(2-ethylhexyl)phthalate, or methoxychlor (MXC) during E11-PND7. MXC and DES advanced the age of vaginal opening and ARC gene expression was impacted. These data indicate that EDCs alter ARC genes involved in reproduction and energy homeostasis in females.
Collapse
|
44
|
Moehlecke M, Canani LH, Silva LOJE, Trindade MRM, Friedman R, Leitão CB. Determinants of body weight regulation in humans. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2016; 60:152-62. [PMID: 26910628 DOI: 10.1590/2359-3997000000129] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 10/07/2015] [Indexed: 11/21/2022]
Abstract
Body weight is regulated by the ability of hypothalamic neurons to orchestrate behavioral, endocrine and autonomic responses via afferent and efferent pathways to the brainstem and the periphery. Weight maintenance requires a balance between energy intake and energy expenditure. Although several components that participate in energy homeostasis have been identified, there is a need to know in more detail their actions as well as their interactions with environmental and psychosocial factors in the development of human obesity. In this review, we examine the role of systemic mediators such as leptin, ghrelin and insulin, which act in the central nervous system by activating or inhibiting neuropeptide Y, Agouti-related peptide protein, melanocortin, transcript related to cocaine and amphetamine, and others. As a result, modifications in energy homeostasis occur through regulation of appetite and energy expenditure. We also examine compensatory changes in the circulating levels of several peripheral hormones after diet-induced weight loss.
Collapse
|
45
|
Tao YX. Constitutive activity in melanocortin-4 receptor: biased signaling of inverse agonists. ADVANCES IN PHARMACOLOGY 2015; 70:135-54. [PMID: 24931195 DOI: 10.1016/b978-0-12-417197-8.00005-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The melanocortin-4 receptor (MC4R) is a critical regulator of energy homeostasis, including both energy intake and energy expenditure. It mediates the actions of a number of hormones on energy balance. The endogenous ligands for MC4R include peptide agonists derived from processing of proopiomelanocortin and the antagonist Agouti-related peptide (AgRP). Wild-type MC4R has some basal (constitutive) activity. Naturally occurring and laboratory-generated mutations have been identified, which results in either increased or decreased basal activities. Impaired basal signaling has been suggested to be a cause of dysregulated energy homeostasis and early-onset obesity, although several constitutively active mutations have also been identified from obese patients. AgRP and several small-molecule antagonists have been shown to be inverse agonists in the Gs-cAMP pathway. However, in the extracellular signal-regulated kinase (ERK) 1/2 pathway, we showed that these inverse agonists are potent agonists, demonstrating convincingly that they are biased ligands. We also showed that some mutations that do not cause constitutive activation in the Gs-cAMP pathway cause constitutive activation in the ERK1/2 pathway, suggesting that they are biased receptors. The physiological and potential pathophysiological relevance of the biased constitutive signaling in MC4R and therapeutic potential remain to be investigated.
Collapse
Affiliation(s)
- Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA.
| |
Collapse
|
46
|
Kappel V, van Noort B, Ritschel F, Seidel M, Ehrlich S. [Anorexia nervosa - from a neuroscience perspective]. ZEITSCHRIFT FUR KINDER-UND JUGENDPSYCHIATRIE UND PSYCHOTHERAPIE 2013; 42:39-48; quiz 49-50. [PMID: 24365962 DOI: 10.1024/1422-4917/a000268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Anorexia nervosa is a frequent disorder especially among adolescent girls and young women, with high morbidity, mortality, and relapse rates. To date, no single therapeutic approach has proved to be superior to others (Herpertz et al., 2011). It remains unclear how its etiology and pathology are encoded within cognitive, neural, and endocrinological processes that modulate important mechanisms in appetitive processing and weight regulation. Yet, several trait characteristics have been identified in AN which might reflect predisposing factors. Further, altered levels of neuropeptides and hormones that regulate appetite and feeding behavior have been found during both the acute and the recovered state, pointing to dysfunctional mechanisms in AN that persist even after malnutrition has ceased. Researchers are also hoping that brain imaging techniques will allow for a more detailed investigation of the neural basis of reward and punishment sensitivity that appears to be altered in AN. The integration and extension of recent findings in these areas will hopefully provide a more comprehensive understanding of the disorder and hence enable the development of more effective treatments.
