1
|
Kahe K, Laferrère B, Castellanos FX, Zhang Y, Mozaffarian D. Monosodium glutamate: A hidden risk factor for obesity? Obes Rev 2025; 26:e13903. [PMID: 39914377 PMCID: PMC12168872 DOI: 10.1111/obr.13903] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/18/2024] [Accepted: 11/17/2024] [Indexed: 05/14/2025]
Abstract
Monosodium glutamate (MSG) has become one of the most widely used food additives in the global food supply. Although it has been classified for decades as a food ingredient that is generally recognized as safe, concerns about the health impacts of chronic MSG use, especially its potential effect on weight, are still ongoing. This comprehensive review summarizes the available human and animal evidence, highlighting potential mechanisms linking MSG use to weight gain or obesity, and discusses challenges and future research directions. Because of MSG intake worldwide as well as hidden MSG in food labeling, there is a pressing need for a mechanistic understanding of the health impacts of MSG use especially on weight. To generate robust scientific evidence and to clarify public concerns, rigorous mechanistic studies and randomized controlled clinical trials are warranted.
Collapse
Affiliation(s)
- Ka Kahe
- Department of Obstetrics and Gynecology and Department of Epidemiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Blandine Laferrère
- Division of Endocrinology, New York Nutrition Obesity Research Center, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Francisco X. Castellanos
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
- Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Yijia Zhang
- Department of Obstetrics and Gynecology and Department of Epidemiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Dariush Mozaffarian
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| |
Collapse
|
2
|
Kim N, Kim S, Park S, Kim EK. Adenosine transmission from hypothalamic tanycytes to AGRP/NPY neurons regulates energy homeostasis. Exp Mol Med 2025:10.1038/s12276-025-01449-6. [PMID: 40316705 DOI: 10.1038/s12276-025-01449-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/14/2025] [Accepted: 03/03/2025] [Indexed: 05/04/2025] Open
Abstract
Tanycytes are a pivotal component of the hypothalamic network that controls energy homeostasis. Despite their importance, the regulatory mechanisms governing tanycyte-neuron interactions in response to metabolic signals remain unexplored. Here we report that adenosine signaling between tanycytes and AGRP/NPY neurons is crucial for tanycytic metabolic regulation mediated by translocator protein 18 kDa (TSPO). Tanycyte-specific Tspo-knockout mice displayed reduced food consumption and weight loss associated with the downregulation of Agrp and Npy expression under high-fat diet feeding. Tspo-deficient tanycytes had elevated levels of intracellular ATP, which was released via connexin 43 hemichannels and extracellularly converted into adenosine by tanycytic ectonucleotidases. The adenosine signal was perceived by adenosine A1 receptors on adjacent AGRP/NPY neurons, reducing ERK phosphorylation, which in turn downregulated Agrp and Npy expression. Our findings underscore the anorexic role of adenosine as a gliotransmitter in the intricate communication between tanycytes and neurons for regulating appetite and body weight.
Collapse
Affiliation(s)
- Nayoun Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Seolsong Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Seokjae Park
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
- Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Eun-Kyoung Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea.
- Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea.
| |
Collapse
|
3
|
Diaz AV, Tekin I, Reis T. Drosophila as a Genetic Model System to Study Organismal Energy Metabolism. Biomolecules 2025; 15:652. [PMID: 40427545 PMCID: PMC12108566 DOI: 10.3390/biom15050652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Metabolism is the essential process by which an organism converts nutrients into energy to fuel growth, development, and repair. Metabolism at the level of a multicellular, multi-organ animal is inherently more complex than metabolism at the single-cell level. Indeed, each organ also must maintain its own homeostasis to function. At all three scales, homeostasis is a defining feature: as energy sources and energetic demands wax and wane, the system must be robust. While disruption of organismal energy homeostasis can be manifested in different ways in humans, obesity (defined as excess body fat) is an increasingly common outcome of metabolic imbalance. Here we will discuss the genetic basis of metabolic dysfunction that underlies obesity. We focus on what we are learning from Drosophila melanogaster as a model organism to explore and dissect genetic causes of metabolic dysfunction in the context of a whole organism.
Collapse
Affiliation(s)
| | | | - Tânia Reis
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
4
|
Cheraghi S, Mobaderi T, Mottaghi A, Movahedi Motlagh F, Taghizadeh S, Eghbali M. Genetic variants in the MC4R gene and risk of obesity/overweight: A systematic review and meta-analysis. Diabetes Obes Metab 2025. [PMID: 40302631 DOI: 10.1111/dom.16425] [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: 01/04/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/02/2025]
Abstract
AIM Obesity is a significant health issue worldwide, progressing due to genetic factors and lifestyle. Melanocortin 4 receptor (MC4R) gene polymorphisms have been identified as a cause of overweight and obesity risk. The aim of this study was a comprehensive assessment of MC4R polymorphism effects on overweight/obesity risk. METHODS All retrieved literature from PubMed, Web of Science and Scopus according to PRISMA guidelines up to June 2022 was reviewed. Inclusion criteria are restricted to English-language, human case-control/cohort studies with genotype distributions of MC4R polymorphisms and their association with obesity and overweight in any geographic regions and age. The heterogeneity using the I-squared statistic (I2), the Q-test and Prediction Interval (PI) and publication bias using Begg's and Egger's tests were examined, and the pooled odds ratios in different genetic models were estimated using a random effect model. Subgroup analysis was performed by the geographic regions and age groups. Risk of bias for individual studies was not assessed. The review is limited by restricted racial diversity and exclusion of environmental factors, incomplete data and limited access to certain articles. This work received no specific funding, and the review was not prospectively registered. RESULTS In our study, 39 eligible studies with 43 697 overweight and obese cases and 52 272 normal weights were included. In mixed-age populations, rs17700633, rs17782313, rs11872992, rs12970134, rs2229616 and rs571312 were evaluated. The remarkable association was seen by rs17782313 and rs12970134 in the Homozygous model (OR = 1.73; 95% CI: 1.51, 1.98 and 1.74; 95% CI: 1.29; 2.35, respectively). In addition, rs17782313 and rs12970134 were found to be more strongly linked to overweight and obesity in Asian and European population groups, as determined by a subgroup analysis of the geographic regions. CONCLUSION The present study confirms the high association of rs17782313 and rs12970134 with obesity and overweight in all age groups and geographic regions. However, further functional studies and high-population research on other MC4R SNPs must validate their role.
Collapse
Affiliation(s)
- Sara Cheraghi
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Tofigh Mobaderi
- Minimally Invasive Surgery Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Azadeh Mottaghi
- Research Center for Prevention of Cardiovascular Diseases, Endocrinology & Metabolism, Institute of Endocrinology Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | | | - Sara Taghizadeh
- Translational Ophthalmology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Eghbali
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Miro C, Cicatiello AG, Nappi A, Sagliocchi S, Acampora L, Restolfer F, Cuomo O, de Alteris G, Pugliese G, Torabinejad S, Maritato R, Murolo M, Di Cicco E, Velotti N, Capuano M, La Civita E, Terracciano D, Ciampaglia R, Stornaiuolo M, Musella M, Aprea G, Pignataro G, Savastano S, Dentice M. Leptin enhances the intracellular thyroid hormone activation in skeletal muscle to boost energy balance. Cell Metab 2025; 37:936-953.e7. [PMID: 39986272 DOI: 10.1016/j.cmet.2025.01.025] [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: 03/29/2024] [Revised: 10/15/2024] [Accepted: 01/27/2025] [Indexed: 02/24/2025]
Abstract
Thyroid hormones (THs) are key modulators of energy metabolism and cross-talk with other endocrine and metabolic factors. Notably, leptin can increase hypothalamic control of TH synthesis as an adaptive metabolic response regulating body weight. In this study, we found that the TH signal is heightened in overweight humans and is lost with obesity. In mice, systemic and intracerebroventricular leptin injection induces the expression of type 2 deiodinase (D2), the TH-activating enzyme, in skeletal muscle. Mechanistically, leptin enhances the transcription of D2 by a STAT3- and α-melanocyte-stimulating hormone (α-MSH)/cyclic AMP (cAMP)-dependent regulation. Notably, mice lacking D2 or with a mutation in the TH receptor do not exhibit the metabolic effects of leptin, such as increased insulin sensitivity and oxygen consumption, indicating that leptin's peripheral metabolic effects in skeletal muscle are mediated by TH. These findings underscore the critical role of leptin in integrating the TH-induced metabolic activation, while also contributing to appetite suppression in response to perceived fat stores.
Collapse
Affiliation(s)
- Caterina Miro
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | | | - Annarita Nappi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Serena Sagliocchi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Lucia Acampora
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Federica Restolfer
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Ornella Cuomo
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II," 80131 Naples, Italy
| | - Giulia de Alteris
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Gabriella Pugliese
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Sepehr Torabinejad
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Rosa Maritato
- Department of Translational Medical Sciences, University of Naples "Federico II," 80131 Naples, Italy
| | - Melania Murolo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Emery Di Cicco
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Nunzio Velotti
- Department of Advanced Biomedical Sciences, University of Naples "Federico II," 80131 Naples, Italy
| | - Marianna Capuano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Evelina La Civita
- Department of Translational Medical Sciences, University of Naples "Federico II," 80131 Naples, Italy
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University of Naples "Federico II," 80131 Naples, Italy
| | - Roberto Ciampaglia
- Department of Pharmacy, University of Naples "Federico II," 80149 Naples, Italy
| | - Mariano Stornaiuolo
- Department of Pharmacy, University of Naples "Federico II," 80149 Naples, Italy
| | - Mario Musella
- Department of Advanced Biomedical Sciences, University of Naples "Federico II," 80131 Naples, Italy
| | - Giovanni Aprea
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II," 80131 Naples, Italy
| | - Silvia Savastano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy; CEINGE - Biotecnologie Avanzate S.c.a.r.l., 80131 Naples, Italy.
| |
Collapse
|
6
|
Grajales-Reyes JG. Advances in energy balance & metabolism circuitry. ADVANCES IN GENETICS 2025; 113:1-28. [PMID: 40409794 DOI: 10.1016/bs.adgen.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Advancements in informatics, genetics, and neuroscience have greatly expanded our understanding of how the central nervous system (CNS) regulates energy balance and metabolism. This chapter explores the key neural circuits within the hypothalamus and brainstem that integrate behavioral and physiological processes to maintain metabolic homeostasis. It also examines the dynamic interplay between the CNS and peripheral organs, mediated through hormonal and neuronal signals, which fine-tune appetite, energy expenditure, and body weight. Furthermore, we highlight groundbreaking research that unveils molecular and cellular pathways governing energy regulation, representing a new frontier in addressing obesity and metabolic disorders. Innovative approaches, such as neurogenetic and neuromodulation techniques, are explored as promising strategies for improving weight management and metabolic health. By providing a comprehensive perspective on the mechanisms underlying energy balance, this chapter underscores the transformative potential of emerging therapeutic innovations.
Collapse
Affiliation(s)
- Jose G Grajales-Reyes
- Department of Anesthesiology, Yale University, New Haven, CT, United States; Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, United States.
| |
Collapse
|
7
|
Haddad HK, Mercado-Reyes JI, Mustafá ER, D’Souza SP, Chung CS, Nestor RRM, Olinski LE, Martinez Damonte V, Saskin J, Vemaraju S, Raingo J, Kauer JA, Lang RA, Oancea E. Hypothalamic opsin 3 suppresses MC4R signaling and potentiates Kir7.1 to promote food consumption. Proc Natl Acad Sci U S A 2025; 122:e2403891122. [PMID: 39951488 PMCID: PMC11874419 DOI: 10.1073/pnas.2403891122] [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: 02/23/2024] [Accepted: 12/02/2024] [Indexed: 02/16/2025] Open
Abstract
Mammalian opsin 3 (OPN3) is a member of the opsin family of G-protein-coupled receptors with ambiguous light sensitivity. OPN3 was first identified in the brain (and named encephalopsin) and subsequently found to be expressed in other tissues. In adipocytes, OPN3 is necessary for light responses that modulate lipolysis and glucose uptake, while OPN3 in human skin melanocytes regulates pigmentation in a light-independent manner. Despite its initial discovery in the brain, OPN3 functional mechanisms in the brain remain elusive. Here, we investigated the molecular mechanism of OPN3 function in the paraventricular nucleus (PVN) of the hypothalamus. We show that Opn3 is coexpressed with the melanocortin 4 receptor (Mc4r) in a population of PVN neurons, where it negatively regulates MC4R-mediated cAMP signaling in a specific and Gαi/o-dependent manner. Under baseline conditions, OPN3 via Gαi/o potentiates the activity of the inward rectifying Kir7.1 channel, previously shown to be closed in response to agonist-mediated activation of MC4R in a Gαs-independent manner. In mice, we found that Opn3 in Mc4r-expressing neurons regulates food consumption. Our results reveal the first mechanistic insight into OPN3 function in the hypothalamus, uncovering a unique mechanism by which OPN3 functions to potentiate Kir7.1 activity and negatively regulate MC4R-mediated cAMP signaling, thereby promoting food intake.
Collapse
Affiliation(s)
- Hala K. Haddad
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Jonathan I. Mercado-Reyes
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - E. Román Mustafá
- Electrophysiology Lab, Instituto Multidisciplinario de Biología Celular, La Plata, Buenos Aires1900, Argentina
| | - Shane P. D’Souza
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - C. Sean Chung
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Ramses R. M. Nestor
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Lauren E. Olinski
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Valentina Martinez Damonte
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
| | - Joshua Saskin
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Shruti Vemaraju
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Jesica Raingo
- Electrophysiology Lab, Instituto Multidisciplinario de Biología Celular, La Plata, Buenos Aires1900, Argentina
| | - Julie A. Kauer
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
| | - Richard A. Lang
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Department of Ophthalmology, University of Cincinnati, Cincinnati, OH45229
| | - Elena Oancea
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| |
Collapse
|
8
|
Simonds SE, Pryor JT, Lam BYH, Dowsett GK, Mustafa T, Munder A, Elysee K, Balland E, Cowley LO, Yeo GSH, Lawrence A, Spanswick DC, Cowley MA. The metabolic and cardiovascular effects of amphetamine are partially mediated by the central melanocortin system. Cell Rep Med 2025; 6:101936. [PMID: 39914386 PMCID: PMC11866487 DOI: 10.1016/j.xcrm.2025.101936] [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: 04/12/2022] [Revised: 09/02/2024] [Accepted: 01/09/2025] [Indexed: 02/21/2025]
Abstract
Amphetamine (AMPH) exerts metabolic and cardiovascular effects. The central melanocortin system is a key regulator of both metabolic and cardiovascular functions. Here, we show that the melanocortin system partially mediates AMPH-induced anorexia, energy expenditure, tachycardia, and hypertension. AMPH increased α-melanocyte stimulating hormone (αMSH) secretion from the hypothalamus, elevated blood pressure and heart rate (HR), increased brown adipose tissue (BAT) thermogenesis, and reduced both food intake (FI) and body weight (BW). In melanocortin 4 receptor-deficient (MC4R knockout [KO]) mice, metabolic and cardiovascular effects of AMPH were significantly attenuated. Antagonism of serotonergic and noradrenergic neurotransmitter systems attenuated AMPH-induced αMSH secretion as well as AMPH-induced metabolic and cardiovascular effects. We propose that AMPH increases serotonergic activation of proopiomelanocortin (POMC) neurons and reduces the noradrenergic inhibition of POMC neurons, thereby disinhibiting them. Together, these presynaptic mechanisms result in increased POMC activity, increased αMSH secretion, and increased activation of MC4R pathways that regulate both the metabolic and cardiovascular systems.
Collapse
MESH Headings
- Animals
- Amphetamine/pharmacology
- Pro-Opiomelanocortin/metabolism
- alpha-MSH/metabolism
- Receptor, Melanocortin, Type 4/metabolism
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/deficiency
- Mice
- Mice, Knockout
- Male
- Energy Metabolism/drug effects
- Blood Pressure/drug effects
- Melanocortins/metabolism
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/drug effects
- Mice, Inbred C57BL
- Hypothalamus/metabolism
- Hypothalamus/drug effects
- Thermogenesis/drug effects
- Heart Rate/drug effects
- Cardiovascular System/drug effects
- Cardiovascular System/metabolism
- Neurons/metabolism
- Neurons/drug effects
- Body Weight/drug effects
- Eating/drug effects
Collapse
Affiliation(s)
- Stephanie E Simonds
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia.
| | - Jack T Pryor
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| | - Brian Y H Lam
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Georgina K Dowsett
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Tomris Mustafa
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| | - Astrid Munder
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| | - Kayla Elysee
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| | - Eglantine Balland
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| | - Lachlan O Cowley
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| | - Giles S H Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Andrew Lawrence
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia
| | - David C Spanswick
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia; Warwick Medical School, Division of Biomedical Sciences, University of Warwick, CV4 7AL Coventry, UK
| | - Michael A Cowley
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia.
| |
Collapse
|
9
|
Weirath NA, Zajac JWP, Donow HM, Lavoi TM, Pinilla C, Santos RG, Prajapati R, Speth R, Ericson MD, Sarupria S, Giulianotti MA, Haskell-Luevano C. N-Branched Tricyclic Guanidines as Novel Melanocortin-3 Receptor Agonists and Melanocortin-4 Receptor Antagonists. J Med Chem 2025; 68:2504-2527. [PMID: 39832483 DOI: 10.1021/acs.jmedchem.4c01556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The melanocortin receptors are a class of centrally and peripherally expressed G protein-coupled receptors, of which the MC3R and MC4R subtypes are implicated in the regulation of appetite and energy homeostasis and can serve as potential therapeutic targets for disorders such as obesity and cachexia. An unbiased high-throughput mixture-based library screen was implemented to identify novel ligands with an emphasis on the identification of nanomolar-potent agonists of the mouse melanocortin-3 receptor. This screen yielded the discovery of an N-branched tricyclic guanidine scaffold (TPI2408) that contained three nanomolar potent mMC3R agonists and additional compounds that possessed antagonism for the mMC4R. The antagonist character of this scaffold library at the mMC4R was confirmed by a follow-up positional scanning antagonist screen. Additionally, molecular dynamics simulations herein provide mechanistic insight into the polypharmacological characteristics of melanocortin receptors. The disclosed materials have the potential to serve as important tools and SAR scaffolds in the study of melanocortin receptor function.
Collapse
MESH Headings
- Receptor, Melanocortin, Type 3/agonists
- Receptor, Melanocortin, Type 3/antagonists & inhibitors
- Receptor, Melanocortin, Type 3/metabolism
- Receptor, Melanocortin, Type 4/agonists
- Receptor, Melanocortin, Type 4/antagonists & inhibitors
- Receptor, Melanocortin, Type 4/metabolism
- Animals
- Mice
- Humans
- Structure-Activity Relationship
- Guanidines/pharmacology
- Guanidines/chemistry
- Guanidines/chemical synthesis
- Molecular Dynamics Simulation
- HEK293 Cells
Collapse
Affiliation(s)
- Nicholas A Weirath
- Department of Medicinal Chemistry and the Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jonathan W P Zajac
- Department of Chemistry and Chemical Theory Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Haley M Donow
- Florida International University, Port St. Lucie, Florida 34978, United States
| | - Travis M Lavoi
- Florida International University, Port St. Lucie, Florida 34978, United States
| | - Clemencia Pinilla
- Department of Medicinal Chemistry and the Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Radleigh G Santos
- Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, Florida 33314, United States
| | - Ritu Prajapati
- Department of Medicinal Chemistry and the Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Robert Speth
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, 3300 S. University Dr., Fort Lauderdale, Florida 33328, United States
- Department of Pharmacology and Physiology, Georgetown University School of Medicine, 3900 Reservoir Rd. NW, Washington, District of Columbia 20007, United States
| | - Mark D Ericson
- Department of Medicinal Chemistry and the Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Sapna Sarupria
- Department of Chemistry and Chemical Theory Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Marcello A Giulianotti
- Department of Medicinal Chemistry and the Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Carrie Haskell-Luevano
- Department of Medicinal Chemistry and the Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
10
|
Christensen CW, Weed SE, Brown TE, Hentges ST. Exploring the role of beta-endorphin in activity-based anorexia in mice. Physiol Rep 2025; 13:e70201. [PMID: 39930661 PMCID: PMC11810985 DOI: 10.14814/phy2.70201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/20/2024] [Accepted: 01/06/2025] [Indexed: 02/14/2025] Open
Abstract
Anorexia nervosa (AN) remains one of the most lethal mental health disorders and is poorly understood from a neurobiological perspective. The most widely used animal model of AN is activity-based anorexia (ABA) where scheduled food presentation leads to a spontaneous maladaptive increase in running-wheel activity and rapid weight loss in rodents, recapitulating specific aspects of AN. Research using the ABA paradigm to probe the role of hedonic and homeostatic circuits has indicated that the hypothalamic proopiomelanocortin (POMC) system may play a role in both the increased activity and reduced food intake observed. Previous work has shown that Pomc mRNA and its peptide product beta-endorphin (β-end) are increased during the onset of ABA. β-end is reinforcing and increases locomotor activity, and mice lacking the mu opioid receptor (MOR), the primary target of β-end, display blunted food-anticipatory activity in the ABA paradigm. Thus, the current work was designed to determine if aspects of ABA would be diminished in mice lacking β-end. We did not find any significant differences in wheel-running, food intake, or body weight loss in β-end knockout mice of either sex during ABA compared to wild-type littermates. Therefore, we conclude that the development of ABA does not require β-end.
Collapse
Affiliation(s)
- Connor W. Christensen
- Department of Integrative Physiology and NeuroscienceWashington State UniversityPullmanWashingtonUSA
| | - Samantha E. Weed
- Department of Integrative Physiology and NeuroscienceWashington State UniversityPullmanWashingtonUSA
| | - Travis E. Brown
- Department of Integrative Physiology and NeuroscienceWashington State UniversityPullmanWashingtonUSA
| | - Shane T. Hentges
- Department of Integrative Physiology and NeuroscienceWashington State UniversityPullmanWashingtonUSA
| |
Collapse
|
11
|
Schöneberg T. Modulating vertebrate physiology by genomic fine-tuning of GPCR functions. Physiol Rev 2025; 105:383-439. [PMID: 39052017 DOI: 10.1152/physrev.00017.2024] [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: 04/22/2024] [Revised: 07/08/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024] Open
Abstract
G protein-coupled receptors (GPCRs) play a crucial role as membrane receptors, facilitating the communication of eukaryotic species with their environment and regulating cellular and organ interactions. Consequently, GPCRs hold immense potential in contributing to adaptation to ecological niches and responding to environmental shifts. Comparative analyses of vertebrate genomes reveal patterns of GPCR gene loss, expansion, and signatures of selection. Integrating these genomic data with insights from functional analyses of gene variants enables the interpretation of genotype-phenotype correlations. This review underscores the involvement of GPCRs in adaptive processes, presenting numerous examples of how alterations in GPCR functionality influence vertebrate physiology or, conversely, how environmental changes impact GPCR functions. The findings demonstrate that modifications in GPCR function contribute to adapting to aquatic, arid, and nocturnal habitats, influencing camouflage strategies, and specializing in particular dietary preferences. Furthermore, the adaptability of GPCR functions provides an effective mechanism in facilitating past, recent, or ongoing adaptations in animal domestication and human evolution and should be considered in therapeutic strategies and drug development.
Collapse
Affiliation(s)
- Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
- School of Medicine, University of Global Health Equity, Kigali, Rwanda
| |
Collapse
|
12
|
Possa‐Paranhos IC, Butts J, Pyszka E, Nelson C, Congdon S, Cho D, Sweeney P. Medial hypothalamic MC3R signalling regulates energy rheostasis in adult mice. J Physiol 2025; 603:379-410. [PMID: 39718394 PMCID: PMC11737543 DOI: 10.1113/jp286699] [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: 04/08/2024] [Accepted: 11/11/2024] [Indexed: 12/25/2024] Open
Abstract
Although mammals resist both acute weight loss and weight gain, the neural circuitry mediating bi-directional defense against weight change is incompletely understood. Global constitutive deletion of the melanocortin-3-receptor (MC3R) impairs the behavioural response to both anorexic and orexigenic stimuli, with MC3R knockout mice demonstrating increased weight gain following anabolic challenges and increased weight loss following anorexic challenges (i.e. impaired energy rheostasis). However, the brain regions mediating this phenotype are not well understood. Here, we utilized MC3R floxed mice and viral injections of Cre-recombinase to selectively delete MC3R from the medial hypothalamus (MH) in adult mice. Behavioural assays were performed on these animals to test the role of MC3R in MH in the acute response to orexigenic and anorexic challenges. Complementary chemogenetic approaches were used in MC3R-Cre mice to localize and characterize the specific medial hypothalamic brain regions mediating the role of MC3R in energy homeostasis. Finally, we performed RNAscope in situ hybridization to map changes in the mRNA expression of MC3R, pro-opiomelanocortin and agouti-related peptide following energy rheostatic challenges, as well as to characterize the MC3R expressing cells in dorsal MH. Our results demonstrate that MC3R deletion in MH increases feeding and weight gain following high-fat diet feeding, and enhances the anorexic effects of semaglutide, in a sexually dimorphic manner. Furthermore, although the arcuate nucleus exerts an important role in MC3R-mediated effects on energy homeostasis, viral deletion in the dorsal MH also resulted in altered energy rheostasis, indicating that brain regions outside of the arcuate nucleus also contribute to the role of MC3R in energy rheostasis. Together, these results demonstrate that MC3R-mediated effects on energy rheostasis result from the loss of MC3R signalling in medial hypothalamic neurons and suggest an important role for dorsal-MH MC3R signalling in energy rheostasis. KEY POINTS: Melanocortin-3-receptor (MC3R) signalling regulates energy rheostasis in adult mice. Medial hypothalamus regulates energy rheostasis in adult mice. Energy rheostatic stimuli alter mRNA levels of agouti-related peptide, pro-opiomelanocortin and MC3R. Dorsal-medial hypothalamus (DMH) MC3R neurons increase locomotion and energy expenditure. MC3R cell types in DMH are sexually dimorphic.
Collapse
Affiliation(s)
| | - Jared Butts
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
- University of Illinois Urbana‐Champaign Neuroscience ProgramUrbanaILUSA
| | - Emma Pyszka
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
| | - Christina Nelson
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
| | - Samuel Congdon
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
| | - Dajin Cho
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
- University of Illinois Urbana‐Champaign Neuroscience ProgramUrbanaILUSA
| | - Patrick Sweeney
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
- University of Illinois Urbana‐Champaign Neuroscience ProgramUrbanaILUSA
| |
Collapse
|
13
|
Zhang SX, Kim A, Madara JC, Zhu PK, Christenson LF, Lutas A, Kalugin PN, Sunkavalli PS, Jin Y, Pal A, Tian L, Lowell BB, Andermann ML. Stochastic neuropeptide signals compete to calibrate the rate of satiation. Nature 2025; 637:137-144. [PMID: 39506113 PMCID: PMC11981016 DOI: 10.1038/s41586-024-08164-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/07/2024] [Indexed: 11/08/2024]
Abstract
Neuropeptides have important roles in neural plasticity, spiking and behaviour1. Yet, many fundamental questions remain regarding their spatiotemporal transmission, integration and functions in the awake brain. Here we examined how MC4R-expressing neurons in the paraventricular nucleus of the hypothalamus (PVHMC4R) integrate neuropeptide signals to modulate feeding-related fast synaptic transmission and titrate the transition to satiety2-6. We show that hunger-promoting AgRP axons release the neuropeptide NPY to decrease the second messenger cAMP in PVHMC4R neurons, while satiety-promoting POMC axons release the neuropeptide αMSH to increase cAMP. Each release event is all-or-none, stochastic and can impact multiple neurons within an approximately 100-µm-diameter region. After release, NPY and αMSH peptides compete to control cAMP-the amplitude and persistence of NPY signalling is blunted by high αMSH in the fed state, while αMSH signalling is blunted by high NPY in the fasted state. Feeding resolves this competition by simultaneously elevating αMSH release and suppressing NPY release7,8, thereby sustaining elevated cAMP in PVHMC4R neurons throughout a meal. In turn, elevated cAMP facilitates potentiation of feeding-related excitatory inputs with each bite to gradually promote satiation across many minutes. Our findings highlight biochemical modes of peptide signal integration and information accumulation to guide behavioural state transitions.