Collapse
Affiliation(s)
- Viola Kappel
- Charité-Universitätsmedizin Berlin, Klinik für Psychiatrie, Psychosomatik und Psychotherapie des Kindes- und Jugendalters
| | - Betteke van Noort
- Charité-Universitätsmedizin Berlin, Klinik für Psychiatrie, Psychosomatik und Psychotherapie des Kindes- und Jugendalters
| | - Franziska Ritschel
- Technische Universität Dresden, Universitätsklinikum Carl Gustav Carus Dresden, Klinik und Poliklinik für Kinder- und Jugendpsychiatrie und -psychotherapie
| | - Maria Seidel
- Technische Universität Dresden, Universitätsklinikum Carl Gustav Carus Dresden, Klinik und Poliklinik für Kinder- und Jugendpsychiatrie und -psychotherapie
| | - Stefan Ehrlich
- Technische Universität Dresden, Universitätsklinikum Carl Gustav Carus Dresden, Klinik und Poliklinik für Kinder- und Jugendpsychiatrie und -psychotherapie Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Psychiatric Neuroimaging Research Program
| |
Collapse
|
47
|
Lerma-Cabrera JM, Carvajal F, Alcaraz-Iborra M, de la Fuente L, Navarro M, Thiele TE, Cubero I. Adolescent binge-like ethanol exposure reduces basal α-MSH expression in the hypothalamus and the amygdala of adult rats. Pharmacol Biochem Behav 2013; 110:66-74. [PMID: 23792540 DOI: 10.1016/j.pbb.2013.06.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 06/03/2013] [Accepted: 06/09/2013] [Indexed: 01/31/2023]
Abstract
Melanocortins (MC) are central peptides that have been implicated in the modulation of ethanol consumption. There is experimental evidence that chronic ethanol exposure reduces α-MSH expression in the limbic and hypothalamic brain regions and alters central pro-opiomelanocortin (POMC) mRNA activity in adult rats. Adolescence is a critical developmental period of high vulnerability in which ethanol exposure alters corticotropin releasing factor, neuropeptide Y, substance P and neurokinin neuropeptide activities, all of which have key roles in ethanol consumption. Given the involvement of MC and the endogenous inverse agonist AgRP in ethanol drinking, here we evaluate whether a binge-like pattern of ethanol treatment during adolescence has a relevant impact on basal and/or ethanol-stimulated α-MSH and AgRP activities during adulthood. To this end, adolescent Sprague-Dawley rats (beginning at PND25) were pre-treated with either saline (SP group) or binge-like ethanol exposure (BEP group; 3.0 g/kg given in intraperitoneal (i.p.) injections) of one injection per day over two consecutive days, followed by 2 days without injections, repeated for a total of 8 injections. Following 25 ethanol-free days, we evaluated α-MSH and AgRP immunoreactivity (IR) in the limbic and hypothalamic nuclei of adult rats (PND63) in response to ethanol (1.5 or 3.0 g/kgi.p.) and saline. We found that binge-like ethanol exposure during adolescence significantly reduced basal α-MSH IR in the central nucleus of the amygdala (CeA), the arcuate nucleus (Arc) and the paraventricular nucleus of the hypothalamus (PVN) during adulthood. Additionally, acute ethanol elicited AgRP IR in the Arc. Rats given the adolescent ethanol treatment required higher doses of ethanol than saline-treated rats to express AgRP. In light of previous evidence that endogenous MC and AgRP regulate ethanol intake through MC-receptor signaling, we speculate that the α-MSH and AgRP disturbances induced by binge-like ethanol exposure during adolescence may contribute to excessive ethanol consumption during adulthood.
Collapse
|
48
|
Kraemer RR, Castracane VD, Francois M, Ghanbari-Niaki A, Sirikul B, Valverde RA. Effects of prolonged exercise on agouti-related protein: a pilot study. Endocrine 2012; 42:436-41. [PMID: 22477065 DOI: 10.1007/s12020-012-9663-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 03/22/2012] [Indexed: 10/28/2022]
Abstract
Agouti-related protein (AgRP), is a signaling peptide that affects feeding behavior, energy homeostasis, and has also been shown to stimulate the hypothalamic-pituitary-adrenal axis. The purpose of this study was to determine the effects of 90 min of treadmill exercise on circulating AgRP concentrations and the relationship of AgRP responses to cortisol. Seven young males completed a preliminary trial followed by counterbalanced experimental and control trials 4-5 weeks apart. The experimental trial began 2.5 h after consumption of a standard nutrient beverage and consisted of treadmill exercise at 60 % of previously determined VO(2max) for 90 min. Blood samples were collected before (-30 and 0 min), during (18, 36, 54, 72, and 90 min), and following exercise (20, 40, and 60 min). Blood samples were collected in a resting, control trial at the same time points as the experimental trial. Plasma lactate was significantly higher in the exercise than the control trial. Although AgRP increased from 18 min of exercise to peak at 90 min, these increases were not significantly different than values in the control trial. Cortisol responses during the exercise trial were significantly higher than the control trial. AgRP concentrations during early exercise were positively correlated with cortisol levels later in recovery. The obtained data suggest that AgRP concentrations during prolonged steady-state exercise are associated with subsequent cortisol increases, but further study is required to determine whether there is a causal effect.