Collapse
Affiliation(s)
- Stephen X Zhang
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Angela Kim
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Joseph C Madara
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Paula K Zhu
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lauren F Christenson
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Andrew Lutas
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter N Kalugin
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Praneel S Sunkavalli
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yihan Jin
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Neuroscience Graduate Group, Center for Neuroscience, University of California, Davis, Sacramento, CA, USA
- Max Planck Florida Institute for Neuroscience, One Max Planck Way, Jupiter, FL, USA
| | - Akash Pal
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Max Planck Florida Institute for Neuroscience, One Max Planck Way, Jupiter, FL, USA
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Max Planck Florida Institute for Neuroscience, One Max Planck Way, Jupiter, FL, USA
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mark L Andermann
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Zhu X, Potterfield R, Gruber KA, Zhang E, Newton SD, Norgard MA, Levasseur PR, Bai P, Chen X, Gu Q, Grossberg AJ, Marks DL. Melanocortin-4 receptor antagonist TCMCB07 alleviates chemotherapy-induced anorexia and weight loss in rats. J Clin Invest 2024; 135:e181305. [PMID: 39509261 DOI: 10.1172/jci181305] [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: 03/21/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024] Open
Abstract
Cancer patients undergoing chemotherapy often experience anorexia and weight loss that substantially deteriorates overall health, reduces treatment tolerance and quality of life, and worsens oncologic outcomes. There are currently few effective therapeutic options to mitigate these side effects. The central melanocortin system, which plays a pivotal role in regulating appetite and energy homeostasis, presents a logical target for treating anorexia and weight loss. In this preclinical study, we evaluated the efficacy of TCMCB07, a synthetic antagonist of the melanocortin-4 receptor, in mitigating anorexia and weight loss in several rat models of chemotherapy: cisplatin, 5-fluorouracil, cyclophosphamide, vincristine, doxorubicin, and a combination of irinotecan and 5-fluorouracil. Our results indicate that peripheral administration of TCMCB07 improved appetite, stabilized body weight, preserved fat and heart mass, and slightly protected lean mass after multiple cycles of chemotherapy. Furthermore, combining TCMCB07 with a growth differentiation factor 15 antibody enhanced treatment effectiveness. Similar effects from TCMCB07 treatment were observed in a rat tumor model following combination chemotherapy. No notable adverse effects nor increased chemotherapy-related toxicities were observed with TCMCB07 treatment. These findings suggest that peripheral administration of TCMCB07 holds promise as a therapeutic approach for alleviating chemotherapy-induced anorexia and weight loss, potentially benefiting numerous patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Xinxia Zhu
- Papé Family Pediatric Research Institute and
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, Oregon, USA
| | | | - Kenneth A Gruber
- Endevica Bio, Northbrook, Illinois, USA
- Department of Medical Pharmacology and Physiology and the Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | | | | | | | - Peter R Levasseur
- Papé Family Pediatric Research Institute and
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, Oregon, USA
| | - Peng Bai
- In Vivo Pharmacology Unit, WuXi App Tec, Nantong, Jiangsu, China
| | - Xu Chen
- In Vivo Pharmacology Unit, WuXi App Tec, Shanghai, China
| | - Qingyang Gu
- In Vivo Pharmacology Unit, WuXi App Tec, Shanghai, China
| | - Aaron J Grossberg
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, Oregon, USA
- Department of Radiation Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | | |
Collapse
|
15
|
Colarusso B, Ortiz R, Yeboah J, Chang A, Gupta M, Kulkarni P, Ferris CF. APOE4 rat model of Alzheimer's disease: sex differences, genetic risk and diet. BMC Neurosci 2024; 25:57. [PMID: 39506641 PMCID: PMC11539573 DOI: 10.1186/s12868-024-00901-z] [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: 04/29/2024] [Accepted: 09/24/2024] [Indexed: 11/08/2024] Open
Abstract
The strongest genetic risk factor for Alzheimer's disease (AD) is the ε4 allele of apolipoprotein E (ApoE ε4). A high fat diet also adds to the risk of dementia and AD. In addition, there are sex differences as women carriers have a higher risk of an earlier onset and rapid decline in memory than men. The present study looked at the effect of the genetic risk of ApoE ε4 together with a high fat/high sucrose diet (HFD/HSD) on brain function in male and female rats using magnetic resonance imaging. We hypothesized female carriers would present with deficits in cognitive behavior together with changes in functional connectivity as compared to male carriers. Four-month-old wildtype and human ApoE ε4 knock-in (TGRA8960), male and female Sprague Dawley rats were put on a HFD/HSD for four months. Afterwards they were imaged for changes in function using resting state BOLD functional connectivity. Images were registered to, and analyzed, using a 3D MRI rat atlas providing site-specific data on 173 different brain areas. Resting state functional connectivity showed male wildtype had greater connectivity between areas involved in feeding and metabolism while there were no differences between female and male carriers and wildtype females. The data were unexpected. The genetic risk was overshadowed by the diet. Male wildtype rats were most sensitive to the HFD/HSD presenting with a deficit in cognitive performance with enhanced functional connectivity in neural circuitry associated with food consumption and metabolism.
Collapse
Affiliation(s)
- Bradley Colarusso
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA
| | - Richard Ortiz
- Department of Psychology, Northern Illinois University, DeKalb, IL, 60115, USA
| | - Julian Yeboah
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA
| | - Arnold Chang
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA
| | - Megha Gupta
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA
| | - Praveen Kulkarni
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA
| | - Craig F Ferris
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA.
- Departments of Psychology and Pharmaceutical Sciences, Northeastern University, 125 NI Hall, 360 Huntington Ave, Boston, MA, 02115-5000, USA.
| |
Collapse
|
16
|
Thapa K, Ghimire B, Pokharel K, Cai M, Savontaus E, Rinne P. Hepatocyte-specific loss of melanocortin 1 receptor disturbs fatty acid metabolism and promotes adipocyte hypertrophy. Int J Obes (Lond) 2024; 48:1625-1637. [PMID: 39117851 PMCID: PMC11502480 DOI: 10.1038/s41366-024-01600-9] [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: 04/03/2024] [Revised: 07/17/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND/OBJECTIVES Melanocortins mediate their biological functions via five different melanocortin receptors (MC1R - MC5R). MC1R is expressed in the skin and leukocytes, where it regulates skin pigmentation and inflammatory responses. MC1R is also present in the liver and white adipose tissue, but its functional role in these tissues is unclear. This study aimed at determining the regulatory role of MC1R in fatty acid metabolism. METHODS Male recessive yellow (Mc1re/e) mice, a model of global MC1R deficiency, and male hepatocyte-specific MC1R deficient mice (Mc1r LKO) were fed a chow or Western diet for 12 weeks. The mouse models were characterized for body weight and composition, liver adiposity, adipose tissue mass and morphology, glucose metabolism and lipid metabolism. Furthermore, qPCR and RNA sequencing analyses were used to investigate gene expression profiles in the liver and adipose tissue. HepG2 cells and primary mouse hepatocytes were used to study the effects of pharmacological MC1R activation. RESULTS Chow- and Western diet-fed Mc1re/e showed increased liver weight, white adipose tissue mass and plasma triglyceride (TG) concentration compared to wild type mice. This phenotype occurred without significant changes in food intake, body weight, physical activity or glucose metabolism. Mc1r LKO mice displayed a similar phenotype characterized by larger fat depots, increased adipocyte hypertrophy and enhanced accumulation of TG in the liver and plasma. In terms of gene expression, markers of de novo lipogenesis, inflammation and apoptosis were upregulated in the liver of Mc1r LKO mice, while enzymes regulating lipolysis were downregulated in white adipose tissue of these mice. In cultured hepatocytes, selective activation of MC1R reduced ChREBP expression, which is a central transcription factor for lipogenesis. CONCLUSIONS Hepatocyte-specific loss of MC1R disturbs fatty acid metabolism in the liver and leads to an obesity phenotype characterized by enhanced adipocyte hypertrophy and TG accumulation in the liver and circulation.
Collapse
Affiliation(s)
- Keshav Thapa
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Drug Research Doctoral Programme (DRDP), University of Turku, Turku, Finland
| | - Bishwa Ghimire
- Institute for Molecular Medicine Finland (FIMM), HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medicity Research Laboratory, University of Turku, Turku, Finland
| | - Kisun Pokharel
- Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Minying Cai
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | - Eriika Savontaus
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Petteri Rinne
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.
- Turku Center for Disease Modeling, University of Turku, Turku, Finland.
| |
Collapse
|
17
|
Xing M, Li Y, Zhang Y, Zhou J, Ma D, Zhang M, Tang M, Ouyang T, Zhang F, Shi X, Sun J, Chen Z, Zhang WJ, Zhang S, Xie X. Paraventricular hypothalamic RUVBL2 neurons suppress appetite by enhancing excitatory synaptic transmission in distinct neurocircuits. Nat Commun 2024; 15:8939. [PMID: 39414808 PMCID: PMC11484884 DOI: 10.1038/s41467-024-53258-6] [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: 03/08/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
The paraventricular hypothalamus (PVH) is crucial for food intake control, yet the presynaptic mechanisms underlying PVH neurons remain unclear. Here, we show that RUVBL2 in the PVH is significantly reduced during energy deficit, and knockout (KO) of PVH RUVBL2 results in hyperphagic obesity in mice. RUVBL2-expressing neurons in the PVH (PVHRUVBL2) exert the anorexigenic effect by projecting to the arcuate hypothalamus, the dorsomedial hypothalamus, and the parabrachial complex. We further demonstrate that PVHRUVBL2 neurons form the synaptic connections with POMC and AgRP neurons in the ARC. PVH RUVBL2 KO impairs the excitatory synaptic transmission by reducing presynaptic boutons and synaptic vesicles near active zone. Finally, RUVBL2 overexpression in the PVH suppresses food intake and protects against diet induced obesity. Together, this study demonstrates an essential role for PVH RUVBL2 in food intake control, and suggests that modulation of synaptic plasticity could be an effective way to curb appetite and obesity.
Collapse
Affiliation(s)
- Mingming Xing
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Yang Li
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuqi Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Juemou Zhou
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Danting Ma
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Mengqi Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Minglei Tang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Ting Ouyang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Fumiao Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Xiaofeng Shi
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Jianyuan Sun
- University of Chinese Academy of Sciences, Beijing, 100049, China
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Weiping J Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Shuli Zhang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xiangyang Xie
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China.
| |
Collapse
|
18
|
Friedman MI, Sørensen TIA, Taubes G, Lund J, Ludwig DS. Trapped fat: Obesity pathogenesis as an intrinsic disorder in metabolic fuel partitioning. Obes Rev 2024; 25:e13795. [PMID: 38961319 DOI: 10.1111/obr.13795] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 05/24/2024] [Accepted: 06/13/2024] [Indexed: 07/05/2024]
Abstract
Our understanding of the pathophysiology of obesity remains at best incomplete despite a century of research. During this time, two alternative perspectives have helped shape thinking about the etiology of the disorder. The currently prevailing view holds that excessive fat accumulation results because energy intake exceeds energy expenditure, with excessive food consumption being the primary cause of the imbalance. The other perspective attributes the initiating cause of obesity to intrinsic metabolic defects that shift fuel partitioning from pathways for mobilization and oxidation to those for synthesis and storage. The resulting reduction in fuel oxidation and trapping of energy in adipose tissue drives a compensatory increase in energy intake and, under some conditions, a decrease in expenditure. This theory of obesity pathogenesis has historically garnered relatively less attention despite its pedigree. Here, we present an updated comprehensive formulation of the fuel partitioning theory, focused on evidence gathered over the last 80 years from major animal models of obesity showing a redirection of fuel fluxes from oxidation to storage and accumulation of excess body fat with energy intake equal to or even less than that of lean animals. The aim is to inform current discussions about the etiology of obesity and by so doing, help lay new foundations for the design of more efficacious approaches to obesity research, treatment and prevention.
Collapse
Affiliation(s)
| | - Thorkild I A Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Center for Childhood Health, Copenhagen, Denmark
| | | | - Jens Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - David S Ludwig
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
| |
Collapse
|
19
|
Lord MN, Noble EE. Hypothalamic cannabinoid signaling: Consequences for eating behavior. Pharmacol Res Perspect 2024; 12:e1251. [PMID: 39155548 PMCID: PMC11331011 DOI: 10.1002/prp2.1251] [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: 11/29/2023] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 08/20/2024] Open
Abstract
In parallel to the legalization of cannabis for both medicinal and recreational purposes, cannabinoid use has steadily increased over the last decade in the United States. Cannabinoids, such as tetrahydrocannabinol and anandamide, bind to the central cannabinoid-1 (CB1) receptor to impact several physiological processes relevant for body weight regulation, including appetite and energy expenditure. The hypothalamus integrates peripheral signals related to energy balance, houses several nuclei that orchestrate eating, and expresses the CB1 receptor. Herein we review literature to date concerning cannabinergic action in the hypothalamus with a specific focus on eating behaviors. We highlight hypothalamic areas wherein researchers have focused their attention, including the lateral, arcuate, paraventricular, and ventromedial hypothalamic nuclei, and interactions with the hormone leptin. This review serves as a comprehensive analysis of what is known about cannabinoid signaling in the hypothalamus, highlights gaps in the literature, and suggests future directions.
Collapse
Affiliation(s)
- Magen N. Lord
- Department of Nutritional SciencesUniversity of GeorgiaAthensGeorgiaUSA
| | - Emily E. Noble
- Department of Nutritional SciencesUniversity of GeorgiaAthensGeorgiaUSA
| |
Collapse
|
20
|
Fang P, She Y, Yu M, Yan J, Yu X, Zhao J, Jin Y, Min W, Shang W, Zhang Z. Novel hypothalamic pathways for metabolic effects of spexin. Pharmacol Res 2024; 208:107399. [PMID: 39245191 DOI: 10.1016/j.phrs.2024.107399] [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: 06/17/2024] [Revised: 08/22/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
One of the main underlying etiologies of type 2 diabetes (T2DM) is insulin resistance, which is most frequently caused by obesity. Notably, the deregulation of adipokine secretion from visceral adiposity has been identified as a crucial characteristic of type 2 diabetes and obesity. Spexin is an adipokine that is released by many different tissues, including white adipocytes and the glandular stomach, and is negatively connected with the state of energy storage. This peptide acts through GALR2/3 receptors to control a wide range of metabolic processes, including inflammation, browning, lipolysis, energy expenditure, and eating behavior. Specifically, spexin can enter the hypothalamus and regulate the hypothalamic melanocortin system, which in turn balances energy expenditure and food intake. This review examines recent advances and the underlying mechanisms of spexin in obesity and T2DM. In particular, we address a range of topics from basic research to clinical findings, such as an analysis of the possible function of spexin in the hypothalamic melanocortin response, which involves reducing energy intake and increasing energy expenditure while also enhancing insulin sensitivity and glucose tolerance. Gaining more insight into the mechanisms that underlie the spexin system's control over energy metabolism and homeostasis may facilitate the development of innovative treatment approaches that focus on combating obesity and diabetes.
Collapse
Affiliation(s)
- Penghua Fang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yuqing She
- Department of Endocrinology, Nanjing Pukou People's Hospital, Nanjing 211899, China
| | - Mei Yu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jing Yan
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xizhong Yu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Juan Zhao
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu Jin
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wen Min
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Wenbin Shang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Zhenwen Zhang
- Department of Endocrinology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China.
| |
Collapse
|
21
|
Værøy H, Skar-Fröding R, Hareton E, Fetissov SO. Possible roles of neuropeptide/transmitter and autoantibody modulation in emotional problems and aggression. Front Psychiatry 2024; 15:1419574. [PMID: 39381606 PMCID: PMC11458397 DOI: 10.3389/fpsyt.2024.1419574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/30/2024] [Indexed: 10/10/2024] Open
Abstract
The theoretical foundations of understanding psychiatric disorders are undergoing changes. Explaining behaviour and neuroendocrine cell communication leaning towards immunology represents a different approach compared to previous models for understanding complex central nervous system processes. One such approach is the study of immunoglobulins or autoantibodies, and their effect on peptide hormones in the neuro-endocrine system. In the present review, we provide an overview of the literature on neuropeptide/transmitter and autoantibody modulation in psychiatric disorders featuring emotional problems and aggression, including associated illness behaviour. Finally, we discuss the role of psycho-immunology as a growing field in the understanding of psychiatric disorders, and that modulation and regulation by IgG autoAbs represent a relatively new subcategory in psycho-immunology, where studies are currently being conducted.
Collapse
Affiliation(s)
- Henning Værøy
- R&D Department, Division of Mental Health Services, Akershus University Hospital, Lørenskog, Norway
| | - Regina Skar-Fröding
- R&D Department, Division of Mental Health Services, Akershus University Hospital, Lørenskog, Norway
| | - Elin Hareton
- Department of Multidiciplinary Laboratory Medicine and Medical Biochemistry, (TLMB), Akershus University Hospital, Lørenskog, Norway
| | - Sergueï O. Fetissov
- Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| |
Collapse
|
22
|
M JN, Bharadwaj D. The complex web of obesity: from genetics to precision medicine. Expert Rev Endocrinol Metab 2024; 19:403-418. [PMID: 38869356 DOI: 10.1080/17446651.2024.2365785] [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: 11/06/2023] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
INTRODUCTION Obesity is a growing public health concern affecting both children and adults. Since it involves both genetic and environmental components, the management of obesity requires both, an understanding of the underlying genetics and changes in lifestyle. The knowledge of obesity genetics will enable the possibility of precision medicine in anti-obesity medications. AREAS COVERED Here, we explore health complications and the prevalence of obesity. We discuss disruptions in energy balance as a symptom of obesity, examining evolutionary theories, its multi-factorial origins, and heritability. Additionally, we discuss monogenic and polygenic obesity, the converging biological pathways, potential pharmacogenomics applications, and existing anti-obesity medications - specifically focussing on the leptin-melanocortin and incretin pathways. Comparisons between childhood and adult obesity genetics are made, along with insights into structural variants, epigenetic changes, and environmental influences on epigenetic signatures. EXPERT OPINION With recent advancements in anti-obesity drugs, genetic studies pinpoint new targets and allow for repurposing existing drugs. This creates opportunities for genotype-informed treatment options. Also, lifestyle interventions can help in the prevention and treatment of obesity by altering the epigenetic signatures. The comparison of genetic architecture in adults and children revealed a significant overlap. However, more robust studies with diverse ethnic representation is required in childhood obesity.
Collapse
Affiliation(s)
- Janaki Nair M
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Dwaipayan Bharadwaj
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
23
|
Yu Z, Zhang T, Yang X, Xu B, Yu Z, An L, Xu T, Jing X, Wang Y, Lu M. Neuregulin4-ErbB4 signalling pathway is driven by electroacupuncture stimulation to remodel brown adipose tissue innervation. Diabetes Obes Metab 2024; 26:3880-3896. [PMID: 38951947 DOI: 10.1111/dom.15735] [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: 04/10/2024] [Revised: 05/25/2024] [Accepted: 06/05/2024] [Indexed: 07/03/2024]
Abstract
AIM To show that electroacupuncture stimulation (ES) remodels sympathetic innervation in brown adipose tissue (BAT) via the bone morphogenic protein 8B (BMP8B)-neuregulin 4 (NRG4)-ErbB4 axis, with somatotopic dependence. MATERIALS AND METHODS We established a high-fat diet (HFD) model with C57BL/6J mice to measure the thermogenesis and metabolism of BAT. In addition, the sympathetic nerve activity (SNA) was measured with the electrophysiological technique, and the immunostaining of c-Fos was used to detect the central nervous system sources of sympathetic outflows. Finally, the key role of the BMP8B-NRG4-ErbB4 axis was verified by peripheral specific antagonism of ErbB4. RESULTS ES at the forelimb and abdomen regions significantly up-regulate SNA, whereas ES at the hindlimb region has a limited regulatory effect on SNA but still partially restores HFD-induced BAT dysfunction. Mechanistically, ES at the forelimb and abdomen regions driving catecholaminergic signals in brown adipocytes depends on neural activities projected from the ventromedial nucleus of the hypothalamus (VMH) to the spinal cord intermediolateral column (IML). Notably, the peripheral suppression of ErbB4 in BAT inhibits the thermogenesis and metabolic function of BAT, as well as significantly hindering the SNA activation and metabolic benefits induced by ES. CONCLUSION These results suggest that ES appears to be an effective approach for remodeling sympathetic innervation in BAT, which is closely related to neuronal activity in the VMH and the NRG4-ErbB4 signaling pathway.
Collapse
Affiliation(s)
- Ziwei Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ting Zhang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xingyu Yang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bin Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhi Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Li An
- School of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tiancheng Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinyue Jing
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yaling Wang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mengjiang Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
24
|
Reynolds RP, Fan RR, Tinajero A, Luo X, Huen SC, Fujikawa T, Lee S, Lemoff A, Mountjoy KG, Elmquist JK. Alpha-melanocyte-stimulating hormone contributes to an anti-inflammatory response to lipopolysaccharide. Mol Metab 2024; 87:101986. [PMID: 38992428 PMCID: PMC11362619 DOI: 10.1016/j.molmet.2024.101986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024] Open
Abstract
OBJECTIVE During infection, metabolism and immunity react dynamically to promote survival through mechanisms that remain unclear. Pro-opiomelanocortin (POMC) cleavage products are produced and released in the brain and in the pituitary gland. One POMC cleavage product, alpha-melanocyte-stimulating hormone (α-MSH), is known to regulate food intake and energy expenditure and has anti-inflammatory effects. However, it is not known whether α-MSH is required to regulate physiological anti-inflammatory responses. We recently developed a novel mouse model with a targeted mutation in Pomc (Pomctm1/tm1 mice) to block production of all α-MSH forms which are required to regulate metabolism. To test whether endogenous α-MSH is required to regulate immune responses, we compared acute bacterial lipopolysaccharide (LPS)-induced inflammation between Pomctm1/tm1 and wild-type Pomcwt/wt mice. METHODS We challenged 10- to 14-week-old male Pomctm1/tm1 and Pomcwt/wt mice with single i.p. injections of either saline or low-dose LPS (100 μg/kg) and monitored immune and metabolic responses. We used telemetry to measure core body temperature (Tb), ELISA to measure circulating cytokines, corticosterone and α-MSH, and metabolic chambers to measure body weight, food intake, activity, and respiration. We also developed a mass spectrometry method to measure three forms of α-MSH produced in the mouse hypothalamus and pituitary gland. RESULTS LPS induced an exaggerated immune response in Pomctm1/tm1 compared to Pomcwt/wt mice. Both groups of mice were hypoactive and hypothermic following LPS administration, but Pomctm1/tm1 mice were significantly more hypothermic compared to control mice injected with LPS. Pomctm1/tm1 mice also had reduced oxygen consumption and impaired metabolic responses to LPS compared to controls. Pomctm1/tm1 mice had increased levels of key proinflammatory cytokines at 2 h and 4 h post LPS injection compared to Pomcwt/wt mice. Lastly, Pomcwt/wt mice injected with LPS compared to saline had increased total α-MSH in circulation 2 h post injection. CONCLUSIONS Our data indicate endogenous α-MSH contributes to the inflammatory immune responses triggered by low-dose LPS administration and suggest that targeting the melanocortin system could be a potential therapeutic for the treatment of sepsis or inflammatory disease.
Collapse
Affiliation(s)
- R P Reynolds
- Department of Internal Medicine, Center for Hypothalamic Research, Dallas, TX, USA
| | - R R Fan
- Department of Internal Medicine, Center for Hypothalamic Research, Dallas, TX, USA
| | - A Tinajero
- Department of Internal Medicine, Center for Hypothalamic Research, Dallas, TX, USA
| | - X Luo
- Department of Biochemistry, Dallas, TX, USA
| | - S C Huen
- Department of Internal Medicine (Nephrology) and Pharmacology, Dallas, TX, USA
| | - T Fujikawa
- Department of Internal Medicine, Center for Hypothalamic Research, Dallas, TX, USA; The Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - S Lee
- Department of Internal Medicine, Center for Hypothalamic Research, Dallas, TX, USA
| | - A Lemoff
- Department of Biochemistry, Dallas, TX, USA
| | - K G Mountjoy
- Department of Molecular Medicine and Pathology, University of Auckland, Private Bag 92019, Auckland 1043, New Zealand
| | - J K Elmquist
- Department of Internal Medicine, Center for Hypothalamic Research, Dallas, TX, USA; The Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
25
|
Farooqi IS, Xu Y. Translational potential of mouse models of human metabolic disease. Cell 2024; 187:4129-4143. [PMID: 39067442 DOI: 10.1016/j.cell.2024.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 07/30/2024]
Abstract
Obesity causes significant morbidity and mortality globally. Research in the last three decades has delivered a step-change in our understanding of the fundamental mechanisms that regulate energy homeostasis, building on foundational discoveries in mouse models of metabolic disease. However, not all findings made in rodents have translated to humans, hampering drug discovery in this field. Here, we review how studies in mice and humans have informed our current framework for understanding energy homeostasis, discuss their challenges and limitations, and offer a perspective on how human studies may play an increasingly important role in the discovery of disease mechanisms and identification of therapeutic targets in the future.
Collapse
Affiliation(s)
- I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Department of Molecular and Cellular Biology and Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
26
|
Godino-Gimeno A, Rocha A, Chivite M, Saera-Vila A, Rotllant J, Míguez JM, Cerdá-Reverter JM. Agouti-Induced Anxiety-Like Behavior Is Mediated by Central Serotonergic Pathways in Zebrafish. J Neurosci 2024; 44:e1970232024. [PMID: 38977301 PMCID: PMC11308329 DOI: 10.1523/jneurosci.1970-23.2024] [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: 10/30/2023] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/10/2024] Open
Abstract
Overexpression of the agouti-signaling protein (asip1), an endogenous melanocortin antagonist, under the control of a constitutive promoter in zebrafish [Tg(Xla.Eef1a1:Cau.Asip1]iim4] (asip1-Tg) increases food intake by reducing sensitivity of the central satiety systems and abolish circadian activity rhythms. The phenotype also shows increased linear growth and body weight, yet no enhanced aggressiveness in dyadic fights is observed. In fact, asip1-Tg animals choose to flee to safer areas rather than face a potential threat, thus suggesting a potential anxiety-like behavior (ALB). Standard behavioral tests, i.e., the open field test (OFT), the novel object test (NOT), and the novel tank dive test (NTDT), were used to investigate thigmotaxis and ALB in male and female zebrafish. Results showed that the asip1-Tg strain exhibited severe ALB in every test, mainly characterized by pronounced freezing behavior and increased linear and angular swimming velocities. asip1-Tg animals exhibited low central serotonin (5-HT) and dopamine (DA) levels and high turnover rates, thus suggesting that central monoaminergic pathways might mediate melanocortin antagonist-induced ALB. Accordingly, the treatment of asip1-Tg animals with fluoxetine, a selective serotonin reuptake inhibitor (SSRI), reversed the ALB phenotype in NTDT as well as 5-HT turnover. Genomic and anatomical data further supported neuronal interaction between melanocortinergic and serotonergic systems. These results suggest that inhibition of the melanocortin system by ubiquitous overexpression of endogenous antagonist has an anxiogenic effect mediated by serotonergic transmission.