Collapse
Affiliation(s)
- Robert R Kraemer
- Department of Kinesiology and Health Studies, Southeastern Louisiana University, SLU10845, Hammond, LA 70402, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Melanocortin system in cancer-related cachexia. Open Med (Wars) 2011. [DOI: 10.2478/s11536-011-0057-6] [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
AbstractThe melanocortin system plays a pivotal role in the regulation of appetite and energy balance. It was recognized to play an important role in the development of cancer-related cachexia, a debilitating condition characterized by progressive body wasting associated with anorexia, increased resting energy expediture and loss of fat as well as lean body mass that cannot be simply prevented or treated by adequate nutritional support.The recent advances in understanding of mechanisms underlying cancer-related cachexia led to consequent recognition of the melanocortin system as an important potential therapeutic target. Several molecules have been made available for animal experiments, including those with oral bioavailability, that act at various checkpoints of the melanocortin system and that might confer singificant benefits for the patients suffering from cancer-related cachexia. The application of melanocortin 4 receptor antagonists/agouti-related peptide agonists has been however restricted to animal models and more pharmacological data will be necessary to progress to clinical trials on humans. Still, pharmacological targeting of the melanocortin system seem to represent an elegant and promising way of treatment of cancer-related cachexia.
Collapse
|
50
|
Zhang LN, Mitchell SE, Hambly C, Morgan DG, Clapham JC, Speakman JR. Physiological and behavioral responses to intermittent starvation in C57BL/6J mice. Physiol Behav 2011; 105:376-87. [PMID: 21907222 DOI: 10.1016/j.physbeh.2011.08.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 08/08/2011] [Accepted: 08/26/2011] [Indexed: 01/29/2023]
Abstract
The dual intervention point model states that body mass is controlled by upper and lower intervention points, above and below which animals (and humans) intervene physiologically to bring their body mass back into the acceptable range. It has been further suggested that the lower intervention point may be defined by the risk of starvation, while the upper intervention point may be defined by the risk of predation. The objective of the present study was to test whether the risk of starvation determines the lower intervention point and to examine the physiological and behavioral mechanisms that underpin the regulation of body mass, when the risk of starvation is increased. Sixty-four mice were exposed to random days of complete fasting or 50% food restriction and their body mass and fat mass responses were measured. Food intake, physical activity and body temperature were measured throughout the experiment. In addition, plasma leptin and insulin, triglyceride and non-esterified fatty acids, along with hypothalamic neuropeptides gene expression in the arcuate nucleus were assessed after 13 and 42 days of treatment. We found that C57BL/6J mice increased body mass and fatness in response to a short-term (13 days) intermittent fasting, which was restored to baseline as the treatment was prolonged. In contrast, intermittently 50% food restricted mice showed no significant changes in body mass or fatness. Over the first 13 days of treatment the data were consistent with the dual intervention point model as the mice showed both increased body mass and adiposity over this period. Over the more protracted period of 42 days the effect waned and was therefore inconsistent with the model. The body mass and fat mass gains in intermittently fasted mice were mainly accounted for by increased food intake. Elevated NPY gene expression after 13 days (three 24 h fasting events) may have driven the increase in food intake. However, no changes were observed in such neuropeptides as POMC, CART, AgRP, Ob-Rb and SOCS 3 or circulating levels of leptin, insulin, NEFA and TG. Hypothermia during fasting days may have also contributed to the increase in body mass. Over 42 days of treatment (nine 24 h fasting events) cumulative food intake was not affected by intermittent starvation. However physical activity, mainly activity during the light phase was lowered suggesting an adaptation to unpredictable starvation. Overall, mice exhibited different behavioral and physiological responses to intermittent starvation depending on the duration of treatment.
Collapse
Affiliation(s)
- Li-Na Zhang
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | | | | | | | | | | |
Collapse
|