Collapse
Affiliation(s)
- Alejandra Godino-Gimeno
- Fish NeuroBehaviour Lab, Department of Fish Physiology and Biotechnology, Instituto de Acuicultura de Torre de la Sal, IATS-CSIC, Castellon 12595, Spain
| | - Ana Rocha
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Terminal de Cruzeiros do Porto de Leixões, Matosinhos 4450-208, Portugal
| | - Mauro Chivite
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo 36310, Spain
| | | | - Josep Rotllant
- Instituto de Investigaciones Marinas, Consejo Superior de Investigaciones Científicas (IIM-CSIC), Vigo 36208, Spain
| | - Jesús M Míguez
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo 36310, Spain
| | - José Miguel Cerdá-Reverter
- Fish NeuroBehaviour Lab, Department of Fish Physiology and Biotechnology, Instituto de Acuicultura de Torre de la Sal, IATS-CSIC, Castellon 12595, Spain
| |
Collapse
|
27
|
Seitz J, Lahaye E, Andreani NA, Thomas B, Takhlidjt S, Chartrel N, Herpertz-Dahlmann B, Baines JF, Fetissov SO. Long-Term Dynamics of Serum α-MSH and α-MSH-Binding Immunoglobulins with a Link to Gut Microbiota Composition in Patients with Anorexia Nervosa. Neuroendocrinology 2024; 114:907-920. [PMID: 38852579 PMCID: PMC11460951 DOI: 10.1159/000539316] [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: 03/11/2024] [Accepted: 05/06/2024] [Indexed: 06/11/2024]
Abstract
INTRODUCTION Immunoglobulins (Ig) reactive with α-melanocyte-stimulating hormone (α-MSH), an anorexigenic neuropeptide, are present in humans and were previously associated with eating disorders. In this longitudinal study involving patients with anorexia nervosa (AN), we determined whether α-MSH in serum is bound to IgG and analyzed long-term dynamics of both α-MSH peptide and α-MSH-reactive Ig in relation to changes in BMI and gut microbiota composition. METHODS The study included 64 adolescents with a restrictive form of AN, whose serum samples were collected at hospital admission, discharge, and during a 1-year follow-up visit and 41 healthy controls, all females. RESULTS We found that in both study groups, approximately 40% of serum α-MSH was reversibly bound to IgG and that levels of α-MSH-reactive IgG but not of α-MSH peptide in patients with AN were low at hospital admission but recovered 1 year later. Total IgG levels were also low at admission. Moreover, BMI-standard deviation score correlated positively with α-MSH IgG in both groups studied but negatively with α-MSH peptide only in controls. Significant correlations between the abundance of specific bacterial taxa in the gut microbiota and α-MSH peptide and IgG levels were found in both study groups, but they were more frequent in controls. CONCLUSION We conclude that IgG in the blood plays a role as an α-MSH-binding protein, whose characteristics are associated with BMI in both patients with AN and controls. Furthermore, the study suggests that low production of α-MSH-reactive IgG during the starvation phase in patients with AN may be related to altered gut microbiota composition.
Collapse
Affiliation(s)
- Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, University Hospital, RWTH University Aachen, Aachen, Germany
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, LVR-University Hospital, Essen, Germany
| | - Emilie Lahaye
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Nadia Andrea Andreani
- Guest Group Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Section of Evolutionary Medicine, Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Benjamin Thomas
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Saloua Takhlidjt
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Nicolas Chartrel
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Beate Herpertz-Dahlmann
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, University Hospital, RWTH University Aachen, Aachen, Germany
| | - John F. Baines
- Guest Group Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Section of Evolutionary Medicine, Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Sergueï O. Fetissov
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| |
Collapse
|
28
|
Arrari F, Jabri MA, Ayari A, Dakhli N, Ben Fayala C, Boubaker S, Sebai H. Amino acid HPLC-FLD analysis of spirulina and its protective mechanism against the combination of obesity and colitis in wistar rats. Heliyon 2024; 10:e30103. [PMID: 38694088 PMCID: PMC11061748 DOI: 10.1016/j.heliyon.2024.e30103] [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: 09/07/2023] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/03/2024] Open
Abstract
Objective The cafeteria diet (CD), designed as an experimental diet mimicking the obesogenic diet, may contribute to the pathogenesis of inflammatory bowel diseases (IBD). This study delves into the influence of spirulina (SP) on obesity associated with colitis in Wistar rats. Methods The amino acids composition of SP was analyzed using HPLC-FLD. Animals were equally separated into eight groups, each containing seven animals and treated daily for eight weeks as follows: Control diet (SD), cafeteria diet (CD) group, CD + SP (500 mg/kg) and SD + SP. Ulcerative colitis was provoked by rectal injection of acetic acid (AA) (3 % v/v, 5 ml/kg b.w.) on the last day of treatment in the following groups: SD + AA, SD + AA + SP, CD + AA, and CD + AA + SP. Results Findings revealed that UC and/or CD increased the abdominal fat, weights gain, and colons. Moreover, severe colonic alteration, perturbations in the serum metabolic parameters associated with an oxidative stress state in the colonic mucosa, defined by overproduction of reactive oxygen species (ROS) and increased levels of plasma scavenging activity (PSA). Additionally, obesity exacerbated the severity of AA-induced UC promoting inflammation marked by the overexpression of pro-inflammatory cytokines. Significantly, treatment with SP provided notable protection against inflammation severity, reduced histopathological alterations, attenuated lipid peroxidation (MDA), and enhanced antioxidant enzyme activities (CAT, SOD, and GPX) along with non-enzymatic antioxidants (GSH and SH-G). Conclusions Thus, the antioxidant effects and anti-inflammatory proprieties of SP could be attributed to its richness in amino acids, which could potentially mitigate inflammation severity in obese subjects suffering from ulcerative colitis. These results imply that SP hold promise as a therapeutic agent for managing of UC, particularly in individuals with concomitant obesity. Understanding SP's mechanisms of action may lead novel treatment strategies for inflammatory bowel diseases and hyperlipidemia in medical research.
Collapse
Affiliation(s)
- Fatma Arrari
- Université de Jendouba, Institut Supérieur de Biotechnologie de Béja, LR: Physiologie Fonctionnelle et Valorisation des Bio-Ressources, 9000, Béja, Tunisia
| | - Mohamed-Amine Jabri
- Université de Jendouba, Institut Supérieur de Biotechnologie de Béja, LR: Physiologie Fonctionnelle et Valorisation des Bio-Ressources, 9000, Béja, Tunisia
| | - Ala Ayari
- Université de Jendouba, Institut Supérieur de Biotechnologie de Béja, LR: Physiologie Fonctionnelle et Valorisation des Bio-Ressources, 9000, Béja, Tunisia
| | - Nouha Dakhli
- Université de Jendouba, Institut Supérieur de Biotechnologie de Béja, LR: Physiologie Fonctionnelle et Valorisation des Bio-Ressources, 9000, Béja, Tunisia
| | - Chayma Ben Fayala
- Laboratoire d'anatomie Pathologique Humaine et Expérimentale, Institut Pasteur de Tunis, 13, Place Pasteur, Tunis, 1002, Tunisia
| | - Samir Boubaker
- Laboratoire d'anatomie Pathologique Humaine et Expérimentale, Institut Pasteur de Tunis, 13, Place Pasteur, Tunis, 1002, Tunisia
| | - Hichem Sebai
- Université de Jendouba, Institut Supérieur de Biotechnologie de Béja, LR: Physiologie Fonctionnelle et Valorisation des Bio-Ressources, 9000, Béja, Tunisia
| |
Collapse
|
29
|
Possa-Paranhos IC, Butts J, Pyszka E, Nelson C, Cho D, Sweeney P. Neuroanatomical dissection of the MC3R circuitry regulating energy rheostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590573. [PMID: 38712101 PMCID: PMC11071362 DOI: 10.1101/2024.04.22.590573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Although mammals resist both acute weight loss and weight gain, the neural circuitry mediating bi-directional defense against weight change is incompletely understood. Global constitutive deletion of the melanocortin-3-receptor (MC3R) impairs the behavioral response to both anorexic and orexigenic stimuli, with MC3R knockout mice demonstrating increased weight gain following anabolic challenges and increased weight loss following anorexic challenges (i.e. impaired energy rheostasis). However, the brain regions mediating this phenotype remain incompletely understood. Here, we utilized MC3R floxed mice and viral injections of Cre-recombinase to selectively delete MC3R from medial hypothalamus (MH) in adult mice. Behavioral assays were performed on these animals to test the role of MC3R in MH in the acute response to orexigenic and anorexic challenges. Complementary chemogenetic approaches were used in MC3R-Cre mice to localize and characterize the specific medial hypothalamic brain regions mediating the role of MC3R in energy homeostasis. Finally, we performed RNAscope in situ hybridization to map changes in the mRNA expression of MC3R, POMC, and AgRP following energy rheostatic challenges. Our results demonstrate that MC3R deletion in MH increased feeding and weight gain following acute high fat diet feeding in males, and enhanced the anorexic effects of semaglutide, in a sexually dimorphic manner. Additionally, activation of DMH MC3R neurons increased energy expenditure and locomotion. Together, these results demonstrate that MC3R mediated effects on energy rheostasis result from the loss of MC3R signaling in the medial hypothalamus of adult animals and suggest an important role for DMH MC3R signaling in energy rheostasis.
Collapse
Affiliation(s)
| | - Jared Butts
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology
- University of Illinois Urbana-Champaign Neuroscience Program
| | - Emma Pyszka
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology
| | - Christina Nelson
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology
| | - Dajin Cho
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology
- University of Illinois Urbana-Champaign Neuroscience Program
| | - Patrick Sweeney
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology
- University of Illinois Urbana-Champaign Neuroscience Program
| |
Collapse
|
30
|
Strnadová V, Pačesová A, Charvát V, Šmotková Z, Železná B, Kuneš J, Maletínská L. Anorexigenic neuropeptides as anti-obesity and neuroprotective agents: exploring the neuroprotective effects of anorexigenic neuropeptides. Biosci Rep 2024; 44:BSR20231385. [PMID: 38577975 PMCID: PMC11043025 DOI: 10.1042/bsr20231385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/26/2024] [Accepted: 04/05/2024] [Indexed: 04/06/2024] Open
Abstract
Since 1975, the incidence of obesity has increased to epidemic proportions, and the number of patients with obesity has quadrupled. Obesity is a major risk factor for developing other serious diseases, such as type 2 diabetes mellitus, hypertension, and cardiovascular diseases. Recent epidemiologic studies have defined obesity as a risk factor for the development of neurodegenerative diseases, such as Alzheimer's disease (AD) and other types of dementia. Despite all these serious comorbidities associated with obesity, there is still a lack of effective antiobesity treatment. Promising candidates for the treatment of obesity are anorexigenic neuropeptides, which are peptides produced by neurons in brain areas implicated in food intake regulation, such as the hypothalamus or the brainstem. These peptides efficiently reduce food intake and body weight. Moreover, because of the proven interconnection between obesity and the risk of developing AD, the potential neuroprotective effects of these two agents in animal models of neurodegeneration have been examined. The objective of this review was to explore anorexigenic neuropeptides produced and acting within the brain, emphasizing their potential not only for the treatment of obesity but also for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Veronika Strnadová
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Andrea Pačesová
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Vilém Charvát
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Zuzana Šmotková
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Blanka Železná
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Jaroslav Kuneš
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
- Department of Biochemistry and Molecular Biology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Lenka Maletínská
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
31
|
Ericson MD, Freeman KT, Larson CM, Bouchard JL, John K, Lunzer MM, Koerperich ZM, Haskell-Luevano C. Incorporation of Three Extracyclic Arginine Residues into a Melanocortin Macrocyclic Agonist (c[Pro-His-DPhe-Arg-Trp-Dap-Lys(Arg-Arg-Arg-Ac)-DPro]) Decreases Food Intake When Administered Intrathecally or Subcutaneously Compared to a Macrocyclic Ligand Lacking Extracyclic Arginine Residues (c[Pro-His-DPhe-Arg-Trp-Dap-Ala-DPro)]. ACS Pharmacol Transl Sci 2024; 7:1114-1125. [PMID: 38633589 PMCID: PMC11020072 DOI: 10.1021/acsptsci.4c00011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 04/19/2024]
Abstract
Of the three Food and Drug Administration-approved melanocortin peptide drugs, two possess a cyclic scaffold, demonstrating that cyclized melanocortin peptides have therapeutic relevance. An extracyclic Arg residue, critical for pharmacological activity in the approved melanocortin cyclic drug setmelanotide, has also been demonstrated to increase the signal when fluorescently labeled cell-penetrating cyclic peptides are incubated with HeLa cells, with the maximal signal observed with three extracyclic Arg amino acids. Herein, a branching Lys residue was substituted into two macrocyclic melanocortin peptide agonists to incorporate 0-3 extracyclic Arg amino acids. Incorporation of the Arg residues resulted in equipotent or increased agonist potency at the mouse melanocortin receptors in vitro, suggesting that these substitutions were tolerated in the macrocyclic scaffolds. Further in vivo evaluation of one parent ligand (c[Pro-His-DPhe-Arg-Trp-Dap-Ala-Pro]) and the three Arg derivative (c[Pro-His-DPhe-Arg-Trp-Dap-Lys(Ac-Arg-Arg-Arg)-Pro)] demonstrated that the three Arg derivative further decreased food intake compared to the parent macrocycle when the compounds were administered either via intrathecal injection or subcutaneous dosing. This suggests that three extracyclic Arg amino acids may be beneficial in the design of cyclic melanocortin ligands and that in vitro pharmacological profiling may not predict the in vivo efficacy of melanocortin ligands.
Collapse
Affiliation(s)
- Mark D. Ericson
- Department of Medicinal Chemistry, Institute for Translational Neuroscience, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Katie T. Freeman
- Department of Medicinal Chemistry, Institute for Translational Neuroscience, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Courtney M. Larson
- Department of Medicinal Chemistry, Institute for Translational Neuroscience, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jacob L. Bouchard
- Department of Medicinal Chemistry, Institute for Translational Neuroscience, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Kristen John
- Department of Medicinal Chemistry, Institute for Translational Neuroscience, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mary M. Lunzer
- Department of Medicinal Chemistry, Institute for Translational Neuroscience, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Zoe M. Koerperich
- Department of Medicinal Chemistry, Institute for Translational Neuroscience, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Carrie Haskell-Luevano
- Department of Medicinal Chemistry, Institute for Translational Neuroscience, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
32
|
Borgmann D, Fenselau H. Vagal pathways for systemic regulation of glucose metabolism. Semin Cell Dev Biol 2024; 156:244-252. [PMID: 37500301 DOI: 10.1016/j.semcdb.2023.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 06/20/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023]
Abstract
Maintaining blood glucose at an appropriate physiological level requires precise coordination of multiple organs and tissues. The vagus nerve bidirectionally connects the central nervous system with peripheral organs crucial to glucose mobilization, nutrient storage, and food absorption, thereby presenting a key pathway for the central control of blood glucose levels. However, the precise mechanisms by which vagal populations that target discrete tissues participate in glucoregulation are much less clear. Here we review recent advances unraveling the cellular identity, neuroanatomical organization, and functional contributions of both vagal efferents and vagal afferents in the control of systemic glucose metabolism. We focus on their involvement in relaying glucoregulatory cues from the brain to peripheral tissues, particularly the pancreatic islet, and by sensing and transmitting incoming signals from ingested food to the brain. These recent findings - largely driven by advances in viral approaches, RNA sequencing, and cell-type selective manipulations and tracings - have begun to clarify the precise vagal neuron populations involved in the central coordination of glucose levels, and raise interesting new possibilities for the treatment of glucose metabolism disorders such as diabetes.
Collapse
Affiliation(s)
- Diba Borgmann
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Physical Activity Research (CFAS), Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Henning Fenselau
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50937 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany.
| |
Collapse
|
33
|
Xu B, Yao J, Song W, Yan X, Zhu M, Li J, Ma Z, Li Y, Li Y, Fu Y, Liu L, Li L, Lyu J, Zhang C. Evolutionary Identification of the Requirement of the Second Intracellular Loop for the Constitutive Activity of Melanocortin-4 Receptors. ACS Pharmacol Transl Sci 2024; 7:630-640. [PMID: 38481681 PMCID: PMC10928900 DOI: 10.1021/acsptsci.3c00169] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/19/2024] [Accepted: 02/02/2024] [Indexed: 02/17/2025]
Abstract
Melanocortin-4 receptor (MC4R) functions as a crucial neuroendocrine G protein-coupled receptor (GPCR) in the central nervous system of mammals, displaying agonist-independent constitutive activity that is mainly determined by its N-terminal domain. We previously reported that zebrafish MC4R exhibited a much higher basal cAMP level in comparison to mammalian MC4Rs. However, the functional evolution of constitutive activities in chordate MC4Rs remains to be elucidated. Here we cloned and compared the constitutive activities of MC4Rs from nine vertebrate species and showed that the additive action of the N-terminus with the extracellular region or transmembrane domain exhibited a combined pharmacological effect on the MC4R constitutive activity. In addition, we demonstrated that four residues of F149, Q156, V163, and K164 of the second intracellular loop played a vital role in determining MC4R constitutive activity. This study provided novel insights into functional evolution and identified a key motif essential for constitutive modulation of MC4R signaling.
Collapse
Affiliation(s)
- Bingxin Xu
- Fundamental
Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai
Sunshine Rehabilitation Center), School of Life Sciences and Technology, Tongji University, Shanghai 201619, P.R. China
| | - Jindong Yao
- Fundamental
Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai
Sunshine Rehabilitation Center), School of Life Sciences and Technology, Tongji University, Shanghai 201619, P.R. China
| | - Wenqi Song
- Fundamental
Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai
Sunshine Rehabilitation Center), School of Life Sciences and Technology, Tongji University, Shanghai 201619, P.R. China
| | - Xinyi Yan
- Fundamental
Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai
Sunshine Rehabilitation Center), School of Life Sciences and Technology, Tongji University, Shanghai 201619, P.R. China
| | - Ming Zhu
- Fundamental
Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai
Sunshine Rehabilitation Center), School of Life Sciences and Technology, Tongji University, Shanghai 201619, P.R. China
| | - Jiangtao Li
- State
Key Laboratory of Marine Geology, School of Ocean and Earth Science, Tongji University, Shanghai 200092, P.R. China
| | - Zhonglin Ma
- State
Key Laboratory of Marine Geology, School of Ocean and Earth Science, Tongji University, Shanghai 200092, P.R. China
| | - Yanchuan Li
- Hubei
Topgene Research Institute of Hubei Topgene Biotechnology Co., Ltd., East Lake High-Tech Development
Zone, Wuhan 430205, P.R. China
| | - Yihao Li
- Hubei
Topgene Research Institute of Hubei Topgene Biotechnology Co., Ltd., East Lake High-Tech Development
Zone, Wuhan 430205, P.R. China
| | - Yanbin Fu
- Shanghai
Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University
School of Medicine, Shanghai 200127, P.R. China
| | - Liu Liu
- Shanghai
Yuhui Pharmaceutical Technology (Group) Co., Ltd., and Shanghai Ruishen
Technology Development Co., Ltd., Shanghai 201203, P.R. China
| | - Lei Li
- Department
of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Jianjun Lyu
- Hubei
Topgene Research Institute of Hubei Topgene Biotechnology Co., Ltd., East Lake High-Tech Development
Zone, Wuhan 430205, P.R. China
| | - Chao Zhang
- Fundamental
Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai
Sunshine Rehabilitation Center), School of Life Sciences and Technology, Tongji University, Shanghai 201619, P.R. China
| |
Collapse
|
34
|
Inyang I, White HE, Timme K, Keating AF. Biological sex differences in hepatic response to in utero dimethylbenz(a)anthracene exposure. Reprod Toxicol 2024; 124:108553. [PMID: 38307155 DOI: 10.1016/j.reprotox.2024.108553] [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: 09/19/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/04/2024]
Abstract
Fetal hepatic dimethylbenz(a)anthracene (DMBA) biotransformation is not defined, thus, this study investigated whether the fetal liver metabolizes DMBA and differs with biological sex. KK.Cg-a/a (lean; n = 20) or KK.Cg-Ay/J (obese; n = 20) pregnant mice were exposed to corn oil (CT) or DMBA (1 mg/kg bw/day) by intraperitoneal injection (n = 10/treatment) from gestation day 7-14. Postnatal day 2 male or female offspring livers were collected. Total RNA (n = 6) and protein (n = 6) were analyzed via a PCR-based array or LC-MS/MS, respectively. The level of Mgst3 was lower (P < 0.05) in livers of female compared to male offspring. Furthermore, in utero DMBA exposure increased (P < 0.1) Cyp2c29 and Gpx3 levels (P < 0.05) in female offspring. In male offspring, the abundance of Ahr, Comt (P < 0.1), Alox5, and Asna1 (P < 0.05) decreased due to DMBA exposure. Female and male offspring had 34 and 21 hepatic proteins altered (P < 0.05) by in utero DMBA exposure, respectively. Opposing patterns for hepatic CD81 and KRT78 occurred, being decreased in females but increased in males, while YWHAG was decreased by DMBA exposure in both. Functional KEGG pathway analysis identified enrichment of 26 and 13 hepatic metabolic proteins in male and female offspring, respectively, due to in utero DMBA exposure. In silico transcription factor analysis of differentially expressed proteins predicted involvement of female NRF1 but male AHR. Thus, hepatic biological sex differences and capacity to respond to toxicants in utero are supported.
Collapse
Affiliation(s)
| | - Hunter E White
- Department of Animal Science, Iowa State University, USA
| | - Kelsey Timme
- Department of Animal Science, Iowa State University, USA
| | | |
Collapse
|
35
|
Meng W, Lin Z, Bian T, Chen X, Meng L, Yuan T, Niu L, Zheng H. Ultrasound Deep Brain Stimulation Regulates Food Intake and Body Weight in Mice. IEEE Trans Neural Syst Rehabil Eng 2024; 32:366-377. [PMID: 38194393 DOI: 10.1109/tnsre.2024.3351312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Given the widespread occurrence of obesity, new strategies are urgently needed to prevent, halt and reverse this condition. We proposed a noninvasive neurostimulation tool, ultrasound deep brain stimulation (UDBS), which can specifically modulate the hypothalamus and effectively regulate food intake and body weight in mice. Fifteen-min UDBS of hypothalamus decreased 41.4% food intake within 2 hours. Prolonged 1-hour UDBS significantly decreased daily food intake lasting 4 days. UDBS also effectively restrained body weight gain in leptin-receptor knockout mice (Sham: 96.19%, UDBS: 58.61%). High-fat diet (HFD) mice treated with 4-week UDBS (15 min / 2 days) reduced 28.70% of the body weight compared to the Sham group. Meanwhile, UDBS significantly modulated glucose-lipid metabolism and decreased the body fat. The potential mechanism is that ultrasound actives pro-opiomelanocortin (POMC) neurons in the hypothalamus for reduction of food intake and body weight. These results provide a noninvasive tool for controlling food intake, enabling systematic treatment of obesity.
Collapse
|
36
|
Sun W, Jin T, Yang H, Li J, Tian Q, Gao J, Peng R, Zhang G, Zhang X. Alterations of serum neuropeptide levels and their relationship to cognitive impairment and psychopathology in male patients with chronic schizophrenia. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:3. [PMID: 38172494 PMCID: PMC10851704 DOI: 10.1038/s41537-023-00425-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024]
Abstract
Serum neuropeptide levels may be linked to schizophrenia (SCZ) pathogenesis. This study aims to examine the relation between five serum neuropeptide levels and the cognition of patients with treatment-resistant schizophrenia (TRS), chronic stable schizophrenia (CSS), and in healthy controls (HC). Three groups were assessed: 29 TRS and 48 CSS patients who were hospitalized in regional psychiatric hospitals, and 53 HC. After the above participants were enrolled, we examined the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) and the blood serum levels of α-melanocyte stimulating hormone (α-MSH), β-endorphin (BE), neurotensin (NT), oxytocin (OT) and substance.P (S.P). Psychiatric symptoms in patients with SCZ were assessed with the Positive and Negative Syndrome Scale. SCZ patients performed worse than HC in total score and all subscales of the RBANS. The levels of the above five serum neuropeptides were significantly higher in SCZ than in HC. The levels of OT and S.P were significantly higher in CSS than in TRS patients. The α-MSH levels in TRS patients were significantly and negatively correlated with the language scores of RBANS. However, the BE and NT levels in CSS patients were significantly and positively correlated with the visuospatial/constructional scores of RBANS. Moreover, the interaction effect of NT and BE levels was positively associated with the visuospatial/constructional scores of RBANS. Therefore, abnormally increased serum neuropeptide levels may be associated with the physiology of SCZ, and may cause cognitive impairment and psychiatric symptoms, especially in patients with TRS.
Collapse
Affiliation(s)
- Wenxi Sun
- Suzhou Medical College of Soochow University, Suzhou, 215031, Jiangsu, China
- Psychiatry Department of Suzhou Guangji Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China
| | - Tingting Jin
- Psychiatry Department of Suzhou Guangji Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China
| | - Haidong Yang
- Department of Psychiatry, The Fourth People's Hospital of Lianyungang, The Affiliated KangDa College of Nanjing Medical University, Lianyungang, 222003, PR China
| | - Jin Li
- Psychiatry Department of Suzhou Guangji Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China
| | - Qing Tian
- Psychiatry Department of Suzhou Guangji Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China
| | - Ju Gao
- Psychiatry Department of Suzhou Guangji Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China
| | - Ruijie Peng
- Suzhou Medical College of Soochow University, Suzhou, 215031, Jiangsu, China
| | - Guangya Zhang
- Psychiatry Department of Suzhou Guangji Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China.
| | - Xiaobin Zhang
- Psychiatry Department of Suzhou Guangji Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China.
| |
Collapse
|
37
|
Hernandez CC, Gimenez LE, Cone RD. Automated Patch Clamp Recordings of GPCR-Gated Ion Channels: Targeting the MC4-R/Kir7.1 Potassium Channel Complex. Methods Mol Biol 2024; 2796:229-248. [PMID: 38856905 DOI: 10.1007/978-1-0716-3818-7_14] [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] [Indexed: 06/11/2024]
Abstract
Automated patch clamp recording is a valuable technique in drug discovery and the study of ion channels. It allows for the precise measurement and manipulation of channel currents, providing insights into their function and modulation by drugs or other compounds. The melanocortin 4 receptor (MC4-R) is a G protein-coupled receptor (GPCR) crucial to appetite regulation, energy balance, and body weight. MC4-R signaling is complex and involves interactions with other receptors and neuropeptides in the appetite-regulating circuitry. MC4-Rs, like other GPCRs, are known to modulate ion channels such as Kir7.1, an inward rectifier potassium channel, in response to ligand binding. This modulation is critical for controlling ion flow across the cell membrane, which can influence membrane potential, excitability, and neurotransmission. The MC4-R is the target for the anti-obesity drug Imcivree. However, this drug is known to lack optimal potency and also has side effects. Using high-throughput techniques for studying the MC4-R/Kir7.1 complex allows researchers to rapidly screen many compounds or conditions, aiding the development of drugs that target this system. Additionally, automated patch clamp recording of this receptor-channel complex and its ligands can provide valuable functional and pharmacological insights supporting the development of novel therapeutic strategies. This approach can be generalized to other GPCR-gated ion channel functional complexes, potentially accelerating the pace of research in different fields with the promise to uncover previously unknown aspects of receptor-ion channel interactions.
Collapse
Affiliation(s)
- Ciria C Hernandez
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
- Center for Chemical Genomics, Life Sciences Institute, University of Michigan, Ann Arbor, MI, 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
| |
Collapse
|
38
|
Zimmermann A, Böge N, Schuster K, Staffeld A, Lang S, Gill S, Rupprecht H, Frintrop L. Glial cell changes in the corpus callosum in chronically-starved mice. J Eat Disord 2023; 11:227. [PMID: 38111061 PMCID: PMC10726510 DOI: 10.1186/s40337-023-00948-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 12/04/2023] [Indexed: 12/20/2023] Open
Abstract
Anorexia nervosa (AN) is characterized by emaciation, hyperactivity, and amenorrhea. Imaging studies in AN patients have revealed reductions in grey and white matter volume, which correlate with the severity of neuropsychological deficits. However, the cellular basis for the observed brain atrophy is poorly understood. Although distinct hypothalamic centers, including the arcuate nucleus (ARC) are critically involved in regulating feeding behavior, little is known about potential hypothalamic modifications in this disorder. Since glia e.g. astrocytes and microglia influence neuronal circuits, we investigated the glial changes underlying pathophysiology of starvation in the corpus callosum (CC) and hypothalamus. Female mice were given a limited amount of food once a day and had unlimited access to a running wheel until a 20% weight reduction was achieved (acute starvation). This weight reduction was maintained for two weeks to mimic chronic starvation. Immunohistochemistry was used to quantify the density of astrocytes, microglia, oligodendrocytes, and the staining intensity of neuropeptide Y (NPY), a potent orexigenic peptide. Chronic starvation induced a decreased density of OLIG2+ oligodendrocytes, GFAP+ astrocytes, and IBA1+ microglia in the CC. However, the densities of glial cells remained unchanged in the ARC following starvation. Additionally, the staining intensity of NPY increased after both acute and chronic starvation, indicating an increased orexigenic signaling. Chronic starvation induced glial cell changes in the CC in a mouse model of AN suggesting that glia pathophysiology may play a role in the disease.
Collapse
Affiliation(s)
- Annelie Zimmermann
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstr. 9, 18057, Rostock, Germany
| | - Natalie Böge
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstr. 9, 18057, Rostock, Germany
| | - Katharina Schuster
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstr. 9, 18057, Rostock, Germany
| | - Anna Staffeld
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstr. 9, 18057, Rostock, Germany
| | - Stephan Lang
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstr. 9, 18057, Rostock, Germany
| | - Sadaf Gill
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstr. 9, 18057, Rostock, Germany
| | - Hanna Rupprecht
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstr. 9, 18057, Rostock, Germany
| | - Linda Frintrop
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstr. 9, 18057, Rostock, Germany.
| |
Collapse
|
39
|
Tian Y, Wang R, Liu L, Zhang W, Liu H, Jiang L, Jiang Y. The regulatory effects of the apelin/APJ system on depression: A prospective therapeutic target. Neuropeptides 2023; 102:102382. [PMID: 37716179 DOI: 10.1016/j.npep.2023.102382] [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: 06/08/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 09/18/2023]
Abstract
Depression is a debilitating neuropsychological disorder characterized by high incidence, high recurrence, high suicide, and high disability rates, which poses serious threats to human health and imposes heavy psychological and economic burdens on family and society. The pathogenesis of depression is extremely complex, and its etiology is multifactorial. Mounting evidence suggests that apelin and apelin receptor APJ, which compose the apelin/APJ system, are related to the development of depression. However, the specific mechanism is still unclear, and research in this area in human is still insufficient. Acceleration of research into the regulatory effects and underlying mechanisms of the apelin/APJ system in depression may identify attractive therapeutic targets and contribute to the development of novel intervention strategies against this devastating psychological disorder. In this review, we mainly discuss the regulatory effects of apelin/APJ system on depression and its potential therapeutic applications.
Collapse
Affiliation(s)
- Yanjun Tian
- Medical Laboratory of Jining Medical University, Jining Medical University, Jining 272067, China
| | - Ruihao Wang
- School of Mental Health, Jining Medical University, Jining 272067, China
| | - Lin Liu
- School of Mental Health, Jining Medical University, Jining 272067, China
| | - Wenhuan Zhang
- School of Mental Health, Jining Medical University, Jining 272067, China
| | - Haiqing Liu
- Department of Physiology, School of Basic Medical Sciences (Institute of Basic Medical Sciences), Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250024, China
| | - Liqing Jiang
- Department of Clinical Laboratory, Affiliated Hospital of Jining Medical University, Jining, China.
| | - Yunlu Jiang
- School of Mental Health, Jining Medical University, Jining 272067, China.
| |
Collapse
|
40
|
Bloyd M, Sinaii N, Faucz FR, Iben J, Coon SL, Caprio S, Santoro N, Stratakis CA, London E. High-frequency variants in PKA signaling-related genes within a large pediatric cohort with obesity or metabolic abnormalities. Front Endocrinol (Lausanne) 2023; 14:1272939. [PMID: 38027204 PMCID: PMC10679389 DOI: 10.3389/fendo.2023.1272939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/05/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Pediatric obesity has steadily increased in recent decades. Large-scale genome-wide association studies (GWAS) conducted primarily in Eurocentric adult populations have identified approximately 100 loci that predispose to obesity and type II diabetes. GWAS in children and individuals of non-European descent, both disproportionately affected by obesity, are fewer. Rare syndromic and monogenic obesities account for only a small portion of childhood obesity, so understanding the role of other genetic variants and their combinations in heritable obesities is key to developing targeted and personalized therapies. Tight and responsive regulation of the cAMP-dependent protein kinase (PKA) signaling pathway is crucial to maintaining healthy energy metabolism, and mutations in PKA-linked genes represent the most common cause of monogenic obesity. Methods For this study, we performed targeted exome sequencing of 53 PKA signaling-related genes to identify variants in genomic DNA from a large, ethnically diverse cohort of obese or metabolically challenged youth. Results We confirmed 49 high-frequency variants, including a novel variant in the PDE11A gene (c.152C>T). Several other variants were associated with metabolic characteristics within ethnic groups. Discussion We conclude that a PKA pathway-specific variant search led to the identification of several new genetic associations with obesity in an ethnically diverse population.
Collapse
Affiliation(s)
- Michelle Bloyd
- Section on Endocrinology and Genetics, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - Ninet Sinaii
- Biostatistics and Clinical Epidemiology Service, National Institutes of Health (NIH) Clinical Center, Bethesda, MD, United States
| | - Fabio Rueda Faucz
- Section on Endocrinology and Genetics, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - James Iben
- Molecular Genomics Core, National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - Steven L. Coon
- Molecular Genomics Core, National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - Sonia Caprio
- Section on Pediatric Endocrinology and Diabetes, Yale University, New Haven, CT, United States
| | - Nicola Santoro
- Section on Pediatric Endocrinology and Diabetes, Yale University, New Haven, CT, United States
- Department of Medicine and Health Sciences, “V. Tiberio” University of Molise, Campobasso, Italy
| | - Constantine A. Stratakis
- Section on Endocrinology and Genetics, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
- Human Genetics and Precision Medicine, Institute for Molecular Biology and Biotechnology (IMBB), Foundation for Research & Technology Hellas (FORTH), Heraklion, ELPEN Research Institute, Athens, Greece
| | - Edra London
- Section on Endocrinology and Genetics, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| |
Collapse
|
41
|
Kokkorakis M, Katsarou A, Katsiki N, Mantzoros CS. Milestones in the journey towards addressing obesity; Past trials and triumphs, recent breakthroughs, and an exciting future in the era of emerging effective medical therapies and integration of effective medical therapies with metabolic surgery. Metabolism 2023; 148:155689. [PMID: 37689110 DOI: 10.1016/j.metabol.2023.155689] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/11/2023]
Abstract
The 21st century is characterized by an increasing incidence and prevalence of obesity and the burden of its associated comorbidities, especially cardiometabolic diseases, which are reaching pandemic proportions. In the late '90s, the "black box" of adipose tissue and energy homeostasis was opened with the discovery of leptin, transforming the adipose tissue from an "inert fat-storage organ" to the largest human endocrine organ and creating the basis on which more intensified research efforts to elucidate the pathogenesis of obesity and develop novel treatments were based upon. Even though leptin was eventually not proven to be the "standalone magic bullet" for the treatment of common/polygenic obesity, it has been successful in the treatment of monogenic obesity syndromes. Additionally, it shifted the paradigm of treating obesity from a condition due to "lack of willpower" to a disease due to distinct underlying biological mechanisms for which specific pharmacotherapies would be needed in addition to lifestyle modification. Subsequently, the melanocortin pathway proved to be an equally valuable pathway for the pharmacotherapy of obesity. Melanocortin receptor agonists have recently been approved for treating certain types of syndromic obesity. Other molecules- such as incretins, implicated in energy and glucose homeostasis- are secreted by the gastrointestinal tract. Glucagon-like peptide 1 (GLP-1) is the most prominent one, with GLP-1 analogs approved for common/polygenic obesity. Unimolecular combinations with other incretins, e.g., GLP-1 with gastric inhibitory polypeptide and/or glucagon, are expected to be approved soon as more effective pharmacotherapies for obesity and its comorbidities. Unimolecular combinations with other compounds and small molecules activating the receptors of these molecules are currently under investigation as promising future pharmacotherapies. Moreover, metabolic and bariatric surgery has also demonstrated impressive results, especially in the case of morbid obesity. Consequently, this broadening therapeutic armamentarium calls for a well-thought-after and well-coordinated multidisciplinary approach, for instance, through cardiometabolic expertise centers, that would ideally address effectively and cost-effectively obesity and its comorbidities, providing tangible benefits to large segments of the population.
Collapse
Affiliation(s)
- Michail Kokkorakis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Angeliki Katsarou
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Niki Katsiki
- Department of Nutritional Sciences and Dietetics, International Hellenic University, 57400 Thessaloniki, Greece
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
42
|
Farooqi S. Obesity and thinness: insights from genetics. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220205. [PMID: 37661743 PMCID: PMC10475868 DOI: 10.1098/rstb.2022.0205] [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: 03/15/2023] [Accepted: 06/09/2023] [Indexed: 09/05/2023] Open
Abstract
Genetic disruption of key molecular components of the hypothalamic leptin-melanocortin pathway causes severe obesity in mice and humans. Physiological studies in people who carry these mutations have shown that the adipose tissue-derived hormone leptin primarily acts to defend against starvation. A lack of leptin causes an intense drive to eat and increases the rewarding properties of food, demonstrating that human appetite has a strong biological basis. Genetic studies in clinical- and population-based cohorts of people with obesity or thinness continue to provide new insights into the physiological mechanisms involved in weight regulation and identify molecular targets for weight loss therapy. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part II)'.
Collapse
Affiliation(s)
- Sadaf Farooqi
- Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Box 289, Cambridge CB2 0QQ, UK
| |
Collapse
|
43
|
Kim EM, Quinn JG, Reid RE, O'Hare E. Evidence for a feeding related association between melanocortin in the NTS and Neuropeptide-Y in the PVN. Appetite 2023; 188:106618. [PMID: 37257508 DOI: 10.1016/j.appet.2023.106618] [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: 03/20/2023] [Revised: 05/26/2023] [Accepted: 05/27/2023] [Indexed: 06/02/2023]
Abstract
Melanocortin and neuropeptide-Y (NPY) are both involved in feeding and energy regulation, and they have opposite effects in the paraventricular nucleus of the hypothalamus (PVN). The present study examined an interaction between melanocortin in the nucleus of the solitary tract (NTS) and NPY in the PVN. Male Sprague-Dawley rats were implanted with cannulae in the injection sites of interest. In Experiment 1, subjects received either the melanocortin 3/4-receptor (MC3/4) antagonist SHU9119 (0, 10, 50 and 100 pmol/0.5 μl) or the MC3/4 agonist MTII (0, 10, 50, 100 and 200 pmol/0.5 μl) into the NTS. Food intake was measured at 1, 2, 4, 6 and 24-h post-injection. Administration of SHU9119 into the NTS significantly and dose-dependently increased food intake at 1, 2, 4, 6 and 6-24-h, and administration of MTII into the NTS significantly and dose-dependently decreased 24-h free feeding. In Experiment 2, subjects received the MC3/4 agonist MTII (0, 10, 50, 100 and 200 pmol/0.5 μl) into the NTS just prior to NPY (0 and 1μg/0.5 μl) in the PVN. PVN injection of NPY stimulated feeding, and administration of MTII (50, 100 and 200 pmol) into the NTS significantly and dose-dependently decreased NPY-induced feeding at 2, 4, 6 and 6-24-h. These data suggest that there could be a neuronal association between melanocortin in the NTS and NPY in the PVN, and that the melanocortin system in the NTS has an antagonistic effect on NPY-induced feeding in the PVN.
Collapse
Affiliation(s)
- E-M Kim
- School of Psychology, Ulster University, Cromore Road, Coleraine, Northern Ireland, UK.
| | - J G Quinn
- School of Medicine, Queen's University Belfast, Northern Ireland, UK
| | - R E Reid
- School of Psychology, Dublin Business School, Ireland
| | - E O'Hare
- School of Psychology, Queen's University Belfast, Northern Ireland, UK
| |
Collapse
|
44
|
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: 24] [Impact Index Per Article: 12.0] [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
|
45
|
Coogan M, Xing D, Su B, Alston V, Johnson A, Khan M, Khalil K, Elaswad A, Li S, Wang J, Lu C, Wang W, Hettiarachchi D, Shang M, Hasin T, Qin Z, Cone R, Butts IAE, Dunham RA. CRISPR/Cas9-mediated knock-in of masu salmon (Oncorhyncus masou) elongase gene in the melanocortin-4 (mc4r) coding region of channel catfish (Ictalurus punctatus) genome. Transgenic Res 2023; 32:251-264. [PMID: 37468714 DOI: 10.1007/s11248-023-00346-w] [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: 07/08/2022] [Accepted: 03/24/2023] [Indexed: 07/21/2023]
Abstract
Channel catfish, Ictalurus punctatus, have limited ability to synthesize Ω-3 fatty acids. The ccβA-msElovl2 transgene containing masu salmon, Oncorhynchus masou, elongase gene driven by the common carp, Cyprinus carpio, β-actin promoter was inserted into the channel catfish melanocortin-4 receptor (mc4r) gene site using the two-hit two-oligo with plasmid (2H2OP) method. The best performing sgRNA resulted in a knockout mutation rate of 92%, a knock-in rate of 54% and a simultaneous knockout/knock-in rate of 49%. Fish containing both the ccβA-msElovl2 transgene knock-in and mc4r knockout (Elovl2) were 41.8% larger than controls at 6 months post-hatch (p = 0.005). Mean eicosapentaenoic acid (EPA, C20:5n-3) levels in Elov2 mutants and mc4r knockout mutants (MC4R) were 121.6% and 94.1% higher than in controls, respectively (p = 0.045; p = 0.025). Observed mean docosahexaenoic acid (DHA, C22:6n-3) and total EPA + DHA content was 32.8% and 45.1% higher, respectively, in Elovl2 transgenic channel catfish than controls (p = 0.368; p = 0.025). To our knowledge this is the first example of genome engineering to simultaneously target transgenesis and knock-out a gene in a commercially important aquaculture species for multiple improved performance traits. With a high transgene integration rate, improved growth, and higher omega-3 fatty acid content, the use of Elovl2 transgenic channel catfish appears beneficial for application on commercial farms.
Collapse
Affiliation(s)
- Michael Coogan
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA.
| | - De Xing
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Baofeng Su
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Veronica Alston
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Andrew Johnson
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Mohd Khan
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
- Department of Fisheries Biology and Genetics, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh
| | - Karim Khalil
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Ahmed Elaswad
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Shangjia Li
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Jinhai Wang
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Cuiyu Lu
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Wenwen Wang
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Darshika Hettiarachchi
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Mei Shang
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Tasnuba Hasin
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Zhenkui Qin
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Roger Cone
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ian A E Butts
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Rex A Dunham
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| |
Collapse
|
46
|
Zhang SX, Kim A, Madara JC, Zhu PK, Christenson LF, Lutas A, Kalugin PN, Jin Y, Pal A, Tian L, Lowell BB, Andermann ML. Competition between stochastic neuropeptide signals calibrates the rate of satiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.11.548551. [PMID: 37503012 PMCID: PMC10369917 DOI: 10.1101/2023.07.11.548551] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
We investigated how transmission of hunger- and satiety-promoting neuropeptides, NPY and αMSH, is integrated at the level of intracellular signaling to control feeding. Receptors for these peptides use the second messenger cAMP, but the messenger's spatiotemporal dynamics and role in energy balance are controversial. We show that AgRP axon stimulation in the paraventricular hypothalamus evokes probabilistic and spatially restricted NPY release that triggers stochastic cAMP decrements in downstream MC4R-expressing neurons (PVH MC4R ). Meanwhile, POMC axon stimulation triggers stochastic, αMSH-dependent cAMP increments. NPY and αMSH competitively control cAMP, as reflected by hunger-state-dependent differences in the amplitude and persistence of cAMP transients evoked by each peptide. During feeding bouts, elevated αMSH release and suppressed NPY release cooperatively sustain elevated cAMP in PVH MC4R neurons, thereby potentiating feeding-related excitatory inputs and promoting satiation across minutes. Our findings highlight how state-dependent integration of opposing, quantal peptidergic events by a common biochemical target calibrates energy intake.
Collapse
|
47
|
Zheng Y, Rajcsanyi LS, Peters T, Dempfle A, Wudy SA, Hebebrand J, Hinney A. Evaluation of the MC3R gene pertaining to body weight and height regulation and puberty development. Sci Rep 2023; 13:10419. [PMID: 37369769 DOI: 10.1038/s41598-023-37344-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/20/2023] [Indexed: 06/29/2023] Open
Abstract
Recent studies reported an impact of the melanocortin 3 receptor (MC3R) on the regulation of body weight, linear growth and puberty timing. Previously, allele p.44Ile of a frequent non-synonymous variant (NSV) p.Val44Ile was reported to be associated with decreased lean body mass (LBM) and later puberty in both sexes. We Sanger sequenced the coding region of MC3R in 185 children or adolescents with short normal stature (SNS) or 258 individuals with severe obesity, and 192 healthy-lean individuals. Eleven variants (six NSVs) were identified. In-silico analyses ensued. Three rare loss-of-function (LoF) variants (p.Phe45Ser, p.Arg220Ser and p.Ile298Ser) were only found in severely obese individuals. One novel highly conserved NSV (p.Ala214Val), predicted to increase protein stability, was detected in a single lean female. In the individuals with SNS, we observed deviation from Hardy-Weinberg Equilibrium (HWE) (p = 0.012) for p.Val44Ile (MAF = 11.62%). Homozygous p.44Ile carriers with SNS had an increased BMI, but this effect did not remain significant after Bonferroni correction. In line with previous findings, the detected LoF NSVs may suggest that dysfunction in MC3R is associated with decreased body height, obesity and delayed puberty.
Collapse
Affiliation(s)
- Yiran Zheng
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Virchowstr. 174, 45147, Essen, Germany.
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| | - Luisa Sophie Rajcsanyi
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Virchowstr. 174, 45147, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Triinu Peters
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Virchowstr. 174, 45147, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Astrid Dempfle
- Institute of Medical Informatics and Statistics, Kiel University, Kiel, Germany
| | - Stefan A Wudy
- Division of Pediatric Endocrinology and Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Johannes Hebebrand
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Virchowstr. 174, 45147, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Anke Hinney
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Virchowstr. 174, 45147, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
48
|
Ericson MD, Tran LT, Mathre SS, Freeman KT, Holdaway K, John K, Lunzer MM, Bouchard JL, Haskell-Luevano C. Discovery of a Pan-Melanocortin Receptor Antagonist [Ac-DPhe(pI)-Arg-Nal(2')-Orn-NH 2] at the MC1R, MC3R, MC4R, and MC5R that Mediates an Increased Feeding Response in Mice and a 40-Fold Selective MC1R Antagonist [Ac-DPhe(pI)-DArg-Nal(2')-Arg-NH 2]. J Med Chem 2023; 66:8103-8117. [PMID: 37307241 PMCID: PMC10631449 DOI: 10.1021/acs.jmedchem.3c00432] [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] [Indexed: 06/14/2023]
Abstract
Discovery of pan-antagonist ligands for the melanocortin receptors will help identify the physiological activities controlled by these receptors. The previously reported MC3R/MC4R antagonist Ac-DPhe(pI)-Arg-Nal(2')-Arg-NH2 was identified herein, for the first time, to possess MC1R and MC5R antagonist activity. Further structure-activity relationship studies probing the second and fourth positions were performed toward the goal of identifying potent melanocortin antagonists. Of the 21 tetrapeptides synthesized, 13 possessed MC1R, MC3R, MC4R, and MC5R antagonist activity. Three tetrapeptides were more than 10-fold selective for the mMC1R, including 8 (LTT1-44, Ac-DPhe(pI)-DArg-Nal(2')-Arg-NH2) that possessed 80 nM mMC1R antagonist potency and was at least 40-fold selective over the mMC3R, mMC4R, and mMC5R. Nine tetrapeptides were selective for the mMC4R, including 14 [SSM1-8, Ac-DPhe(pI)-Arg-Nal(2')-Orn-NH2] with an mMC4R antagonist potency of 1.6 nM. This compound was administered IT into mice, resulting in a dose-dependent increase in the food intake and demonstrating the in vivo utility of this compound series.
Collapse
Affiliation(s)
- Mark D Ericson
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Linh T Tran
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Sarah S Mathre
- 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
| | - Kelsey Holdaway
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Kristen John
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mary M Lunzer
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jacob L Bouchard
- 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
|
49
|
Ericson MD, Freeman KT, LaVoi TM, Donow HM, Santos RG, Giulianotti MA, Pinilla C, Houghten RA, Haskell-Luevano C. The Parallel Structure-Activity Relationship Screening of Three Compounds Identifies the Common Agonist Pharmacophore of Pyrrolidine Bis-Cyclic Guanidine Melanocortin-3 Receptor (MC3R) Small-Molecule Ligands. Int J Mol Sci 2023; 24:10145. [PMID: 37373293 PMCID: PMC10299128 DOI: 10.3390/ijms241210145] [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: 04/02/2023] [Revised: 05/07/2023] [Accepted: 05/10/2023] [Indexed: 06/29/2023] Open
Abstract
The melanocortin receptors are involved in numerous physiological pathways, including appetite, skin and hair pigmentation, and steroidogenesis. In particular, the melanocortin-3 receptor (MC3R) is involved in fat storage, food intake, and energy homeostasis. Small-molecule ligands developed for the MC3R may serve as therapeutic lead compounds for treating disease states of energy disequilibrium. Herein, three previously reported pyrrolidine bis-cyclic guanidine compounds with five sites for molecular diversity (R1-R5) were subjected to parallel structure-activity relationship studies to identify the common pharmacophore of this scaffold series required for full agonism at the MC3R. The R2, R3, and R5 positions were required for full MC3R efficacy, while truncation of either the R1 or R4 positions in all three compounds resulted in full MC3R agonists. Two additional fragments, featuring molecular weights below 300 Da, were also identified that possessed full agonist efficacy and micromolar potencies at the mMC5R. These SAR experiments may be useful in generating new small-molecule ligands and chemical probes for the melanocortin receptors to help elucidate their roles in vivo and as therapeutic lead compounds.
Collapse
Affiliation(s)
- Mark D. Ericson
- Department of Medicinal Chemistry & Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (M.D.E.); (K.T.F.)
| | - Katie T. Freeman
- Department of Medicinal Chemistry & Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (M.D.E.); (K.T.F.)
| | - Travis M. LaVoi
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA; (T.M.L.); (H.M.D.); (M.A.G.); (C.P.); (R.A.H.)
| | - Haley M. Donow
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA; (T.M.L.); (H.M.D.); (M.A.G.); (C.P.); (R.A.H.)
| | - Radleigh G. Santos
- Department of Mathematics, Nova Southeastern University, Fort Lauderdale, FL 33314, USA;
| | - Marc A. Giulianotti
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA; (T.M.L.); (H.M.D.); (M.A.G.); (C.P.); (R.A.H.)
| | - Clemencia Pinilla
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA; (T.M.L.); (H.M.D.); (M.A.G.); (C.P.); (R.A.H.)
| | - Richard A. Houghten
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA; (T.M.L.); (H.M.D.); (M.A.G.); (C.P.); (R.A.H.)
| | - Carrie Haskell-Luevano
- Department of Medicinal Chemistry & Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (M.D.E.); (K.T.F.)
| |
Collapse
|
50
|
Chen HJC, Mazzaferro S, Tian T, Mali I, Merkle FT. Differentiation, Transcriptomic Profiling, and Calcium Imaging of Human Hypothalamic Neurons. Curr Protoc 2023; 3:e786. [PMID: 37272700 PMCID: PMC7614736 DOI: 10.1002/cpz1.786] [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] [Indexed: 06/06/2023]
Abstract
Neurons in the hypothalamus orchestrate homeostatic physiological processes and behaviors essential for life. Human pluripotent stem cells (hPSCs) can be differentiated into many types of hypothalamic neurons, progenitors, and glia. This updated unit includes published studies and protocols with new advances in the differentiation, maturation, and interrogation by transcriptomic profiling and calcium imaging of human hypothalamic cell populations. Specifically, new methods to freeze and thaw hypothalamic progenitors after they have been patterned and before substantial neurogenesis has occurred are provided that will facilitate experimental flexibility and planning. Also included are updated recipes and protocols for neuronal maturation, with details on the equipment and methods for examining their transcriptomic response and cell-autonomous properties in culture in the presence of synaptic blockers. Together, these protocols facilitate the adoption and use of this model system for fundamental biological discovery and therapeutic translation to human diseases such as obesity, diabetes, sleep disorders, infertility, and chronic stress. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: hPSC maintenance Basic Protocol 2: Hypothalamic neuron differentiation Support Protocol 1: Cortical neuron (control) differentiation Basic Protocol 3: Neuronal maturation Support Protocol 2: Cryopreservation and thawing of neuronal progenitors Support Protocol 3: Quality control: Confirmation of hypothalamic patterning and neurogenesis Support Protocol 4: Bulk RNA sequencing of hypothalamic cultures Basic Protocol 4: Calcium imaging of hypothalamic neurons using Fura-2 AM Alternate Protocol: Calcium imaging of green fluorescent hypothalamic neurons using Rhod-3 AM.
Collapse
Affiliation(s)
- Hsiao-Jou Cortina Chen
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
- Wellcome Trust–Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Simone Mazzaferro
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
- Wellcome Trust–Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Tian Tian
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
- Wellcome Trust–Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Iman Mali
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
| | - Florian T. Merkle
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
- Wellcome Trust–Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|